Monthly Archives: September 2022

FIG 1

Astrocyte, Lipid Metabolism in Alzheimer’s Disease and Glioblastoma

DOI: 10.31038/JCRM.2022555

Abstract

The brain is a central key organ of the body containing the second highest lipid content only after adipose tissue. Lipids as the main structural components of biological membranes play important roles in a vast number of biological processes within the brain such as energy homeostasis, material transport, signal transduction, neurogenesis and synaptogenesis, providing a balanced cellular environment required for proper functioning of brain cells. Lipids and their metabolism are of great physiological importance in view of the crucial roles of lipids in brain development and function. Astrocytes are the most abundant glial cells in the brain and involved in various processes including metabolic homeostasis, blood brain barrier maintenance, neuronal support and crosstalk. Disturbances in lipid metabolism and astrocytic functions may lead to pathological alterations associated with numerous neurological diseases like Alzheimer’s Disease (AD) recognized as the most frequent cause of dementia leading to major progressive memory and cognitive deficits as well as Glioblastoma (GBM) known as the most aggressive malignant brain tumor with a poor prognosis. Herein, we not only review the level and role of altered lipid metabolism in correlation with astrocytic function and astrocyte-neuron crosstalk in AD and GBM, but also discuss important lipid-related metabolites and proteins participating in possible mechanisms of pathologically dysregulated lipid metabolism, offering potential therapeutic targets in targeted molecular therapies for AD and GBM.

Keywords

Astrocyte, Lipid metabolism, Alzheimer’s disease, Glioblastoma

Introduction

The brain is a central and pivotal organ highly enriched in lipids (constituting 50% to 60% of brain dry weight) [1], the major biomacromolecules characterized with poor water-solubility and good solubility in non-polar organic solvent, and is regarded with the second highest lipid content next to the adipose tissue [2]. Lipids are a class of fatty substances differing in overall structure, molecular weight, head group configuration, carbon-carbon bond formation and other factors, among which fatty acids, phospholipids, sphingolipids, sterol lipids and triglycerides are the five main brain lipid classes [3], serving as basic structural components of biological membranes and participating in a broad variety of physiological events, including chemical energy generation and storage, substance transport, cellular signaling, neural differentiation, axonal regeneration, synaptogenesis, synaptic plasticity and brain development [4-14].

The brain consists of neurons and non-neuronal cells such as glial and vascular epithelial cells, of which astrocytes represent the most abundant glial cells [15,16]. Astrocytes mediate diverse biological activities under physiological conditions, including structural and energy support for neurons [17,18], neuronal development and maintenance [19,20], formation, function and plasticity of synapses [21,22], modulation of synaptic transmission [22], metabolomic homeostasis [23] as well as integrity of the Blood-brain Barrier (BBB) [24,25] which is a semipermeable membrane regulating solute exchange between blood and brain parenchyma to maintain CNS homeostasis and function and partially separating local lipid metabolism of the brain from that of the body [25-33]. Apart from the well-known enzymatic capacity of glycogenesis and glycolysis [34-38], equipment of lipid metabolism also exists in astrocytes, providing membrane components for neurons and other glial cells [39,40] and playing fundamental roles in astrocyte function including membrane fluidity, energy generation and intercellular signaling. Emerging evidence has shown that astrocytic usage of lipids stored in droplets via mitochondrial β-oxidation fulfills crucial energy-providing and neuroprotective roles in the brain [18,41], whereby disruption in lipid metabolism, structure and function of astrocytes may lead to pathogenic mechanisms underlying an array of neurological diseases.

Lipid Classification in the Brain

Fatty Acids

As one of the most well-known lipid class, Fatty Acids (FAs), the essential monomeric constituents of all lipids, account for almost 20% of the energy source through oxidation, for which astrocytes as the major provider of fatty acid β-oxidation may be the essential place [42-44]. Additionally, fatty acids can also be utilized by astrocytes for producing ketone bodies under particular conditions (e.g. ischemia), serving as a substrate for neuronal energy production-related Tricarboxylic Acid (TCA) cycle [45]. Fatty acids permeate the Blood-brain Barrier (BBB) via passive (dissociation from albumin carriers, binditheng to luminal membrane which belong to endothelia cells, ATP-independent release and entrance into the cytosol) and/or protein-mediated transport (e.g. Fatty Acid Transport Proteins (FATPs), fatty acid translocase/CD36 (FAT/CD36), Fatty Acid Binding Proteins (FABPs) and caveolin-1) [46,47]. Fatty acids can be further divided into unsaturated and saturated fatty acids, from which the former subclass contains Monounsaturated Fatty Acids (MUFAs) and Polyunsaturated Fatty Acids (PUFAs), while the latter comprises palmitic acid, stearic acid and others [48,49]. PUFAs are highly enriched in the brain, with 3- to 4-fold level over other tissues [50,51]. What’s more, essential PUFAs play key roles in brain activity and development [52,53], in which the ω-3 Docosahexaenoic Acid (DHA) are particularly involved in synaptogenesis, neurogenesis and neuroprotection in the brain [54-57].

Phospholipids

As the most abundant constituent of major categories of membrane lipids [58,59], Phospholipids (PLs) generally consist of two hydrophobic tails of fatty acids differing in length and a backbone-attached hydrophilic phosphate group [60-62].

Phospholipids, which are synthesized in the mitochondria and Endoplasmic Reticulum (ER) tracing from diacylglycerol and phosphatidic acid, spontaneously aggregate into the formation of bimolecular layers in aqueous environments on account of configuration and amphipathic property [63]. Phospholipids can be classified into glycerophospholipids and phosphosphingolipids, of which glycerophospholipids are the prominent glycerol-based class of lipid molecules which can be further subclassified into subtypes such as Phosphatidic Acid (PA), Phosphatidylcholine (PC), Phosphatidylethanolamine (PE), Phosphatidylglycerol (PG), Phosphatidylinositol (PI) and Phosphatidylserine (PS) on the basis of variation in hydrophilic head groups and participate in a variety of physiological activities in the brain [59,64,65]. Moreover, fates of brain cells are influenced by exposure to different phospholipids, such as differentiation of neural cells into astrocytes was promoted and inhibited with PE and PC treatment, respectively [66].

Sphingolipids

Sphingolipids containing sphingoid bases (also known as long-chain bases) and a set of aliphatic amino alcohols that includes sphingosine are mainly synthesized in Endoplasmic Reticulum (ER). Sphingolipids comprise a large group of lipid molecules through compounding with different functional groups, such as ceramide (functional group of single hydrogen atoms) and Sphingomyelin (SM) (functional groups including phosphocholine) with regards to structural composition, functioning as building blocks of membranes (e.g. lipid rafts) [67] and playing fundamental roles in formation and regulation of synapse structure and function [68], cell recognition, signal transmission and inflammatory regulation of astrocytes [69-72]. Besides, sphingolipid metabolites have also been discovered to exert regulatory roles in autophagy, cancer cell growth, response to DNA damage and inflammation [73-75].

Sterol Lipids

Sterol lipids include numerous organic molecules, of which cholesterol with four hydrocarbon rings is the main part. Cholesterol can be synthesized in ER by all nucleated cells, while over 70% of total body cholesterol are provided by the diet [76], namely the cholesterol absorbed in the gut transfers into the liver and then spreads through the body. What is noteworthy is that the brain, unlike other organs, makes its own cholesterol because of effective prevention of peripheral cholesterol exchange between brain tissue and plasma cholesterol-carrying lipoproteins by the BBB [77-79]. In brain tissue, de novo synthesis of cholesterol is mainly performed in astrocytes which are considered as the main cholesterol producer in the brain [80], though the majority of sterol is synthesized in oligodendrocytes in developing brain and has an association with myelination [81] and oligodendrocytes, besides, cholesterol can also be synthesized in many other cell types [82-84]. Apart from de novo synthesis [85], brain cells are able to acquire cholesterol from neighboring cells through the absorption of cholesterol-laden lipoproteins (e.g. Apolipoprotein E (APOE)) in a receptor-mediated way [86,87], in which lipoprotein synthesis for cholesterol transport occurs in astrocytes [88]. With abundant existence in myelin and lipid membranes [81], cholesterol fulfills vital roles in the brain, including BBB integrity, organization of lipid rafts (discrete microdomains present in the external leaflet of plasma membrane), regulation of cell membrane flexibility (through interaction with neighbouring phospholipids) and localization and activity of diverse membrane proteins (e.g. membrane receptor and transporter proteins), axonal guidance, formation and maintenance of synapses and dendrites, synaptic membranerelated fluidity and ion channel function, glucose transport, intracellular signaling and other important neuronal functions [84,89-103].

Triglycerides

As the major form of FA deposition and the optimal form of FA triesters of glycerol, Triglycerides (TGs) are essential ingredients of glycerolipid synthesis by assembling with other glycerol molecules [104]. Triglycerides mainly generated in the adipose tissue and liver can reach other tissues with the package into lipoproteins containing a hydrophilic exterior and a hydrophobic lipid core, including chylomicrons, Very-lowdensity Lipoproteins (VLDL), low-density lipoproteins (LDL), very-high-density lipoproteins (VHDL) and high-density lipoproteins (HDL) only which can cross the BBB [105-108]. Additionally, apolipoprotein E (ApoE) and apolipoprotein J (ApoJ), the most abundant apolipoproteins synthesized in astrocytes, serve as receptor ligands on HDL [109-111] and play fundamental roles in lipid metabolism-associated structural support, enzyme activity and substrate delivery [110,112-114].

Astrocyte-Neuron Coupling of Lipid Metabolism

In humans, the brain representing, on average, merely 2% of total body weight consumes approximately and over 20% of energy substrates during quiet waking and diverse tasks, respectively [115,116], which depends on relatively efficient metabolic coupling between astrocytes and neurons. Physiologically, astrocytes are considered primarily as glycolytic cells with a large enzymatic capacity for glycolysis [115,117,118], whereas neurons are predominantly oxidative [119-121]. Besides the glucose metabolism in which astrocytes participate in the delivery of blood-derived glucose to neurons as an obligatory energy fuel, glycogen storage and activitydependent L-lactate production as a metabolic substrate for neurons during aerobic glycolysis [115,122-124], astrocytes-neuron coupling of lipid metabolism has also been suggested to occur as a response to neuronal activity in protection of neurons from lipotoxicity [125,126]. This is a mechanism proposing that L-lactate-derived de novo synthesis of free fatty acids (FFAs) in overstimulated neurons is triggered during astrocyte-neuron L-lactate shuttle (ANLS), resulting in excess FFAs in association to lipotoxicity-related reactive oxygen species (ROS) and lipid peroxidation chain reaction [127], peroxidized FAs with devastating effects [127] are then transferred from hyperactive neurons to astrocytes via apolipoprotein E-positive lipid particles, where they are directly stored in lipid droplets (LDs) [125,126,128] which are dynamic organelles possessing a core of neutral lipids (e.g. cholesterol esters (CEs) and triacylglycerides (TAGs)), influencing fatty acid breakdown for energy production [129] and buffering excess FFAs to prevent lipid accumulation [130] as well as utilized as an energy substrate in β-oxidation [126] (Figure 1).

FIG 1

Figure 1: Astrocyte-Neuron coupling of lipid metabolism. Excess fatty acids produced in hyperactive neurons are transferred via lipid particles associated with APOE to astrocytes, where fatty acids are delivered to lipid droplets after endocytosis of neuron-derived lipid particles, detoxified as a means of neuron protection under conditions of enhanced activity as well as consumed by mitochondrial oxidation (e.g. β-oxidation). FAs, fatty acids; APOE, apolipoprotein E; LDs, lipid droplets.

AD

With the worldwide increase in longevity, Alzheimer’s disease (AD) as the most common form of senile dementia is rapidly becoming a major health problem [131,133]. AD is a devastating irreversible neurodegenerative disease clinically defined by memory loss, neuropsychiatric abnormalities, cognitive impairment, behaviour deficits and progressive decline of self-care capacity [134-136] as well as pathologically characterized by extracellular amyloid-ß (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated microtubuleassociated protein tau [137-139]. Moreover, accumulation of lipid granules in glia, besides notorious Aβ deposition and tau aggregates, was noticed with the examination of Auguste Deter’s brain (the first described AD patient), initially establishing a possible involvement of perturbations of lipid metabolism in AD pathology [140,141]. Altered lipid metabolism has also been further described with important roles in AD pathogenesis [142-151].

Recent AD pathology-related lipidome studies have demonstrated changes in content of numerous lipids (Table 1). Substantial differences in fatty acid levels were observed in AD brain tissues [152,153], including a decrease in levels of docosahexaenoic acid (DHA) present in frontal cortex gray matter [154] and hippocampus [155] to which damage correlates with impaired learning and memory [156], suggesting a dysregulation of fatty acid metabolism and may potentially marking this neurodegenerative disease [157]. Cholesterol accumulation observed in senile plaques and influenced brain regions from AD patients [158] has been reported in association with region-specific synapse loss [159]. A causal relationship between hypercholesterolemia and dysfunctional cholinergic system, cognitive impairments and pathology of amyloid and tau protein has been also demonstrated [160,161], further supporting important roles of disturbed cholesterol metabolism in AD. What’s more, detection of elevated cholesterol esters was performed in lipid raft-like mitochondria-associated ER membranes (MAMs) [162] of which hyperactivity leads to cholesterol retention and synapse loss and correlates with cognitive deficits [163] and in which accumulated cleaved products of Amyloid Precursor Protein (APP) cause mitochondrial dysfunction, interruption of cellular lipid homeostasis and membrane lipid alterations generally observed in AD pathogenesis [164,165]. Mitochondrial dysfunction, accompanied with increased oxidative stress, in neurons induces a lipid transfer to nearby astrocytes in which lipid droplets accumulate, in turn, mitochondrial dysfunction in glial cells can be caused by accumulation of peroxidated lipids and oxidative stress, contributing to neurodegenerative processes [166-168]. Growing evidence has supported nonnegligible roles of phospholipids and sphingolipids in AD pathogenesis and progression, with studies reporting that phospholipids and sphingolipids, together with acylglycerols, fatty acids and sterol lipids, present significant content changes in AD brain tissues [154,169-175].

Table 1: Summary of lipid changes in AD

Lipids Tissue Changes in AD Ref
Fatty acids Omega-3 fatty acids DHA Brain; CSF; Circulation [176-180]
MFG [179]
FCx [181]
EPA Brain; Circulation [180]
MFG [179]
DPA Brain [182]
ALA Plasma [183]
Omega-6 fatty acids AA Brain; CSF

[177,184,1

85]

MFG [179]
HPC [186]
LA Brain; Plasma [179,187]
Saturated fatty acids Brain; CSF [176]
Eicosanoids PG Brain [188]
Phospholipids Phosphatidylcholine (PC) Total PC lipids Brain [189]
PC-EPA CSF [190]
PC-DHA Plasma [191]
PC-EPA Plasma [191]
Phosphatidylethanolamine (PE) Total PE lipids HPC [186]
PE-SA HPC [192]
PE-OA HPC [192]
PE-AA HPC [192]
PE-DHA HPC [192]
Phosphatidylserine (PS) Total PS lipids Occipital lobe; Inferior parietal [193]
lobule
Sphingolipids Ceramides (CM) Total CM lipids Brain [194]
Sphingomyelin (SM) Total SM lipids CSF [195]
Triglycerides Total TG lipids Serum [196]
Polyunsaturated TG Brain [197]
Sterol lipids Cholesterol Brain [198]
Cholesterol precursors Brain [198]
Total oxidized cholesterol Brain [199]

PC, phosphatidylcholine; PE, phosphatidylethanolamine; PS, phosphatidylserine; CM, ceramides; SM, sphingomyelin; TG, triglyceride; AA, arachidonic acid; ALA, alpha-linolenic acid; DHA, docosahexaenoic acid; DPA, docosapentaenoic acid; EPA, eicosapentaenoic acid; LA, linoleic acid; OA, oleic acid; SA, stearic acid; PG, prostaglandin; CSF, cerebral spinal fluid; FCx, frontal cortex; HPC, hippocampus; MFG, medial frontal gyrus;↑; increased from control ↓; decreased from control.

APOE

In comparison with early-onset familial AD (EOFAD), late-onset AD (LOAD) accounts for approximately 95% of all AD cases [200,201], in which genetic predisposition, after aging, plays major roles in the onset of AD. As the strongest risk factor for LOAD, apolipoprotein E (APOE) is the main lipoprotein in the brain and plays pivotal roles in brain lipid metabolism, membrane remodelling and neuronal growth and repair [202-206]. APOE mainly produced by astrocytes is released into extracellular space where essential lipids (e.g. cholesterol) are delivered to neurons adopting APOE-bound cargo through APOE receptors expressed on the neuronal surface [202]. In addition to the capacities of Aβ binding and influencing Aβ aggregation and clearance [204,207], APOE participates in indirect regulation of Aβ metabolism through interactions with receptors (e.g. low-density lipoprotein receptorrelated protein 1 (LRP1)) [206,208-213]. Critical and isoform-specific role of APOE has also been demonstrated in formation of intraparenchymal Aβ deposits in amyloid precursor protein (APP) transgenic mice [214-217]. APOE exists with 3 different alleles namely APOEε2, APOEε3 and APOEε4, translating to 3 protein isoforms termed APOE2, APOE3 and APOE4, of which APOE4 present in approximately 14% of worldwide populations [205,218] is the most prevalent genetic risk factor for AD [219-222]. A single amino acid difference between APOE3 and APOE4 (Cys 112 Arg) brings about a conformational change influencing the binding to Aβ, lipids and apolipoprotein receptors [223]. APOEε2 considered as a protective genetic factor associated with reduced risk for AD and late age at onset [219,224] has been reported to orchestrate differences in lipidome and transcriptome profiles of postmortem AD brain [218,225]. Conversely, APOEε4 markedly elevates AD risk [219,224], in which heterozygous and homozygous APOEε4 allele may increase AD risk by 3 and 12 times, respectively [223], accelerates disease course and worsens brain pathology [226-228]. A correlation between APOE4 genotype and increased expression of Serpina3n, a gene expressed by astrocytes and considered as a strong marker of reactive and aged astrocytes in the brain [229,230], has been reported with a possible contribution to the pathogenic role of APOE4 in AD [231]. Higher APOE4 level in Cerebral Spinal Fluid (CSF) of AD patients compared with that of control individuals has been connected to accelerated Aβ oligomer accumulation [232]. APOE4 may retard Aβ clearance and favour Aβ deposition via binding to Aβ after specific fragmentation [205,223]. APOE4 was reported to trap ATP-binding cassette transporters A1 (ABACA1) (a regulator of APOE4 lapidation in protection from lipidpoor ApoE4 aggregation) in late rather than recycling endosomes and alter ABACA1 membrane trafficking in astrocytes, which might result in reduced Aβ degradation [233]. Insufficient Aβ clearance also affects accumulation in synaptic cleft, contributing to disruption of hippocampal long-term synaptic plasticity related to learning and memory abilities [234]. APOE4 is internalized in APOE receptors such as low-density lipoprotein receptor-related protein 1 (LRP1) which is also a member of Aβ receptors including very low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (APOER2) [209]. Additionally, APOE4-induced reduction of dendritic spine density in mice [234,235] is consistent with pathological changes (dendritic spine density reduction and synapse loss) observed in brain tissues from AD APOEε4-carriers [236]. APOE4 causes widespread AD phenotypes-associated cellular and molecular alterations in brain cells derived from human induced pluripotent stem cells (iPSCs), among which increased Aβ secretion as well as impaired Aβ uptake and cholesterol accumulation occurred in neurons and astrocytes, respectively [237]. Astrocytic lipid metabolism is influenced by APOE4 [237,238], in which increased fatty acid unsaturation and lipid droplet (LD) accumulation were found in APOE4-expressing human iPSC-derived astrocytes, which can be restored to basal state through supplementation of culture medium with choline (a soluble phospholipid precursor) [238]. Furthermore, APOE4 can also impair astrocyte-neuron coupling of fatty acid metabolism via decreased fatty acid (FA) sequestering in LDs, reduced LD transport efficiency and lowered FA oxidation, resulting in lipid accumulation in astrocytes and hippocampus, diminished abilities of astrocytes in neuronal lipid elimination and FA degradation, accelerated lipid dysregulation and increased AD risk [239].

ACSBG1 and ACSL6

Cellular accumulation and activation of fatty acids (FAs) either synthesized de novo or taken up from diets require the ATP-dependent reaction catalyzed by acyl-CoA synthetases (ACSs), a family of enzymes initiating FA metabolism-related reactions through ligation to coenzyme A (CoA) [240]. ACS enzyme family contain various members differing in distribution and fatty acid substrate preference [241], among which only two show specific enrichment in the brain, ACSBG1 and ACSL6 [242,243], suggesting their potentially particular roles in modulation of brain fatty acid metabolism. ACSBG1, almost exclusively expressed in astrocytes, have preferences for a wide range of substrates containing long-chain saturated and unsaturated fatty acids [244,245]. ACSBG1 knockdown in vitro results in decreased ACS enzymatic activity and FA oxidation [245], indicating its participation in astrocytic FA oxidation, however, clear roles of ACSBG1 in brain function and/or dysfunction still remain poorly understood. ACSL6 showing high expression in the brain was reported to be downregulated in age-related neurodegenerative diseases [246,247] and in direct correlation with neurite outgrowth [248-252]. With high substrate preference for docosahexaenoic acid (DHA) of which low levels are associated with AD pathophysiology [253], ACSL6 has been revealed with key roles in regulating DHA incorporation into neuronal membranes using Acsl6 deficient mice with significant reduction in DHA-containing phospholipids and impaired memory [254,255]. Critical roles of ACSL6 in brain DHA retention and neuroprotection are further supported by findings that ACSL6 depletion led to markedly reduced levels of brain membrane phospholipid DHA, spatial memory deficits, hyperlocomotion, increased cholesterol biosynthesis and age-related neuroinflammation [256]. What’s noteworthy is that astrocyte-specific depletion had minimal influence on membrane lipid composition [256] in consideration of ACSL6 enrichment in astrocytes [240,257-261], possibly due to the expression of a DHA-nonpreferring variant [251,262-267] and enrichment of Y-gate domain rather than DHA-preferring F-gate domain in astrocytes [251].

ATAD3A

ATPase family AAA-domain containing protein 3A (ATAD3A), a nuclear-encoded mitochondrial membrane-anchored protein belonging to the AAA+-ATPase protein family and simultaneously interacting with inner and outer mitochondrial membranes, is implicated in a variety of biological processes including stability maintenance of mitochondrial DNA (mtDNA), regulation of mitochondrial dynamics and cholesterol metabolism [268-270]. ATAD3A deficiency led to neurodegenerative phenotypes in association with cholesterol elevation, downregulated expression of cholesterol metabolism-related genes [269], optic atrophy and axonal neuropathy [271]. Oligomerization and accumulation of ATAD3A at MAMs, lipid raft-like ER subdomain rich in sphingomyelin and cholesterol [272] and associated with diverse metabolic functions such as lipid metabolism, mitochondrial function and calcium homeostasis [273-277], have been discovered in both mouse models and postmortem human brain tissues of Alzheimer’s disease [278]. Aberrantly oligomerized ATAD3A leads to cholesterol accumulation via expression inhibition of cholesterol clearancemediating cytochrome P450 family 46 subfamily A member 1 (CYP46A1) located on MAMs of which deficiency correlates with cholesterol disturbance, amyloid aggregation and cognitive impairments [279], AD-like MAM hyperconnectivity (e.g. impaired MAM integrity) [277] as well as synapse loss [278]. MAM-resident cholesterol imbalance facilitates amyloidogenic APP cleavage [165], in turn, retention of APP proteolytic fragments at MAMs interrupts cholesterol trafficking and homeostasis [280]. Additionally, blocking ATAD3A oligomerization by heterozygous knockout or pharmacological inhibition treated with DA1 peptide has been reported in causal relationship with cholesterol turnover normalization, MAM integrity enhancement, APP processing suppression, synapse loss mitigation and ultimate reduction of AD-like neuropathology and cognitive impairments [278], further revealing a role of ATAD3A in AD pathology and suggesting a potential therapeutic strategy of retarding AD progression through manipulation of abnormal ATAD3A oligomerization.

FoxO3

Forkhead box O transcription factor 3 (FoxO3) belonging to the forkhead box (FOX) family sharing an evolutionarily conserved forkhead DNA-binding domain composed of 80 to 100 amino acids [281]and possessing single nucleotide polymorphisms (SNPs) associated with human longevity [282,283] functions as a mediator of biological processes promoting lifespan and preventing aging-related diseases [284,285], of which alterations are involved in carcinoma, cardiovascular and neurodegenerative diseases [283,286-289]. FoxO3 plays a pivotal role in quiescence maintenance of neural stem cells (NSCs) in the brain, removal of which induces NSC differentiation and consequent NSC pool reduction [290-293]. Apart from capacities for neuronal survival promotion or neuronal apoptosis mediation [294,295], FoxO3 has also been shown with astrocyte proliferation controlling through inhibiting inflammatory cytokines (e.g. TNF-α and IL-1β) mediating reactive astrogliosis in neurodegenerative diseases [296-299]. Conditional knockout of FoxO3 in astrocytes was reported to impair consumption of excess fatty acids [300] which are cytotoxic and destructive to mitochondrial function [301]. FoxO3 reduction in aged mice was found to be specific to the cortex rather than the hippocampus, where FoxO3 deficiency caused cortical astrogliosis and dysregulated lipid metabolism [300]. In addition, lipid dysregulation, mitochondrial dysfunction together with Aβ uptake impairment were also observed in cultured astrocytes deficient in FoxO3, which could be reversed by astrocytic FoxO3 overexpression [300], potentially supporting the concept that FoxO3 elevation in astrocytes may retard or restore cortical astrogliosis and AD-associated impairments.

GSAP

Under typical conditions, Amyloid-β (Aβ) peptides as the products of body’s cholesterol disturbance are cleaved from amyloid precursor protein (APP) which may occur in two cellular pools, namely lipid raft-associated pool preferentially favouring APP cleavage by β- and γ-secretase as well as non-raft pools where cleavage is performed by α-secretase in a non-amyloidogenic pathway [302] and rapidly eliminated to maintain normal Aβ levels [303]. γ‐secretase activating protein (GSAP) was first reported for its regulatory roles in γ-secretase activity and specificity and its significant and selective enhancement of Aβ production through interactions with γsecretase and amyloid precursor protein carboxy‐terminal fragment (APP-CTF) [304]. Significantly upregulated GSAP level has been demonstrated in both AD mouse models and postmortem brain tissues from AD patients [305-307]. Single-nucleotide polymorphisms (SNPs) at the GSAP locus have been shown association with AD diagnosis [308,309], of which one SNP was found to correlate with GSAP expression and AD risk [310]. Genetic knockdown and pharmacological inhibition of GSAP suppress Aβ generation and deposition and tau phosphorylation in AD mouse models [304,305,311]. Apart from the promotion of APP-CTF partitioning into Aβ production-favoring lipid rafts, GSAP has also been shown to be enriched in mitochondria-associated membranes (MAMs), an intracellular domain where amyloidogenic APP processing responsible for dysregulated lipid metabolism is performed [312,313]. GSAP depletion lowers APP-CTF accumulation in lipid rafts, reduces ER-mitochondrial contacts elevated in AD [313-316], and alters lipid profiles in a direction opposite to AD pathogenesis (e.g. GSAP depletion-raised levels of phosphatidylethanolamine (PE) and phosphatidylinositol (PI) showing consistent reduction in human AD brain) [310,317]. What’s more, interactions between GSAP and multiple components related to ER-associated degradation (ERAD) regulating mitochondrial function through MAM and participating in AD pathogenesis have also been revealed, further supporting crucial roles of GSAP in attenuating AD-associated pathogenic process.

Glioblastoma

Glioma as a malignant primary brain tumor originating from astrocytes or other glial cells accounts for approximately 80% of all malignant brain tumors [318], of which glioblastoma (GBM) is the most aggressive type of brain tumor known with a 5-year survival rate below 5% [319-321]. Metabolic reprogramming has been recognized as a fundamental hallmark for carcinogenesis and progression of multiple tumors including GBM [322-324], through which tumor cells meet the high-energy demands of rapid proliferation [325]. Except for the representative metabolic feature named the Warburg effect, a phenomenon in which GBM cells rely on glycolysis for energy production under oxygen-sufficient and oxygen-insufficient conditions [323,325-327], GBM cells can also be fueled by fatty acid oxidation (FAO) as an alternative crucial energy resource to meet high-energy consumption in GBM aggressiveness [328-332], of which inhibition negatively impacted GBM proliferation and progression [333]. Oxidation of fatty acids is achieved by two major pathways, namely enzymatic oxidation mediated by peroxidases (e.g cyclooxygenase (COX), cytochrome P450 (CYP450), lipoxygenase (LOX) and phospholipase A2 (PLA2)) [334] as well as nonenzymatic self-catalyzed peroxidation (Figure 2A) of which 4-hydroxynonenal (4HNE) is an end-product showing elevated expression proportional to the grade of brain tumor malignancy [335-337]. Moreover, lipid metabolism reprogramming in association with numerous pathophysiological processes such as tumor proliferation and development [338-343] has been further evidenced with the observation of large amounts of lipid droplets (LDs) in GBM [344-346] and other tumors [347-354]. Neutral lipid core of a single LD includes cholesteryl esters and triglycerides (TGs) composed of glycerol molecules with triple hydroxyl groups esterified by fatty acids [355-358]. TGs have been demonstrated to serve as an important energy reservoir for supporting GBM cell survival, in which LDs were rapidly broken down by GBM cells via autophagy, a pivotal cellular process degrading damaged organelles and protein aggregates and recycling nutrients via hydrolysis of cytoplasmic components to ultimately maintain cellular homeostasis [359-362], to release stored fatty acids for producing energy upon energetic stress like glucose deprivation (Figure 2B), in turn, inhibition of FAO or autophagy led to LD retention and significant potentiation of GBM cell death [363], suggesting that LDs may play critical roles in regulating GBM growth and limitation of LD usage might be indispensable in GBM treatment. What’s more, cholesterol metabolism in GBM is different from that in healthy brain tissues where nearly all brain cholesterol is synthesized de novo [364-366]. Contrary to normal astrocytes mainly synthesizing cholesterol from glucose or glutamine [367,369] and converting excess cholesterol to oxysterol as an endogenous ligand of liver X receptors (LXRs) to consequently trigger efflux of surplus cholesterol via ATPbinding cassette transporter A1 (ABCA1) and suppression of cholesterol uptake by low-density lipoprotein receptors (LDLRs) [370-374], GBM cells are insufficient to de novo synthesize cholesterol and thus dependent on exogenously supplied cholesterol for survival through upregulated LDLR expression [364,375] (Figure 2C), in which LXR agonists could induce GBM cell death by lowering intracellular cholesterol content via ABCA1-dependent cholesterol efflux and LDLR inhibition [364]. Additionally, intracellular cholesterol level has been revealed to be involved in resistance against GBM cell death induced by temozolomide (TMZ), a blood-brain barrier (BBB) penetrant chemotherapy agent currently used in the standard therapy for patients with GBM [376,377]. Furthermore, sphingomyelins (SMs), an important group of phospholipids in cell membranes, together with their hydrolysis by sphingomyelinases (SMase) are crucial to effects of radio- and chemotherapy [378,381]. Ceramides which are generated by SMase-mediated SM hydrolysis caused by TMZ and radiation can induce cell apoptosis [382-384], which can be evaded through conversion of ceramides to sphingosine-1-phosphate (S1P) (Figure 3) [385-387] linked to tumor grade and implicated in GBM aggressive phenotypes [383,388].

FIG 2

Figure 2: A. Scheme of non-enzymatic self-catalyzed lipid peroxidation. Abstraction of allylic hydrogen from PUFA induces lipid radical formation and initiates a chain reaction of lipid peroxidation, which is followed by conjugated diene-yielding molecular rearrangement. Conjugated dienes, in presence of molecular oxygen, are transformed to lipid peroxyl radical abstracting allylic hydrogen from another PUFA, forming lipid hydroperoxide and another lipid radical. Lipid hydroperoxide can be further catalyzed and transformed to lipid alkoxyl radical and lipid peroxyl radical. Lipid peroxidation is terminated when non-radical products are formed because of interaction with antioxidants. Reaction between two lipid peroxyl radicals or two lipid alkoxyl radicals will consequently form a peroxide-bridged lipid dimer, while lipid dimers can be formed by reaction between lipid hydroperoxides and lipid radicals. PUFA, polyunsaturated fatty acids. B. Schematic model of LDs hydrolysis maintaining GBM cell survival. GBM cells mainly utilize glucose to produce energy under glucose-rich conditions, while LDs can be rapidly broken down after autophagy activation upon glucose starvation, released FAs then enter mitochondria for energy production. FAs, fatty acids; LDs, lipid droplets; GBM, glioblastoma. C. Astrocytes are relied upon by neurons and GBM cells to provide de novo synthesized cholesterol. Neurons and GBM cells take up astrocyte-secreted cholesterol in APOE-containing lipoproteins. Following cholesterol uptake mediated by LDLR, oxysterol and cholesterol derivatives produced in neurons are physiological agonists for LXR of which activation leads to dimerization with RXR and subsequent elevation in ABCA1 expression. LXR activation also inhibits LDLR expression, resulting in decreased cholesterol uptake and regulating intracellular cholesterol level. On the contrary, mechanisms surveilling and regulating cholesterol are disrupted in GBM cells, in which oxysterol and cholesterol derivatives cannot activate LXR inducing intracellular cholesterol accumulation. ABCA1, ATP-binding cassette transporter A1; APOE, apolipoprotein E; GBM, glioblastoma; LDLR, low-density lipoprotein receptor; LXR, liver X receptor; RXR, retinoid X receptor.

FIG 3

Figure 3: Sphingolipid metabolism in tumor progression. Sphingomyelin, after chemotherapy and radiation, is broken down into ceramide involved in blocking tumor progression. Ceramide can be converted by tumor cells to S1P (S1P can also be produced by astrocytes and other cells) exerting protumor effects including tumor proliferation, migration, invasion and angiogenesis. Involvement of S1P in tumor progression is specifically mediated by S1PRs (S1PR1-S1PR5) which can signal through phospholipase mechanisms. Each S1PR can couple to one or more GPCRs to signal through different phospholipases and induce phenotypes (e.g. angiogenesis, proliferation, migration and invasion). CDase, ceramidase; GPCRs, G protein-coupled receptors; SMase, sphingomyelinase; S1P, sphingosine-1-phosphate; S1PRs, S1P receptors.

S1PRs

GBM cells utilize exogenous source of S1P synthesized and exported by astrocytes and neuronal cells [389] and endogenous S1P production [390] for tumor progression. Involvement of S1P in tumor growth, migration, invasion, survival and angiogenesis [391-394] is specifically mediated by the family of G-protein coupled receptors named S1P receptors (S1PRs, S1PR1-S1PR5) [395-400]. S1PR1, S1PR2, S1PR3 and S1PR5 are expressed in human GBM cells [401-403], and elevated levels of S1PR1, S1PR2, and S1PR3 have been detected in brain tissues from GBM patients compared with healthy tissues, while only S1PR1 and S1PR2 showed significant association with GBM survival rates [401,402]. S1PRs are essential for mediating diverse S1P functions, whereas orientations in which they influence cell phenotypes still remain unclear. S1PR1 inhibition was reported to promote GBM cell proliferation, which collides with studies suggesting increased GBM proliferation by S1PR1-3, of which S1PR1 showed the strongest effects [402,404]. S1PR2 was shown to both reduce GBM migration through Rho/Rho kinase signaling pathway and participate in promoting GBM invasion [405,406]. In addition, S1PR5 has also been identified as an independent prognostic factor of GBM patients’ survival, aligning with reported role of S1PR5 in proliferation promotion [404,407]. Pharmacologically altered S1PR expression by fingolimod (FTY720), a sphingosine analogue leading to S1PR1 internalization, has been revealed to suppress astrocyte activation and change astrocytic secretion of C-X-C motif chemokine 5 (CXCL5) known to promote GBM proliferation and migration [408-410]. Furthermore, functions of individual S1P receptor subtypes are dependent upon activation of diverse downstream effector proteins, especially coupling to different G-proteins [399], such as binding of S1PR1, S1PR2 and S1PR5 with Gi, activation of Gq by S1PR2 and S1PR3 as well as signaling of S1PR2, S1PR3, and S1PR5 via G12/13 (Figure 3) [411], which alters signaling of phospholipases (particularly phospholipase C (PLC) cleaving proximal phosphodiester bonds of glycerophospholipids in production of phosphorylated headgroups and diacylglycerols [399,400]) and further activates downstream signaling molecules (e.g. extracellular signal-regulated kinase (ERK), phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MEK)) (Table 2). What’s noteworthy is that a S1PR-targeted liposomal drug delivery system, named S1P/JS-K/Lipo, capable of blood-brain tumor barrier (BBTB) penetration and enhanced tumor-targeted delivery has recently been described, efficiently delivering a nitric oxide (NO) prodrug (JS-K, O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate) to GBM tissues via specific interactions with S1PRs highly expressed on GBM cells [412], representing a promising targeted approach for GBM therapy.

Table 2: Summary of S1PR-mediated effects in GBM

Models Involved S1PRs Signaling Pathways Findings Ref
LN18 GBM cells;

U87MG GBM cells.

S1PR1 ↑

S1PR2 ↑

S1PR3 ↑

PI3K/AKT1 pathway Demonstrated association between S1P1 and S1P2 with GBM patient’s [413]
survival. S1PR1/2 inhibition reduces GBM migration.
U373MG GBM cells S1PR1 ↑

S1PR2 ↑

S1PR3 ↑

MAPK/ERK and PI3Kβ pathway S1P promotes glioma cell proliferation. [414]
U373MG GBM cells;  GBM6 cells;  GBM12 cells. S1PR2 MEK1/2 and Rho/ROCK S1P induces mRNA and protein expression of PAI-1 and uPAR, which are important for GBM invasiveness. [415]
U373MG GBM cells; U118MG GBM cells. S1PR1↑

S1PR2

S1PR3

MAPK-ERK

Rho/ROCK

S1PR, S1PR2 and S1PR3 all positively contribute to S1P-stimulated glioma cell proliferation, of which S1PR1 makes the major contribution. [416]
C6 glioma cells S1PR2 MAPK/ERK, PKC, PLC, PLD and Ca2+ signaling S1PRs are linked to at least two signaling pathways (i.e. PTX-sensitive Gi/Go-protein pathway and toxin- insensitive Gq/G11-PLC pathway). [417]
C6 glioma cells; 1321-N1 astrocytoma cells. S1PR2 PI3K/Cdc42/p38MAPK and PI3K/Rac1/JNK S1PR2 mediates S1P-induced negative regulation of glioma cell migration. [418]
U373MG GBM cells; U87MG GBM cells; M059K cells; U-1242 cells; A172 cells. S1PR1 ↑

S1PR2 ↑

S1PR3 ↑

MAPK/ERK and PI3K S1P potently enhances glioma cell motility by signaling through coupling of S1PRs to Gi proteins. [419]
T98G glioma cells; G112 glioma cells. S1P1, S1P2, S1P3 and S1P5 PTEN/AKT/Egr S1PR1 is a significant prognostic factor for glioma; [420]
Downregulated S1PR1 expression increases glioma cell proliferation and enhances glioma malignancy.
Human GBM specimens; U87 glioma cells; U251 glioma cells; T98G glioma cells; G112 glioma cells. S1PR1↓ Downregulated S1PR1 expression in GBM patients with a poor survival. S1PR1 signaling negatively controls glioma cell proliferation. [421]

AKT, v-akt murine thymoma viral oncogene homolog; Cdc42, cell division control protein 42 homolog; ERK, extracellular signal-regulated kinase; JNK, c-Jun Nterminal kinase; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; PLC, phospholipase C; PLD, phospholipase D; PTEN, phosphatase and tensin homolog; PTX, pertussis toxin; Rac1, Ras-related C3 botulinum toxin substrate 1; ROCK, Rho-associated protein kinase.

FABP7

Fatty acid binding protein 7 (FABP7), a member of the multi-gene FABP family comprised of structurally related proteins with expression patterns specific to cell, tissue and development, binds to very long chain polyunsaturated fatty acids (VLCPUFAs) such as docosahexaenoic acid (DHA) with high affinity [422,423]. FABP7 abundant in astrocytes [424-426] is a lipid chaperone mediating cellular uptake, intracellular trafficking and subsequent oxidation of fatty acids (FAs), whose expression was reported to be elevated in GBM and GBM stem-like cells forming neurospheres (NS) and might accounting for GBM aggressiveness [427,428] and recurrence as well as associated with proliferation, migration and invasion of GBM cells, GBM histology and reduced survival time [429-434]. Under metabolic stresses (e.g. hypoxia), fatty acids are stored as lipid droplets (LDs) and subsequently oxidized in a FABP-dependent manner for energy production in GBM cells [435]. Slowcycling cells (SCCs), a subpopulation of GBM cells preferentially utilizing mitochondrial oxidative phosphorylation (OXPHOS), showing elevated lipid contents specifically metabolized under glucose deprivation and displaying enhanced capabilities of migration, invasion and chemoresistance, have been revealed with the characterization of higher FABP expression and larger LD amounts in cultured conditions of normal oxygen levels or nutrients [436]. Additionally, resistance of SCCs against deprived glucose or inhibited glycolysis could be restrained by FA uptake blocking via genetic deletion or pharmacological inhibition of FABP7 [436].

Moreover, promotion effects of FABP7 on GBM cell migration can be mitigated with DHA supplementation through specific and dramatic inhibition of DHA supplementation in culture medium on plasma membrane lipid order of FABP7expressing GBM cells which positively correlates with GBM cell migration as well as DHA supplementation-mediated disruption of nanodomains formed by FABP7 on GBM cell membranes [437], further suggesting a critical role of FABP7 in lipid metabolism in GBM cells.

SCD

Stearoyl-CoA desaturase (SCD) is an endoplasmic reticulum (ER)-localized delta-9 fatty acid desaturase forming carbon-carbon double bonds at the 9th to 10th position from the COOH-terminus of saturated fatty acids (SFAs), stearic acid and palmitic acid and thus generating monounsaturated fatty acids (MUFAs), oleic acid and palmitoleic acid, respectively [438,439], whose expression correlates with the ratio of MUFA to SFA in which a disequilibrium contributes to alterations in cell growth and differentiation [438-441]. SCD has 4 isoforms in mice (SCD1, SCD2, SCD3 and SCD4), while only two paralogs are expressed in human, namely SCD sharing approximately 85% amino acid identity with mouse SCDs and SCD5 unique to primates [440,442]. SCD has been described as a hypermethylated gene member contributing to the CpG island methylator phenotype which defines a distinct glioma subgroup [443]. SCD expression in GBM, in contrast to SCD upregulation often observed in multiple human tumors [444-447], was reported to be lower than normal brain tissues because of hypermethylation and monoallelic deletion together with phosphatase and tensin homolog (PTEN) frequently deleted in GBM [448] in a subset of GBM patients [449]. In addition, GBM cells without epigenetic and genetic changes mentioned above were revealed to express elevated SCD levels on which tumor cells rely for their survival [449]. SCD inhibition by CAY10566, an inhibitor with a modest BBB penetration ability, has been demonstrated to not only significantly suppress intracranial GBM growth, but also obviously affect tumor vasculature including nearly complete blocking of intratumoral bleeding and possible normalization of blood vessels, potentially allowing enhanced delivery of combinedly used antitumor drugs such as temozolomide (TMZ) [449,450].

Conclusions

The brain is highly enriched in lipids where they are crucial for multiple physiological processes ranging from maintenance of structural integrity and metabolic homeostasis to brain function and development. Metabolism of lipids is a complicated process in which a wide range of lipid-related effector proteins are involved and whose alteration is strongly associated with brain dysfunctions and diseases such as Alzheimer’s disease (AD) and glioblastoma (GBM). In this review, we throw light upon basic classes of lipids including fatty acids, phospholipids, sphingolipids, sterol lipids and triglycerides, of which dysregulated metabolism can be regarded as disease biomarkers. We also briefly discuss the role of lipids within the brain and altered lipid profile correlated with astrocytic function and astrocyte-neuron crosstalk in AD and GBM. Moreover, we have discussed lipid-related metabolites and proteins critical for disease-associated lipid dyshomeostasis and how these proteins together with lipids in correlation with astrocytic functions modulate disease pathogenesis and development, enlightening their therapeutic potential in preventing onset and progression of AD and GBM. However, there are still several lipids whose association with AD and GBM and availability as clinically valuable biomarkers for disease detection at early stages need further evaluation, which can be performed by newly-developed and improved techniques of gradually matured lipidomic platforms. What’s more, there remains much to be discovered about benefits and risks of manipulation of compounds affecting effector proteins involved in lipid metabolism, and further characterization of pathways in which important lipid-related proteins participate along with clinical studies will aid the understanding of pathogenesis mechanisms behind AD and GBM and identification of novel therapeutic targets to help ameliorate disease courses, facilitate disease treatments and consequently benefit patients.

References

  1. Mita R, Coles JE, Glubrecht DD, Sung R, Sun X, et al. (2007) B-FABP-expressing radial glial cells: The malignant glioma cell of origin? Neoplasia 9: 734-744. [crossref]
  2. Mita R, Beaulieu MJ, Field C, Godbout R (2010) Brain fatty acid-binding protein and omega-3/omega-6 fatty acids: Mechanistic insight into malignant glioma cell migration. J Biol Chem 285: 37005-37015. [crossref]
  3. Antonella DR, Serena P, Marco R, Patrizia T, Letizia M, et al. (2012) A radial glia gene marker, fatty acid binding protein 7 (FABP7), is involved in proliferation and invasion of glioblastoma cells. Plos One 7: e52113. [crossref]
  4. (2020) 2020 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia 16: 391-460.
  5. Brookmeyer R, Evans DA, Hebert L, Langa KM, Heeringa SG, et al. (2011) National estimates of the prevalence of Alzheimer’s disease in the United States. Alzheimer’s & Dementia 7: 61-73. [crossref]
  6. Rlhh A, Cagw A, Pa A, Maj B, Frjv A, Jls C (2014) Diagnosing Alzheimer’s disease: A systematic review of economic evaluations. Alzheimer’s & Dementia 10: 225-237.
  7. Wilfling F, Haas JT, Walther TC, Jr R (2014) Lipid droplet biogenesis. Curr Opin Cell Biol 59: 88-96.
  8. Olzmann James A, Carvalho P (2019) Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol 20: 137-155. [crossref]
  9. Liu L, Zhang K, Sandoval H, Yamamoto S, Jaiswal M, et al. (2015) Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration. Cell 160: 177-190.
  10. Yoon H, Shaw JL, Haigis MC, Greka A (2021) Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell 81: 3708-3730. [crossref]
  11. Ioannou MS, Jackson J, Sheu SH, Chang CL, Liu Z (2019) Neuron-Astrocyte Metabolic Coupling Protects against Activity-Induced Fatty Acid Toxicity. Cell 177: 1522-1535.
  12. Liu L, Mackenzie KR, Putluri N, Maleti-Savati M, Bellen HJ (2017) The Glia-Neuron lactate shuttle and elevated ROS promote lipid synthesis in neurons and lipid droplet accumulation in glia via APOE/D. Cell Metab 26: 719-737. [crossref]
  13. Magistretti PJ, Allaman I (2018) Lactate in the brain: From metabolic endproduct to signalling molecule. Nat Rev Neurosci 19: 235-249. [crossref]
  14. Magistretti PPJ (1994) Glutamate uptake into astrocytes stimulates aerobic glycolysis: A mechanism coupling neuronal activity to glucose utilization. Proceedings of the National Academy of ences of the United States of America 91: 10625-10629. [crossref]
  15. Alexei, Verkhratsky, Vladimir, Parpura, Nina, Vardjan, et al. (2019) Physiology of astroglia. Adv Exp Med Biol 1175: 45-91.
  16. Afridi R, Kim JH, Rahman MH, Suk K (2020) Metabolic regulation of glial phenotypes: Implications in neuron-glia interactions and neurological disorders. Front Cell Neurosci 14: 20. [crossref]
  17. Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, et al. (2015) Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci 18: 1819-1831. [crossref]
  18. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O Keeffe S, et al. (2014) An RNA-Sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34: 11929-11947. [crossref]
  19. Abe T, Takahashi S, Suzuki N (2006) Oxidative metabolism in cultured rat astroglia: Effects of reducing the glucose concentration in the culture medium and of D-aspartate or potassium stimulation. Journal of Cerebral Blood Flow & Metabolism J Cereb Blood Flow Metab 26: 153-160. [crossref]
  20. Goyal MS, Hawrylycz M, Miller JA, Snyder AZ, Raichle ME (2014) Aerobic glycolysis in the human brain is associated with development and neotenous gene expression. Cell Metab 19: 49-57. [crossref]
  21. Dienel GA (2019) Brain glucose metabolism: Integration of energetics with function. Physiol Rev 99: 949-1045. [crossref]
  22. Dienel GA, Cruz NF (2016) Aerobic glycolysis during brain activation: Adrenergic regulation and influence of norepinephrine on astrocytic metabolism. J Neurochem 138: 14-52. [crossref]
  23. Ramasamy Indra (2014) Recent advances in physiological lipoprotein metabolism. Clin Chem Lab Med 52. [crossref]
  24. Filou S, Lhomme M, Karavia EA, Kalogeropoulou C, Theodoropoulos V, et al (2016) Distinct roles of apolipoproteins a1 and e in the modulation of High-Density lipoprotein composition and function. Biochemistry 55: 3752-3762 [crossref]
  25. Bolanos-Garcia VM, Miguel RN (2003) On the structure and function of apolipoproteins: More than a family of lipid-binding proteins. Prog Biophys Mol Biol 83: 47-68. [crossref]
  26. Ito JI, Nagayasu Y, Miura Y, Yokoyama S, Michikawa M (2014) Astrocytes endogenous apoE generates HDL-like lipoproteins using previously synthesized cholesterol through interaction with ABCA1. Brain Res 1570: 1-12. [crossref]
  27. Center M, Francisco S Introduction to lipids and lipoproteins.
  28. Pitas RE, Boyles JK, Lee SH, Foss D, Mahley RW (1987) Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins. Biochim Biophys Acta 917: 148-161. [crossref]
  29. Balazs Z, Panzenboeck U, Hammer A, Sovic A, Quehenberger O, et al. (2010) Uptake and transport of high-density lipoprotein (HDL) and HDLassociated alpha-tocopherol by an in vitro blood-brain barrier model. J Neurochem 89: 939-950. [crossref]
  30. Braun V, Hantke K (2019) Lipoproteins: Structure, function, biosynthesis. Subcell Biochem 92: 39-77. [crossref]
  31. Hoofnagle AN, Heinecke JW (2009) Lipoproteomics: Using mass spectrometry-based proteomics to explore the assembly, structure, and function of lipoproteins. J Lipid Res 50: 1967-1975. [crossref]
  32. Alaupovic P (1996) Significance of apolipoproteins for structure, function, and classification of plasma lipoproteins. Method Enzymol 263: 32-60. [crossref]
  33. Ghosh S, Strum JC, Bell RM (1997) Lipid biochemistry: Functions of glycerolipids and sphingolipids in cellular signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 11: 45. [crossref]
  34. Zhu X, Owen JS, Wilson MD, Li H, Griffiths GL, et al. (2010) Macrophage ABCA1 reduces MyD88-dependent Toll-like receptor trafficking to lipid rafts by reduction of lipid raft cholesterol. J Lipid Res 51: 3196-3206. [crossref]
  35. Woller SA, Choi S, An EJ, Low H, Schneider DA, Ramachandran R, et al. (2018) Inhibition of neuroinflammation by AIBP: Spinal effects upon facilitated pain states. Cell Rep 23: 2667-2677. [crossref]
  36. Zhang J, Li Q, Wu Y, Wang D, Xu L, Zhang Y, et al. (2019) Cholesterol content in cell membrane maintains surface levels of ErbB2 and confers a therapeutic vulnerability in ErbB2-positive breast cancer. Cell Commun Signal 17. [crossref]
  37. Banach-Orowska M, Wyszyska R, Pyrzyska B, Maksymowicz M, et al. (2019) Cholesterol restricts lymphotoxin receptor-triggered NF-B signaling. Cell Commun Signal 17. [crossref]
  38. Luchtman DW, Song C (2013) Cognitive enhancement by omega-3 fatty acids from child-hood to old age: Findings from animal and clinical studies. Neuropharmacology 64: 550-565. [crossref]
  39. Hamilton JA, Hillard CJ, Spector AA, Watkins PA (2007) Brain uptake and utilization of fatty acids, lipids and lipoproteins: Application to neurological disorders. J Mol Neurosci 33: 2-11. [crossref]
  40. Dawson G 1999 Basic Neurochemistry, 6th Edition. J Neurosci Res
  41. Subramaniam S, Fahy E, Gupta S, Sud M, Byrnes RW, et al. (2011) Bioinformatics and systems biology of the lipidome. Chem Rev 111: 6452-6490. [crossref]
  42. Han X (2016) Lipidomics for studying metabolism. Nat Rev Endocrinol 12: 668-679. [crossref]
  43. Li M, Yang L, Bai Y, Liu H (2014) Analytical methods in lipidomics and their applications. Anal Chem 86: 161-175. [crossref]
  44. Cermenati G, Mitro N, Audano M, et al. (2015) Lipids in the nervous system: From biochemistry and molecular biology to patho-physiology. Biochim Biophys Acta 1851: 51-60. [crossref]
  45. Qiuhui X, Chunxiu H, Di Y, Melcangi RC, Crestani M, et al. (2018) Development of a high coverage pseudotargeted lipidomics method based on UltraHigh performance liquid Chromatography-Mass spectrometry. Anal Chem 90: 7608-7616. [crossref]
  46. Wood PL, Cebak JE (2018) Lipidomics biomarker studies: Errors, limitations, and the future. Biochem Biophys Res Commun 504: 569-575. [crossref]
  47. Rustam YH, Reid GE (2017) Analytical challenges and recent advances in mass spectrometry based lipidomics. Anal Chem 90: 374-397. [crossref]
  48. Chaves ED, Rusinol AE, Vance DE, Campenot RB, Vance JE (1997) Role of lipoproteins in the delivery of lipids to axons during axonal regeneration. J Biol Chem 272: 30766-30773. [crossref]
  49. Fester L, Zhou L, Bütow A, Huber C, von Lossow R, et al. (2009) Cholesterol promoted synaptogenesis requires the conversion of cholesterol to estradiol in the hippocampus. Hippocampus 19: 692-705. [crossref]
  50. Sang N, Chen C (2006) Lipid signalling and synaptic plasticity. Neuroscientist 12: 425-434. [crossref]
  51. Hussain G, Wang J, Rasul A, et al. (2019) Role of cholesterol and sphingolipids in brain development and neurological diseases. Lipids Health Dis 18: 1-2. [crossref]
  52. Jorge C, Farez MF (2015) The role of astrocytes in multiple sclerosis progression. Front Neurol 6: 180. [crossref]
  53. Joe EH, Choi DJ, An J, et al. (2018) Astrocytes, microglia, and parkinson’s disease. Exp Neurobiol 27: 77-87. [crossref]
  54. Almad A, Maragakis NJ (2018) A stocked toolbox for understanding the role of astrocytes in disease. Nat Rev Neurol 14: 351-362. [crossref]
  55. Belanger M, Allaman I, Magistretti PJ (2011) Brain energy metabolism: Focus on astrocyte-neuron metabolic cooperation. Cell Metab 14: 724-738. [crossref]
  56. Ullian EM, Sapperstein SK, Christopherson KS, et al. (2001) Control of synapse number by glia. Science 291: 657-661. [crossref]
  57. Varcianna A, Myszczynska MA, Castelli LM, et al. (2018) Micro-RNAs secreted through astrocyte-derived extracellular vesicles cause neuronal network degeneration in C9orf72 ALS. Ebiomedicine 40: 626-635. [crossref]
  58. Chung WS, Allen NJ, Eroglu C (2015) Astrocytes control synapse formation, function, and elimination. Cold Spring Harb Perspect Biol 7: a020370. [crossref]
  59. Sofroniew MV, Vinters HV, Vinters HV (2013) REVIEW Astrocytes: Biology and pathology. Acta Neuropathol 119: 7-35. [crossref]
  60. Deitmer JW, Theparambil SM, Ruminot I, et al. (2019) Energy dynamics in the brain: Contributions of astrocytes to metabolism and pH homeostasis. Front Neurosci-Switz 13: 1301. [crossref]
  61. Ballabh P, Braun A, Nedergaard M (2004) The blood-brain barrier: An overview: Structure, regulation, and clinical implications. Neurobiol Dis 16: 1-13. [crossref]
  62. Abbott NJ, Patabendige AAK, Dolman DEM, et al (2010) Structure and function of the blood–brain barrier. Neurobiol Dis 37: 13-25. [crossref]
  63. Edmond J (2001) Essential polyunsaturated fatty acids and the barrier to the brain. J Mol Neurosci 16: 181-193. [crossref]
  64. Mitchell RW, Hatch GM (2011) Fatty acid transport into the brain: Of fatty acid fables and lipid tails. Prostaglandins Leukot Essent. Fatty Acids 85: 293-302. [crossref]
  65. Saunders NR, Ek CJ, Habgood MD (2008) Barriers in the brain: A renaissance? Trends Neurosci 31: 279-286. [crossref]
  66. Bradbury MW (1984) The structure and function of the blood-brain barrier. Federation Proceedings 43: 186-190.
  67. Betsholtz C (2014) Physiology: Double function at the blood brain barrier. Nature 509: 432-433.
  68. Daneman R, Prat A (2015) The Blood鈥揃rain Barrier. Csh Perspect Biol 7: 32-34.
  69. Ferreira L (2019) What human blood-brain barrier models can tell us about BBB function and drug discovery? Expert Opin Drug Dis 14: 1113-1123. [crossref]
  70. Moura RP, Martins C, Pinto S, et al. (2019) Blood-brain barrier receptors and transporters: An insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Del 16: 271-285. [crossref]
  71. Dienel GA, Cruz NF (2016) Aerobic glycolysis during brain activation: Adrenergic regulation and influence of norepinephrine on astrocytic metabolism. J Neurochem 138: 14-52. [crossref]
  72. Goyal MS, Hawrylycz M, Miller JA, et al. (2014) Aerobic glycolysis in the human brain is associated with development and neotenous gene expression. Cell Metab 19: 49-57. [crossref]
  73. Abe T, Takahashi S, Suzuki N (2006) Oxidative metabolism in cultured rat astroglia: Effects of reducing the glucose concentration in the culture medium and of D-aspartate or potassium stimulation. J Cereb Blood Flow Metab 26: 153-160. [crossref]
  74. Cataldo AM, Broadwell RD (1986) Cytochemical identification of cerebral glycogen and glucose-6-phosphatase activity under normal and experimental conditions. II. Choroid plexus and ependymal epithelia, endothelia and pericytes. J Neurocytol 15: 511-524. [crossref]
  75. Pfeiffer-Guglielmi B, Fleckenstein B, Jung G, et al. (2010) Immunocytochemical localization of glycogen phosphorylase isozymes in rat nervous tissues by using isozyme-specific antibodies. J Neurochem 85: 73-81. [crossref]
  76. Camargo N, Goudriaan A, Deijk ALFV, et al. (2017) Oligodendroglial myelination requires astrocyte-derived lipids. Plos Biol 15: e1002605. [crossref]
  77. Van Deijk ALF, Camargo N, Timmerman J, et al. (2017) Astrocyte lipid metabolism is critical for synapse development and function in vivo. 65: 670-682. [crossref]
  78. Ioannou MS, Jesse J, Shu-Hsien S, et al. (2020) Neuron-Astrocyte Metabolic Coupling Protects against Activity-Induced Fatty Acid Toxicity. Cell 177: 1522-1535. [crossref]
  79. Ebert D, Haller RG, Walton ME (2003) Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J Neurosci 23: 5928-5935. [crossref]
  80. Chang CY, Ke DS, Chen JY (2009) Essential fatty acids and human brain. Acta Neurol Taiwan 18: 231-241. [crossref]
  81. Edmond J, Robbins RA, Bergstrom JD, et al. (2010) Capacity for substrate utilization in oxidative metabolism by neurons, astrocytes, and oligodendrocytes from developing brain in primary culture. J Neurosci Res 18: 551-561. [crossref]
  82. Takahashi S, Iizumi T, Mashima K, et al. (2014) Roles and regulation of ketogenesis in cultured astroglia and neurons under hypoxia and hypoglycemia. Asn Neuro 6: [crossref]
  83. Simard JR, Pillai BK, Hamilton JA (2008) Fatty acid flip-flop in a model membrane is faster than desorption into the aqueous phase. Biochemistry 47: 9081-9089. [crossref]
  84. Mitchell RW, On NH, Bigio MRD, et al. (2011) Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. J Neurochem 117: 735-746. [crossref]
  85. Marianne Haag (2003) Essential fatty acids and the brain. Can J Psychiatry 48: 195-203.
  86. Tracey TJ, Steyn FJ, Wolvetang EJ, et al. (2018) Neuronal lipid metabolism: Multiple pathways driving functional outcomes in health and disease. Front Mol Neurosci 11: 10. [crossref]
  87. Crawford MA, Casperd NM, Sinclair AJ (1976) The long chain metabolites of linoleic and linolenic acid in liver and brain in herbivores and carnivores. Comparative biochemistry and physiology. Comp Biochem Physiol B 54: 395-401. [crossref]
  88. Crawford MA, Broadhurst CL, Guest M, et al. (2013) A quantum theory for the irreplaceable role of docosahexaenoic acid in neural cell signalling throughout evolution. Prostaglandins Leukot Essent Fatty Acids 88: 5-13. [crossref]
  89. Bazinet RP, Lay S (2014) Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat Rev Neurosci 15: 771-785. [crossref]
  90. Luchtman DW, Song C (2013) Cognitive enhancement by omega-3 fatty acids from child-hood to old age: Findings from animal and clinical studies. 64: 550-565. [crossref]
  91. Denis I, Potier B, Vancassel S, et al (2013) Omega-3 fatty acids and brain resistance to ageing and stress: Body of evidence and possible mechanisms. Ageing Res Rev 12: 579-594. [crossref]
  92. Igarashi M, Santos RA, Cohen-Cory S (2015) Impact of Maternal n-3 Polyunsaturated Fatty Acid Deficiency on Dendritic Arbor Morphology and Connectivity of DevelopingXenopus laevisCentral NeuronsIn Vivo. J Neurosci 35: 6079-6092. [crossref]
  93. Lim GP, Calon F, Morihara T, et al. (2005) A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged alzheimer mouse model. J Neurosci 25: 3032-3040. [crossref]
  94. Michael A, Crawford (2006) Docosahexaenoic acid in neural signaling systems. Nutrition & Health
  95. Hamilton JA, Hillard CJ, Spector AA, et al. (2007) Brain uptake and utilization of fatty acids, lipids and lipoproteins: Application to neurological disorders. J Mol Neurosci 33: 2-11. [crossref]
  96. Farooqui AA, Horrocks LA, Farooqui T (2000) Glycerophospholipids in brain: Their metabolism, incorporation into membranes, functions, and involvement in neurological disorders. Chem Phys Lipids 106: 1-29. [crossref]
  97. Van Meer G, Voelker DR, Feigenson GW (2008) Membrane lipids: Where they are and how they behave. Nat Rev Mol Cell Bio 9: 112-124. [crossref]
  98. Harayama T, Riezman H (2018) Understanding the diversity of membrane lipid composition. Nat Rev Mol Cell Biol 19: 281-296. [crossref]
  99. Li J, Wang X, Zhang T, et al. (2015) A review on phospholipids and their main applications in drug delivery systems. Asian J Pharm Sci 10: 81-98. [crossref]
  100. Ross BM, Moszczynska A, Blusztajn JK, et al. (1997) Phospholipid biosynthetic enzymes in human brain. Lipids 32: 351-358. [crossref]
  101. Paltauf F, Hermetter A (1990) Phospholipids natural, semisynthetic, synthetic: Phospholipids 1-12. [crossref]
  102. Tracey TJ, Steyn FJ, Wolvetang EJ, et al. (2018) Neuronal lipid metabolism: Multiple pathways driving functional outcomes in health and disease. Front Mol Neurosci 11: 10. [crossref]
  103. Montaner A, da Silva Santana TT, Schroeder T, et al. (2018) Author correction: Specific phospholipids regulate the acquisition of neuronal and astroglial identities in Post-Mitotic cells. Sci Rep 9: 20222. [crossref]
  104. Sonnino S, Prinetti A (2012) Membrane domains and the lipid raft concept. Curr Med Chem. 20: 4-21. [crossref]
  105. West RJ, Briggs L, Perona Fjeldstad M (2018) Sphingolipids Regulate Neuromuscular Synapse Structure and function in Drosophila. J Comp Neurol 526: 1995-2009. [crossref]
  106. Vance Dennis E (2008) Biochemistry of lipids, lipoproteins and membranes (Fifth edition).:v-vi
  107. Hannun YA, Obeid LM (2018) Author Correction: Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Bio 19: 673. [crossref]
  108. Schnaar Ronald L (2016) Gangliosides of the vertebrate nervous system. J Mol Biol 428: 3325-3336. [crossref]
  109. Novgorodov SA, Voltin JR, Wang W, et al. (2019) Acid sphingomyelinase deficiency protects mitochondria and improves function recovery after brain injury. J Lipid Res 60: 609-623. [crossref]
  110. Hartmann D, Lucks J, Fuchs S, et al. (2012) Long chain ceramides and very long chain ceramides have opposite effects on human breast and colon cancer cell growth. Int J Biochem Cell Biol 44: 620-628. [crossref]
  111. Xu R, Wang K, Mileva I, et al. (2016) Alkaline ceramidase 2 and its bioactive product sphingosine are novel regulators of the DNA damage response. Oncotarget 7: 18440-18457. [crossref]
  112. Gomez-Mu Oz A, Presa N, Gomez-Larrauri A, et al. (2016) Control of inflammatory responses by ceramide, sphingosine 1phosphate and ceramide 1-phosphate. Prog Lipid Res 61: 51-62. [crossref]
  113. Blom T, Somerharju P, Ikonen E (2011) Synthesis and biosynthetic trafficking of membrane lipids. Cold Spring Harb Perspect Biol 3: a4713. [crossref]
  114. Björkhem I, Meaney S (2004) Brain cholesterol: Long secret life behind a barrier. Arterioscler Thromb Vasc Biol 24: 806-815. [crossref]
  115. Kang S, Kim CH, Jung H, et al. (2017) Agmatine ameliorates type 2 diabetes induced-Alzheimer’s disease-like alterations in high-fat diet-fed mice via reactivation of blunted insulin signalling. Neuropharmacology 113: 467-479. [crossref]
  116. Saeed AA, Genove G, Li T, Lutjohann D, Olin M, et al. (2014) Effects of a disrupted Blood-Brain barrier on cholesterol homeostasis in the brain. J Biol Chem [crossref]
  117. Reinhart MP, Billheimer JT, Faust JR, Gaylor JL (1987) Subcellular localization of the enzymes of cholesterol biosynthesis and metabolism in rat liver. J Biol Chem 262: 9649. [crossref]
  118. Muse ED, Jurevics H, Toews AD, Matsushima GK, Morell P (2010) Parameters related to lipid metabolism as markers of myelination in mouse brain. J Neurochem 76: 77-86. [crossref]
  119. Zarrouk, Amira, Debbabi, Meryam, Bezine, Maryem, et al. (2018) Lipid biomarkers in alzheimer’s disease. Curr Alzheimer Res 15: 303-312. [crossref]
  120. Genaro-Mattos TC, Anderson A, Allen LB, Korade Z, Mirnics K (2019) Cholesterol biosynthesis and uptake in developing neurons. Acs Chem Neurosci [crossref]
  121. Deijk V, Anne-Lieke F, Camargo, Nutabi, Timmerman Jaap, et al. (2017) Astrocyte lipid metabolism is critical for synapse development and function in vivo. Glia 65: 670-682. [crossref]
  122. Wang H, Eckel RH (2014) What are lipoproteins doing in the brain? Trends in Endocrinology & Metabolism Tem 25: 8-14. [crossref]
  123. Orth M, Bellosta S (2012) Cholesterol: Its regulation and role in central nervous system disorders. Cholesterol 2012: 292598. [crossref]
  124. Pitas RE, Boyles JK, Lee SH, Hui D, Weisgraber KH (1987) Lipoproteins and their receptors in the central nervous system. Characterization of the lipoproteins in cerebrospinal fluid and identification of apolipoprotein B,E (LDL) receptors in the brain. J Biol Chem 262: 14352-14360. [crossref]
  125. Jing C, Zhang X, Kusumo H, Costa LG, Guizzetti M (2013) Cholesterol efflux is differentially regulated in neurons and astrocytes: Implications for brain cholesterol homeostasis. Biochim Biophys Acta 1831. [crossref]
  126. Rushworth JV, Hooper NM (2010) Lipid rafts: Linking alzheimer’s amyloidbeta production, aggregation, and toxicity at neuronal membranes. International Journal of Alzheimer’s Disease 2011: 603052. [crossref]
  127. Kabouridis PS, Janzen J, Magee AL, Ley SC (2015) Cholesterol depletion disrupts lipid rafts and modulates the activity of multiple signaling pathways in T lymphocytes. Eur J Immunol 30: 954-963. [crossref]
  128. Pike LJ (2003) Lipid rafts bringing order to chaos. J Lipid Res 44: 655-667. [crossref]
  129. Hussain G, Wang J, Rasul A, Anwar H, Imran A, et al. (2019) Role of cholesterol and sphingolipids in brain development and neurological diseases. Lipids Health Dis 18. [crossref]
  130. Mauch DH, Ngler K, Schumacher S, Gritz C, Miller E, Otto A, et al. (2001) CNS synaptogenesis promoted by Glia-Derived cholesterol. Science 294. [crossref]
  131. Fukui K, Ferris HA, Kahn CR (2015) Effect of Cholesterol Reduction on Receptor Signaling in Neurons. J Biol Chem 290: 26383-26392. [crossref]
  132. Chaves ED, Rusinol AE, Vance DE, Campenot RB, Vance JE (1997) Role of lipoproteins in the delivery of lipids to axons during axonal regeneration. J Biol Chem 272: 30766. [crossref]
  133. Xue-Shan Z, Juan P, Qi W, Zhong R, Li-Hong P, et al. (2016) Imbalanced cholesterol metabolism in Alzheimer’s disease. Clin Chim Acta 456: 107-114. [crossref]
  134. Kandimalla R, Thirumala V, Reddy PH (2016) Is Alzheimer’s disease a Type 3 Diabetes? A Critical Appraisal. Biochimica et Biophysica Acta 1863: 1078-1089. [crossref]
  135. S Sonnino, A Prinetti (2013) Membrane domains and the “lipid raft” concept. Curr Med Chem [crossref]
  136. Caliceti C, Zambonin L, Prata C, Sega F, Hakim G, et al. (2012) Effect of plasma membrane cholesterol depletion on glucose transport regulation in leukemia cells. Plos One 7. [crossref]
  137. Group BMJP (2009) Alzheimer’s disease. BMJ 338.
  138. Mattson MP (2016) Pathways towards and away from Alzheimer’s disease. Nature 430: 631-639. [crossref]
  139. Vishal S, Sourabh A, Harkirat S (2011) Alois Alzheimer (1864-1915) and the Alzheimer syndrome. Journal of Medical Biography 19: 32-33. [crossref]
  140. D’Errico P, Luehmann MM (2020) Mechanisms of pathogenic tau and a protein spreading in alzheimer’s disease. Front Aging Neurosci 12: 265. [crossref]
  141. Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82: 239-259. [crossref]
  142. El Kadmiri, Said N, Slassi Ilham, Moutawakil BE, Nadifi S (2018) Biomarkers for alzheimer disease: Classical and novel candidates’ review. Neuroscience 370: 181-190. [crossref]
  143. Foley P (2010) Lipids in Alzheimer’s disease: A century-old story. BBA – Molecular and Cell Biology of Lipids 1801: 750-753. [crossref]
  144. Alzheimer A (1907) Uber eine eigenartige Erkrankung der Hirnride. Allg.z.psychiatr
  145. Kim M, Nevado-Holgado A, Whiley L, Snowden SG, Soininen H, et al. (2016) Association between plasma ceramides and phosphatidylcholines and hippocampal brain volume in late onset alzheimer’s disease. Journal of Alzheimers Disease 60: 809-817. [crossref]
  146. González-Domínguez R, García-Barrera T, Vitorica J, Gómez-Ariza JL (2015) Metabolomic screening of regional brain alterations in the APP/PS1 transgenic model of Alzheimer’s disease by direct infusion mass spectrometry. J Pharmaceut Biomed 102: 425-435. [crossref]
  147. Bennett S, Valenzuela N, Xu H, Franko B, Fai S, et al. (2013) Using neurolipidomics to identify phospholipid mediators of synaptic (dys)function in Alzheimer’s Disease. Front Physiol 4: 168. [crossref]
  148. Kao YC, Ho PC, Tu YK, Jou IM, Tsai KJ (2020) Lipids and alzheimer’s disease. Int J Mol Sci 21: 1505. [crossref]
  149. Grimm MO, Michaelson D, Hartmann T (2017) Omega-3 fatty acids, lipids and apoE lipidation in Alzheimer’s disease: A rationale for multi-nutrient dementia prevention. J Lipid Res 58: 2083-2101. [crossref]
  150. Wood PL (2012) Lipidomics of Alzheimer’s disease: Current status. Alzheimers Research & Therapy 4: 5. [crossref]
  151. Bales KR (2010) Brain lipid metabolism, apolipoprotein E and the pathophysiology of Alzheimer’s disease. Neuropharmacology 59: 295-302. [crossref]
  152. Ledesma MD, Martin MG, Dotti CG (2012) Lipid changes in the aged brain: Effect on synaptic function and neuronal survival. Prog Lipid Res 51: 23-35. [crossref]
  153. Sultana R, Perluigi M, Allan Butterfield D (2013) Lipid peroxidation triggers neurodegeneration: A redox proteomics view into the Alzheimer disease brain. Free Radical Biology & Medicine 62: 157-169. [crossref]
  154. Tian-Bi, Zhu, Zhao, Zhang, Piao, Luo, et al. (2018) Lipid metabolism in Alzheimer’s disease. Brain Res Bull
  155. Nasaruddin ML, Holscher C, Kehoe P, Graham SF, Green BD (2016) Wideranging alterations in the brain fatty acid complement of subjects with late Alzheimer’s disease as detected by GC-MS. Am J Transl Res 8: 154-165. [crossref]
  156. Nasaruddin ML, Pan X, Mcguinness B, Passmore P, Kehoe PG, et al. (2018) Evidence that parietal lobe fatty acids may be more profoundly affected in moderate alzheimer’s disease (AD) pathology than in severe AD pathology. Metabolites 8: 69. [crossref]
  157. Wood PL, Barnette BL, Kaye JA, Quinn JF, Woltjer RL (2015) Non-targeted lipidomics of CSF and frontal cortex grey and white matter in control, mild cognitive impairment, and Alzheimer’s disease subjects. Acta Neuropsychiatr 27: 270-278. [crossref]
  158. Belkouch M, Hachem M, Elgot A, Van AL, Picq M, et al. (2016) The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer’s disease. J Nutr Biochem 38: 1-11. [crossref]
  159. Sarrafpour S, Ormseth C, Chiang A, Arakaki X, Harrington M, et al. (2019) Lipid Metabolism in Late-Onset Alzheimer’s Disease Differs from Patients Presenting with Other Dementia Phenotypes. Int J Env Res Pub He 16: 1995. [crossref]
  160. Wong MW, Braidy N, Poljak A, Pickford R, Thambisetty M (2017) Dysregulation of lipids in Alzheimer’s disease and their role as potential biomarkers. Alzheimer’s & Dementia 13: 810-827. [crossref]
  161. Xiong H, Callaghan D, Jones A, Walker DG, Lue LF, et al. (2008) Cholesterol retention in Alzheimer’s brain is responsible for high – and secretase activities and a production. Neurobiol Dis 29: 422-437. [crossref]
  162. Pfrieger FW (2003) Cholesterol homeostasis and function in neurons of the central nervous system. Cellular & Molecular Life Sciences 60: 1158-1171. [crossref]
  163. Chang TY, Yamauchi Y, Hasan MT, Chang C (2017) Cellular cholesterol homeostasis and Alzheimers J Lipid Res 58: 2239-2254. [crossref]
  164. Scala CD, Chahinian H, Yahi N, Garmy N, Fantini J (2014) Interaction of alzheimer’s-Amyloid peptides with cholesterol: Mechanistic insights into amyloid pore formation. Biochemistry 53: 4489-4502. [crossref]
  165. Area-Gomez E, Maria DCLC, Tambini MD, Guardia-Laguarta C, De Groof AJC, et al. (2012) Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. Embo J 31: 4106-4123.
  166. Area-Gomez E, Schon EA (2016) Mitochondria-associated ER membranes and Alzheimer disease. Curr Opin Genet Dev 38: 90-96. [crossref]
  167. Pera M, Larrea D, Guardia Laguarta C, Montesinos J, Velasco KR, Agrawal RR, et al. (2017) Increased localization of APP C99 in mitochondria associated ER membranes causes mitochondrial dysfunction in Alzheimer disease. The EMBO Journal 36: 3356-3371. [crossref]
  168. Pera M, Montesinos J, Larrea D, Agrawal RR, Area-Gomez E (2020) MAM and C99, key players in the pathogenesis of Alzheimer’s disease. Int Rev Neurobiol 154: 235-278. [crossref]
  169. Andrew P Bailey, Grielof Koster, Christelle Guillermier, Hirst EMA, Macrae JI, et al. (2015) Antioxidant role for lipid droplets in a stem cell niche of drosophila. Cell 163: 340-353. [crossref]
  170. Liu L, Zhang K, Sandoval H, Yamamoto S, Jaiswal M, et al. (2015) Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration. Cell 160: 177-190. [crossref]
  171. Liu L, Mackenzie KR, Putluri N, Maleti-Savati M, Bellen HJ (2017) The Glia-Neuron lactate shuttle and elevated ROS promote lipid synthesis in neurons and lipid droplet accumulation in glia via APOE/D. Cell Metab 26: 719-737. [crossref]
  172. Wong MW, Braidy N, Poljak A, Sachdev PS (2017) The application of lipidomics to biomarker research and pathomechanisms in Alzheimer’s disease. Curr Opin Psychiatr 30: 136-144. [crossref]
  173. Zhamg AH, Ma MZ, Kong L, Gao HL, Sun H (2020) High-throughput lipidomics analysis to discover lipid biomarkers and profiles as potential targets for evaluating efficacy of an against APP/PS1 transgenic mice based on UPLC S. Biomed Chromatogr 34: 4724. [crossref]
  174. Barupal DK, Baillie R, Fan S, Saykin AJ, Kaddurah-Daouk R (2019) Sets of coregulated serum lipids are associated with Alzheimer’s disease pathophysiology. Alzheimer s & Dementia Diagnosis Assessment & Disease Monitoring 11: 619-627. [crossref]
  175. Kim J, Basak JM, Holtzman DM (2009) The role of apolipoprotein e in alzheimer’s disease. Neuron 63: 287-303. [crossref]
  176. Monteiro-Cardoso VF, Oliveira MM, Melo T, Domingues MRM, Videira RA (2015) Cardiolipin profile changes are associated to the early synaptic mitochondrial dysfunction in alzheimer’s disease. Journal of Alzheimers Disease 43: 1375-1392. [crossref]
  177. Basu Ball W, Neff JK, Gohil VM (2017) The role of nonbilayer phospholipids in mitochondrial structure and function. Febs Lett 592: 1273-1290. [crossref]
  178. Calzada E, Onguka O, Claypool SM (2016) Phosphatidylethanolamine metabolism in health and disease. Int Rev Cel Mol Bio 321: 29-88. [crossref]
  179. Fonteh AN, Cipolla M, Chiang J, Arakaki X, Harrington MG (2014) Human cerebrospinal fluid fatty acid levels differ between supernatant fluid and Brain Derived nanoparticle fractions, and are altered in alzheimer’s disease. Plos One 9: 100519. [crossref]
  180. Giudetti AM, Shu G, Vergara D, Fonteh AN (2020) Polyunsaturated fatty acid composition of cerebrospinal fluid fractions shows their contribution to cognitive resilience of a pre-symptomatic alzheimer’s disease cohort. Front Physiol 11: 1-14.
  181. Wilde MD, Vellas B, Girault E, Yavuz AC, Sijben JW (2017) Lower brain and blood nutrient status in Alzheimer’s disease: Results from meta-analyses. Alzheimers & Dementia: Translational Research & Clinical Interventions 3: 416-431. [crossref]
  182. Snowden SG, Ebshiana AA, Hye A, An Y, Thambisetty M (2017) Association between fatty acid metabolism in the brain and Alzheimer disease neuropathology and cognitive performance: A nontargeted metabolomic study. Plos Med 14: 1002266. [crossref]
  183. Mh A, Apab C, Nb A, Jc A, Psa D (2020) Blood fatty acids in Alzheimer’s disease and mild cognitive impairment: A meta-analysis and systematic review. Ageing Res Rev 60: 101043. [crossref]
  184. Wood PL, Barnette BL, Kaye JA, Quinn JF, Woltjer RL (2015) Non-targeted lipidomics of CSF and frontal cortex grey and white matter in control, mild cognitive impairment and Alzheimer’s disease subjects. Acta Neuropsychiatr 27: 270-278. [crossref]
  185. Dyall SC (2015) Long-chain omega-3 fatty acids and the brain: A review of the independent and shared effects of EPA, DPA and DHA. Front Aging Neurosci 7: 52. [crossref]
  186. Di Iorio, Benedetta Bartali, Annamaria Corsi, Stefania Bandinelli, Mark P Mattson, et al. (2007) Low plasma N-3 fatty acids and dementia in older persons: The InCHIANTI study. J Gerontol A Biol Med 62: 1120-1126. [crossref]
  187. Thomas MH, Pelleieux S, Vitale N, Olivier JL (2016) Dietary arachidonic acid as a risk factor for age-associated neurodegenerative diseases: Potential mechanisms. Biochimie: 168-177. [crossref]
  188. Goozee K, Chatterjee P, James I, Shen K, Sohrabi HR, Asih PR, et al. (2017) Alterations in erythrocyte fatty acid composition in preclinical Alzheimer’s disease. Sci Rep-Uk
    7.
  189. Prasad MR, Lovell MA, Yatin M, Dhillon H, Markesbery WR (1998) Regional membrane phospholipid alterations in alzheimer’s disease. Neurochem Res 23: 81-88. [crossref]
  190. Cunnane SC, Schneider JA, Tangney C, Tremblay-Mercier J, Morris MC (2012) Plasma and brain fatty acid profiles in mild cognitive impairment and alzheimer’s disease. Journal of Alzheimer’s disease: JAD 29: 691-697. [crossref]
  191. Iwamoto N, Kobayashi K, Kosaka K (1989) The formation of prostaglandins in the postmortem cerebral cortex of Alzheimer-type dementia patients. J Neurol 236: 80-84. [crossref]
  192. Wood PL (2012) Lipidomics of Alzheimer’s disease: Current status. Alzheimers Research & Therapy 4: 5. [crossref]
  193. Fonteh AN, Chiang J, Cipolla M, Hale J, Diallo F, et al. (2013) Alterations in cerebrospinal fluid glycerophospholipids and phospholipase A2 activity in Alzheimer’s disease. J Lipid Res 54: 2884-2897. [crossref]
  194. A LW, A AS, B JH, A PP, C DG, et al. (2014) Evidence of altered phosphatidylcholine metabolism in Alzheimer’s disease. Neurobiol Aging 35: 271-278. [crossref]
  195. Guan Z, Wang Y, Cairns NJ, Lantos PL, Gustav D, et al. (1999) Decrease and structural modifications of phosphatidylethanolamine plasmalogen in the brain with alzheimer disease. J Neuropathol Exp Neurol 58: 740-747. [crossref]
  196. Farooqui AA, Horrocks LA (1998) Plasmalogen-selective phospholipase A2 and its involvement in Alzheimer’s disease. Biochem Soc T 26: 243-246. [crossref]
  197. Filippov V, Song MA, Zhang K, Vinters HV, Duerksen-Hughes PJ (2012) Increased ceramide in brains with alzheimer’s and other neurodegenerative diseases. J Alzheimers Dis 29(3):537-547. [crossref]
  198. Alfred N Fonteh, Cora Ormseth, Jiarong Chiang, Cipolla M, Arakaki X, et al. (2015) Sphingolipid metabolism correlates with cerebrospinal fluid Beta amyloid levels in Alzheimer’s disease. Plos One 10: 0125597. [crossref]
  199. Lepara O, Valjevac A, Alajbegovi A, Zairagi A, Nakas-I indi E (2009) Decreased serum lipids in patients with probable Alzheimer’s disease. Bosnian Journal of Basic Medical ences 9: 215-220. [crossref]
  200. Bernath MM, Bhattacharyya S, Nho K, Barupal DK, Saykin AJ (2018) Serum triglycerides in Alzheimer’s disease: Relation to neuroimaging and CSF biomarkers. Neurology 94: 2088-2098. [crossref]
  201. Hascalovici JR, Vaya J, Khatib S, Holcroft CA, Schipper HM (2009) Brain sterol dysregulation in sporadic AD and MCI: Relationship to heme oxygenase-1. J Neurochem 110: 1241-1253. [crossref]
  202. Heverin M, Bogdanovic N, Lutjohann D, Bayer T, Bjrkhem I (2004) Changes in the levels of cerebral and extracerebral sterols in the brain of patients with Alzheimer’s disease. J Lipid Res 45: 186-193. [crossref]
  203. Tanzi T RE (2018) The genetics of alzheimer disease.
  204. Guerreiro RJ, Gustafson DR, Hardy J (2012) The genetic architecture of Alzheimer’s disease: Beyond APP, PSENs and APOE. Neurobiol Aging 33: 437-456. [crossref]
  205. Holtzman DM, Herz J, Bu G (2012) Apolipoprotein e and apolipoprotein e receptors: Normal biology and roles in alzheimer disease. Csh Perspect Med 2: a6312. [crossref]
  206. RW Mahley (1988) Apolipoprotein E: Cholesterol transport protein with expanding role in cell biology. Science 240: 622-630. [crossref]
  207. Kim J, Basak JM, Holtzman DM (2009) The role of apolipoprotein e in alzheimer’s disease. Neuron 63: 287-303. [crossref]
  208. Serrano-Pozo A, Das S, Hyman BT (2021) APOE and Alzheimer’s disease: Advances in genetics, pathophysiology, and therapeutic approaches. The Lancet Neurology 20: 68-80. [crossref]
  209. Tachibana M, Holm ML, Liu CC, Shinohara M, Kanekiyo T (2019) APOE4mediated amyloid-beta pathology depends on its neuronal receptor LRP1. J Clin Invest 129. [crossref]
  210. Castellano JM, Kim J, Stewart FR, Jiang H, Demattos RB, et al. (2011) Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med 3: 57r-89r. [crossref]
  211. Verghese PB, Castellano JM, Garai K, Wang Y, Hong J, Shah A, et al. (2013) ApoE influences amyloid-{beta} (A{beta}) clearance despite minimal apoE/A{beta} association in physiological conditions. Neuroscience 110: E1807-1816. [crossref]
  212. Basak JM, Verghese PB, Yoon H, Kim J, Holtzman DM (2012) Low-density lipoprotein receptor represents an apolipoprotein E-independent pathway of a uptake and degradation by astrocytes. J Biol Chem 287: 13959-13971. [crossref]
  213. Kanekiyo T, Zhang J, Liu Q, Liu CC, Bu G (2011) Heparan sulphate proteoglycan and the Low-Density lipoprotein Receptor-Related protein 1 constitute major pathways for neuronal amyloid- uptake. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience 31: 1644-1651. [crossref]
  214. Sagare A, Deane R, Bell RD, Johnson B, Hamm K, Pendu R, et al. (2007) Clearance of amyloid-beta by circulating lipoprotein receptors. Nat Med 13: 1029-1031. [crossref]
  215. Castellano JM, Deane R, Gottesdiener AJ, Verghese PB, Stewart FR, et al. (2012) Low-density lipoprotein receptor overexpression enhances the rate of brain-to-blood clearance in a mouse model of-amyloidosis. P Natl Acad Sci Usa 109: 15502. [crossref]
  216. Deane ASBV (2008) The role of the cell surface LRP and soluble LRP in blood-brain barrier Abeta clearance in Alzheimer’s disease. Curr Pharm Design 14. [crossref]
  217. Bales KR, Verina T, Dodel RC, Du Y, Altstiel L, Bender M, et al. (1997) Lack of apolipoprotein E dramatically reduces amyloid |[beta]|-peptide deposition. Nat Genet 17: 263-264. [crossref]
  218. Fryer JD (2005) Human apolipoprotein e4 alters the amyloid-? 40:42 ratio and promotes the formation of cerebral amyloid angiopathy in an amyloid precursor protein transgenic model. J Neurosci 25: 2803-2810. [crossref]
  219. Holtzman DM (2000) Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences 97: 2892-2897. [crossref]
  220. Fagan AM, Watson M, Parsadanian M, Bales KR, Holtzman DM (2002) Human and murine ApoE markedly alters a beta metabolism before and after plaque formation in a mouse model of Alzheimer’s disease. Neurobiol Dis 9: 305-318.
  221. Shadlen MF (1998) Effects of Age and Ethnicity on the Link Between APOE4 and Alzheimer Disease. JAMA [crossref]
  222. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Pericak-Vance MA (1993) Gene dose of Apolipoprotein E type 4 allele and the risk of Alzheimers disease in late onset families. Science 8: 41-43. [crossref]
  223. Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PHS, et al. (1993) Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurology (8). [crossref]
  224. Roses AD (1996) Apolipoprotein E alleles as risk factors in Alzheimer’s disease. Annu Rev Med 47: 387-400. [crossref]
  225. Mahley RW, Huang Y, Rall SC (1999) Pathogenesis of type III hyperlipoproteinemia (dysbetalipoproteinemia): Questions, quandaries, and paradoxes. J Lipid Res 40: 1933-1949. [crossref]
  226. Liu CC, Kanekiyo T, Xu H, Bu G (2013) Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat Rev Neurol 9: 106-118. [crossref]
  227. Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS, Pericak-Vance M, et al. (1993) Binding of human apolipoprotein E to synthetic amyloid beta peptide: Isoform-specific effects and implications for late-onset Alzheimer disease. Proc Natl Acad Sci U S A 90: 8098-8102. [crossref]
  228. Lefterov I, Wolfe CM, Fitz NF, et al. (2019) APOE2 orchestrated differences in transcriptomic and lipidomic profiles of postmortem AD brain. Alzheimer’s Research and Therapy 11: 1-6. [crossref]
  229. Juva K, Verkkoniemi A, Viramo P, Polvikoski T, Kainulainen K, et al. (2000) Apolipoprotein e, cognitive function, and dementia in a general population aged 85 years and over. Int Psychogeriatr 12: 379-387. [crossref]
  230. O’Donoghue MC, Murphy SE, Zamboni G, Nobre AC, Mackay CE (2018) APOE genotype and cognition in healthy individuals at risk of Alzheimer’s disease: A review. Cortex 104: 103-123. [crossref]
  231. Trachtenberg AJ, Filippini N, Cheeseman J, et al. (2012) The effects of APOE on brain activity do not simply reflect the risk of Alzheimer’s disease. Neurobiol Aging. 33: 618.e1-618.e13. [crossref]
  232. Boisvert MM, Erikson GA, Shokhirev MN, Allen NJ (2018) The aging astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep 22: 269-285. [crossref]
  233. Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, et al. (2012) Genomic analysis of reactive astrogliosis. J Neurosci 32: 6391-6410. [crossref]
  234. Zhao N, Ren Y, Yamazaki Y, Qiao W, Li F, et al. (2020) Alzheimer’s risk factors age, APOE genotype, and sex drive distinct molecular pathways. Neuron 106: 727-742. [crossref]
  235. Dulewicz M, Kulczyska-Przybik A, Sowik A, Borawska R, Mroczko B (2021) Fatty acid binding protein 3 (FABP3) and apolipoprotein e4 (ApoE4) as lipid Metabolism-Related biomarkers of alzheimer’s disease. J Clin Med 10: 3009. [crossref]
  236. Rawat V, Wang S, Jian S, Bar R, Liraz O, et al. (2019) ApoE4 alters ABCA1 membrane trafficking in astrocytes. J Neurosci 39: 9611-9622. [crossref]
  237. Qiao F, Gao XP, Yuan L, Cai HY, Qi JS (2014) Apolipoprotein e4 impairs in vivo hippocampal Long-Term synaptic plasticity by reducing the phosphorylation of CaMKII伪 and CREB. Journal of Alzheimers Disease Jad 41: 1165-1176. [crossref]
  238. GA Rodriguez, MP, Burns, Weeber EJ, Rebeck GW (2013) Young APOE4 targeted replacement mice exhibit poor spatial learning and memory, with reduced dendritic spine density in the medial entorhinal cortex. Learn Memory 20: 256-266. [crossref]
  239. Terrisse L, Poirier J, Bertrand P, Merched A, Visvikis S, et al. (1998) Increased levels of apolipoprotein d in cerebrospinal fluid and hippocampus of alzheimer’s patients. J Neurochem 71: 1643-1650. [crossref]
  240. Lin YT, Seo J, Gao F, Feldman HM, Wen HL, et al. (2018) APOE4 causes widespread molecular and cellular alterations associated with alzheimer’s disease phenotypes in human iPSC-Derived brain cell types – ScienceDirect. Neuron 98: 1141-1154. [crossref]
  241. Sienski G, Narayan P, Bonner JM, Kory N, Boland S, et al. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med 13: eaaz4564. [crossref]
  242. Qi G, Mi Y, Shi X, Gu H, Brinton RD, et al. (2021) ApoE4 impairs Neuron-Astrocyte coupling of fatty acid metabolism. Cell Rep 34: 108572. [crossref]
  243. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, et al. (2014) An RNA-Sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34: 11929-11947. [crossref]
  244. Watkins PA (1997) Fatty acid activation. Progress in lipid research 36: 55-83.
  245. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, et al. (2015) Tissue-based map of the human proteome. Science 347: 1260419. [crossref]
  246. Ellis JM, Frahm JL, Li LO, Coleman RA (2010) Acyl-coenzyme a synthetases in metabolic control. Curr Opin Lipidol 21: 212. [crossref]
  247. Steinberg SJ, Morgenthaler J, Heinzer AK, Smith KD, Watkins PA (2000) Very long-chain Acyl-CoA synthetases HUMAN “BUBBLEGUM” REPRESENTS a NEW FAMILY of PROTEINS CAPABLE of ACTIVATING VERY LONG-CHAIN FATTY ACIDS. J Biol Chem 275: 35162-35169. [crossref]
  248. Pei Z, Oey NA, Zuidervaart MM, Jia Z, Li Y, et al. (2003) The Acyl-CoA synthetase \”bubblegum\” (Lipidosin): FURTHER CHARACTERIZATION and ROLE in NEURONAL FATTY ACID? -OXIDATION. J Biol Chem 278: 47070-47078. [crossref]
  249. Li MD, Burns TC, Morgan AA, Khatri P (2014) Integrated multi-cohort transcriptional meta-analysis of neurodegenerative diseases. Acta Neuropathol Com 2: 1-23. [crossref]
  250. Jun G, Ibrahim-Verbaas CA, Vronskaya M, Lambert JC, Chung J, et al. (2016) A novel Alzheimer disease locus located near the gene encoding tau protein. Mol Psychiatry 21: 108-117. [crossref]
  251. Ellis JM, Bowman CE, Wolfgang MJ (2015) Metabolic and Tissue-Specific regulation of Acyl-CoA metabolism. Plos One 10: e0116587. [crossref]
  252. Marszalek JR, Kitidis C, Dararutana A, Lodish HF (2004) Acyl-CoA synthetase 2 overexpression enhances fatty acid internalization and neurite outgrowth. J Biol Chem 279: 23882-23891. [crossref]
  253. Klett EL, Chen S, Yechoor A, Lih FB, Coleman RA, et al. (2017) Long-chain acylCoA synthetase isoforms differ in preferences for eicosanoid species and long-chain fatty acids. J Lipid Res 58: 884-894. [crossref]
  254. Horn CGV, Caviglia JM, Li LO, Wang S, Granger DA, et al. (2005) Characterization of recombinant long-chain rat acyl-CoA synthetase isoforms 3 and 6: Identification of a novel variant of isoform 6. Biochemistry-Us 44: 1635-1642 [crossref]
  255. Kim HC, Lee SW, Cho YY, Lim JM, Ryoo ZY, et al. (2009) RNA interference of long-chain acyl-CoA synthetase 6 suppresses the neurite outgrowth of mouse neuroblastoma NB41A3 cells. Mol Med Rep 2: 669-674. [crossref]
  256. Wang X, Zhu M, Hjorth E, Cortés-Toro V, Eyjolfsdottir H, et al. (2015) Resolution of inflammation is altered in Alzheimer’s disease. Alzheimer’s & Dementia 11: 40-50. [crossref]
  257. Fernandez RF, Kim SQ, Zhao Y, Foguth RM, Weera MM, et al. (2018) Acyl-CoA synthetase 6 enriches the neuroprotective omega-3 fatty acid DHA in the brain. Proc Natl Acad Sci 115: 12525-12530. [crossref]
  258. Chouinard-Watkins R, Bazinet RP (2018) ACSL6 is critical for maintaining brain DHA levels. P Natl Acad Sci Usa 115: 12343-12345. [crossref]
  259. Fernandez RF, Pereyra AS, Diaz V, Wilson ES, Litwa KA, et al. (2021) Acyl-CoA synthetase 6 is required for brain docosahexaenoic acid retention and neuroprotection during aging. JCI Insight 6: e144351. [crossref]
  260. Fernandez RF, Ellis JM (2020) Acyl-CoA Synthetases as regulators of brain phospholipid acyl-chain diversity. Prostaglandins, Leukotrienes and Essential Fatty Acids. 161: 102175. [crossref]
  261. Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, et al. (2015) Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci 18: 1819-1831. [crossref]
  262. Atlas AMB (2009) Allen brain atlas. Neuron
  263. Orre M, Kamphuis W, Osborn LM, Melief J, Kooijman L, et al. (2014) Acute isolation and transcriptome characterization of cortical astrocytes and microglia from young and aged mice. Neurobiol Aging 35: 1-4. [crossref]
  264. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, et al. (2008) Transcriptome database for astrocytes, neurons, and oligodendrocytes: A new resource for understanding brain development and function. J Neurosci 28: 264-278. [crossref]
  265. Soupene E, Kuypers FA (2008) Mammalian Long-Chain Acyl-CoA synthetases. Experimental Biology & Medicine 233: 507-521. [crossref]
  266. Hale BJ, Fernandez RF, Kim SQ, Diaz VD, Jackson SN, et al. (2019) Acyl-CoA Synthetase 6 enriches seminiferous tubules with the omega-3 fatty acid DHA and is required for male fertility in the mouse. J Biol Chem 294: 14394-14405. [crossref]
  267. Soupene E, Kuypers FA (2006) Multiple erythroid isoforms of human longchain acyl-CoA synthetases are produced by switch of the fatty acid gate domains. Bmc Mol Biol 7: 1-2. [crossref]
  268. Lee EJ, Kim HC, Yong YC, Byun SJ, Lim JM, et al. (2005) Alternative promotion of the mouse acyl-CoA synthetase 6 (mAcsl6) gene mediate the expression of multiple transcripts with 5′-end heterogeneity: Genetic organization of mAcsl6 variants. Biochem Biophys Res Commun 327: 84-93. [crossref]
  269. Malhotra KT, Malhotra K, Lubin BH, Kuypers FA (1999) Identification and molecular characterization of acyl-CoA synthetase in human erythrocytes and erythroid precursors. Biochem J 3441: 135-143. [crossref]
  270. Soupene E, Dinh NP, Siliakus M, Kuypers FA (2010) Activity of the acylCoA synthetase ACSL6 isoforms: Role of the fatty acid Gate-domains. Bmc Biochem 11: 1-3. [crossref]
  271. Gilquin B, Taillebourg E, Cherradi N, Hubstenberger A, Gay O, et al. (2010) The AAA+ ATPase ATAD3A controls mitochondrial dynamics at the interface of the inner and outer membranes. Molecular & Cellular Biology 30: 1984-96. [crossref]
  272. Desai R, Frazier AE, Durigon R, Patel H, Jones AW, et al. (2017) ATAD3 gene cluster deletions cause cerebellar dysfunction associated with altered mitochondrial DNA and cholesterol metabolism. Brain 140: 1595-1610. [crossref]
  273. Zhao Y, Sun X, Hu D, et al. (2019) ATAD3A oligomerization causes neurodegeneration by coupling mitochondrial fragmentation and bioenergetics defects. Nat Commun 10: 1-20.
  274. Harel T, Yoon WH, Garone C, Gu S, Coban-Akdemir Z, et al. (2016) Recurrent de novo and biallelic variation of ATAD3A, encoding a mitochondrial membrane protein, results in distinct neurological syndromes. Am J Hum Genet 99: 831-845. [crossref]
  275. Janikiewicz J, Szymański J, Malinska D, Patalas-Krawczyk P, Michalska B, et al. (2018) Mitochondria-associated membranes in aging and senescence: Structure,function, and dynamics. Cell Death Dis 9: 1-2. [crossref]
  276. Hayashi T, Rizzuto R, Hajnoczky G, Su TP (2009) MAM: More than just a housekeeper. Trends Cell Biol 19: 81-88. [crossref]
  277. Rusiol AE, Cui Z, Chen MH, Vance JE (1994) A unique mitochondria associated membrane fraction from rat liver has a high capacity for lipid synthesis and contains pre-Golgi secretory proteins including nascent lipoproteins. J Biol Chem 269: 27494-27502. [crossref]
  278. Vance JE (2003) Molecular and cell biology of phosphatidylserine and phosphatidylethanolamine metabolism. Prog Nucleic Acid Res Mol Biol 75: 69-111. [crossref]
  279. Mitsuo Tagaya, Kohei Arasaki (2017) Regulation of mitochondrial dynamics and autophagy by the Mitochondria-Associated membrane. Advances in Experimental Medicine & Biology 997: 33-47. [crossref]
  280. Csords G, Vrnai P, Golenr T, Roy S, Purkins G, et al. (2010) Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol Cell 39: 121-132. [crossref]
  281. Zhao Y, Hu D, Wang R, Sun X, Ropelewski P, et al. (2022) ATAD3A oligomerization promotes neuropathology and cognitive deficits in Alzheimer’s disease models. Nat Commun 13: 1-20. [crossref]
  282. Fathia Djelti, Braudeau J, Hudry E, Dhenain M, Varin J, et al. (2015) CYP46A1 inhibition, brain cholesterol accumulation and neurodegeneration pave the way for Alzheimer’s disease. Brain 138: 2383-2398. [crossref]
  283. Montesinos J, Pera M, Larrea D, Guardia-Laguarta C, Agrawal RR, et al. (2020) The Alzheimer’s disease associated C99 fragment of APP regulates cellular cholesterol trafficking. The EMBO Journal 39: e103791. [crossref]
  284. Weigel D, J Ckle H (1990) The fork head domain: A novel DNA binding motif of eukaryotic transcription factors? Cell. 63: 455-456. [crossref]
  285. Flachsbart F, Caliebe A, Kleindorp R, Blanché H, von Eller-Eberstein H, et al. (2009) Association of FOXO3Avariation with human longevity confirmed in German centenarians. Proceedings of the National Academy of Sciences 106: 2700-2705. [crossref]
  286. Willcox BJ, Portion TA, He Q, Chen R, Grove JS, et al. (2008) FOXO3A genotype is strongly associated with human longevity. Proceedings of the National Academy of Sciences 105: 13987-13992. [crossref]
  287. Shimokawa I, Komatsu T, Hayashi N, Kim SE, Kawata T, et al. (2015) The life-extending effect of dietary restriction requires Foxo3 in mice. Aging Cell 14: 707-709. [crossref]
  288. Chiacchiera F, Simone C (2010) The AMPK-FoxO3A axis as a target for cancer treatment. Cell cycle (Georgetown, Tex.) 9: 1091-1096. [crossref]
  289. Armando V, Burgering B (2007) Stressing the role of FoxO proteins in lifespan and disease. Nat Rev Mol Cell Biol 8: 440-450. [crossref]
  290. Beekman M, Nederstigt C, Suchiman H, Kramer D, Breggen R, et al. (2010) Genome-wide association study (GWAS)-identified disease risk alleles do not compromise human longevity. Proceedings of the National Academy of Sciences 107: 18046-18049. [crossref]
  291. Zettergren A, Kern S, Ryd茅n L, Blennow K, Zetterberg H, et al. (2018) Genetic variation in FOXO3 is associated with Self-Rated health in a Population-Based sample of older individuals. J Gerontol A Biol Med 73: 1453. [crossref]
  292. Soerensen M, Nygaard M, Dato S, Stevnsner T, Christiansen L (2015) Association study of FOXO3A SNPs and aging phenotypes in Danish oldest-old individuals. Aging Cell 14: 60-66. [crossref]
  293. Paik J, Ding Z, Narurkar R, Ramkissoon S, Muller F, et al. (2009) Fox Os cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5: 540-553. [crossref]
  294. Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, et al. (2009) FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5: 527-539. [crossref]
  295. Schffner I, Minakaki G, Khan MA, Balta EA, Schlötzer-Schrehardt U, et al. (2018) FoxO function is essential for maintenance of autophagic flux and neuronal morphogenesis in adult neurogenesis. Neuron 99: 1188-1203. [crossref]
  296. Yeo H, Lyssiotis CA, Zhang Y, Ying H, Asara JM, et al. (2013) FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells. Embo J 32: 2589-2602. [crossref]
  297. Caballero‐Caballero A, Engel T, Martinez‐Villarreal J, Sanz-Rodriguez A, Chang P, et al. (2013) Mitochondrial localization of the Forkhead box class O transcription factor FOXO3a in brain. J Neurochem 124: 749-756. [crossref]
  298. Lehtinen MK, Yuan Z, Boag PR, Yang Y, Villén J, et al. (2006) A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell 125: 987-1001. [crossref]
  299. Sriram K (2006) Deficiency of TNF receptors suppresses microglial activation and alters the susceptibility of brain regions to MPTP-induced neurotoxicity: Role of TNF-Faseb J 20: 670-682. [crossref]
  300. Woiciechowsky C, Sch枚ning B, Stoltenburg-Didinger G, Stockhammer F, Volk HD (2004) Brain-IL-1 beta triggers astrogliosis through induction of IL-6: Inhibition by propranolol and IL-10. Med Sci Monit 10: 325-330. [crossref]
  301. Buffo A, Rolando C, Ceruti S (2010) Astrocytes in the damaged brain: Molecular and cellular insights into their reactive response and healing potential. Biochem Pharmacol 79: 77-89. [crossref]
  302. Cui M, Huang Y, Tian C, et.al (2011) FOXO3a inhibits TNF– and IL-1-induced astrocyte proliferation: Implication for reactive astrogliosis. Glia 59: 641-654. [crossref]
  303. Du S, Jin F, Maneix L, et al. (2021) FoxO3 deficiency in cortical astrocytes leads to impaired lipid metabolism and aggravated amyloid pathology. Aging cell 20: e13432. [crossref]
  304. Cui M, Huang Y, Tian C, Zhao Y, Zheng J (2011) FOXO3a inhibits TNF– and IL-1-induced astrocyte proliferation: Implication for reactive astrogliosis. Glia 59: 641-654. [crossref]
  305. Nguyen TB, Louie SM, Daniele JR, Tran Q, Dillin A, et al. (2017) DGAT1-Dependent Lipid Droplet Biogenesis Protects Mitochondrial Function during Starvation-Induced Autophagy. Dev Cell 42: 9-21. [crossref]
  306. Ehehalt Robert, Keller P, Haass C, Thiele C, Simons K (2003) Amyloidogenic processing of the Alzheimer beta-amyloid precursor protein depends on lipid rafts. J Cell Biol 160: 113-123. [crossref]
  307. Saido T, Leissring MA (2012) Proteolytic degradation of amyloid beta-Protein. Csh Perspect Med 2: a6379. [crossref]
  308. He G, Luo W, Li P, Remmers C, Netzer WJ, et al. (2010) Gamma-secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature 467: 95-98. [crossref]
  309. Chu J, Li JG, Joshi YB, Giannopoulos PF, Hoffman NE, et al. (2015) Gamma Secretase-Activating protein is a substrate for caspase-3: Implications for alzheimer’s disease. Biol Psychiat 77: 720-728. [crossref]
  310. Perez SE, Nadeem M, Malek-Ahmadi MH, He B, et al. (2017) Frontal cortex and hippocampal-Secretase activating protein levels in prodromal alzheimer disease. Neurodegener Dis 17: 235-241. [crossref]
  311. Satoh J, Tabunoki H, Ishida T, Saito Y, Arima K (2012) Immunohistochemical characterization of secretase activating protein expression in Alzheimer’s disease brains. Neuropathology & Applied Neurobiology 38: 132-141. [crossref]
  312. Floudas CS, Um N, Kamboh M, Barmada MM, Visweswaran S (2014) Identifying genetic interactions associated with late-onset Alzheimer’s disease. Biodata Min 7: 35. [crossref]
  313. Zhu M, Tao Y, He Q, Gao H, Song F, et al. (2014) A common GSAP promoter variant contributes to Alzheimer’s disease liability. Neurobiol Aging 35: 2651-2656. [crossref]
  314. Xu P, Chang JC, Zhou X, Wang W, Greengard P (2021) GSAP regulates lipid homeostasis and mitochondrial function associated with Alzheimer’s disease. The Journal of experimental medicine 218: e20202446. [crossref]
  315. Jin Chu, Elisabetta Lauretti, Caryne P, Praticò D (2014) Pharmacological modulation of GSAP reduces amyloid- levels and tau phosphorylation in a mouse model of Alzheimer’s disease with plaques and tangles. Journal of Alzheimers Disease Jad 41: 729-737. [crossref]
  316. Pera M, Larrea D, Guardia aguarta C, Montesinos J, Velasco KR, et al. (2017) Increased localization of APP C99 in mitochondria鈥恆ssociated ER membranes causes mitochondrial dysfunction in Alzheimer disease. The EMBO Journal. [crossref]
  317. Prete DD, Suski JM, Ouls B, Debayle D, Gay AS, et al. (2016) Localization and processing oftheAmyloid-beta protein precursor inMitochondria-Associated membranes. Journal of Alzheimer\”s Disease 55: 1549-1570. [crossref]
  318. Area-Gomez E, Maria DCLC, Tambini MD, Guardia-Laguarta C, De Groof AJC, et al. (2012) Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. Embo J 31: 4106-4123. [crossref]
  319. L Hedskog, CM Pinho, Filadi R, Rönnbäck A, Hertwig L, et al. (2013) Modulation of the endoplasmic reticulum-mitochondria interface in Alzheimer’s disease and related models. Proceedings of the National Academy of Sciences 110: 7916-7921. [crossref]
  320. Adami PVM, Nichtov Z, Weaver DB, Bartok A, Wisniewski T, et al. (2019) Perturbed mitochondriaR contacts in live neurons that model the amyloid pathology of Alzheimer’s disease. The Company of Biologists 132: jcs229906. [crossref]
  321. Kosicek M, Hecimovic S (2013) Phospholipids and alzheimer’s disease: Alterations, mechanisms and potential biomarkers. Int J Mol Sci 14: 1310-1322. [crossref]
  322. Goodenberger M, Jenkins RB (2012) Genetics of adult glioma. Cancer Genet-Ny 205: 613-621. [Crossref]
  323. Venur VA, Peereboom DM, Ahluwalia MS (2015) Current medical treatment of glioblastoma. Cancer Treatment & Research 163: 103-115. [crossref]
  324. Joo K, Kim J, Jin J, Kim M, Seol H, et al. (2013) Patient-Specific orthotopic glioblastoma xenograft models recapitulate the histopathology and biology of human glioblastomas in situ. Cell Rep [crossref]
  325. Ricard D, Idbaih A, Ducray F, Lahutte M, Hoangxuan K, et al. (2012) Primary brain tumours in adults. Lancet 379: 1984-1996. [crossref]
  326. Libby CJ, Tran AN, Scott SE, Griguer C, Hjelmeland AB (2018) The protumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells. Biochimica Et Biophysica Acta 1869: 175-188. [crossref]
  327. Agnihotri S, Zadeh G (2015) Metabolic reprogramming in glioblastoma: The influence of cancer metabolism on epigenetics and unanswered questions. Neuro Oncology 18: 160-172.[crossref]
  328. Intlekofer AM, Finley LWS (2019) Metabolic signatures of cancer cells and stem cells. Nature Metabolism 1: 177-188. [crossref]
  329. Weinberg H, Hanahan D (2011) Hallmarks of cancer: The next generation. Cell 144: 646-674. [crossref]
  330. Lopez-Lazaro M (2008) The warburg effect: Why and how do cancer cells activate glycolysis in the presence of oxygen? Anti-Cancer Agents in Medicinal. Chemistry (Formerly Current Medicinal Chemistry – Anti-Cancer Agents) 8. [crossref]
  331. Duraj T, Garcromero N, Carrinavarro J, Madurga R, Ana O, et al. Beyond the warburg effect: Oxidative and glycolytic phenotypes coexist within the metabolic heterogeneity of glioblastoma. Cells-Basel 10: 202. [crossref]
  332. Hoangnh LB, Siebzehnrubl FA, Yang C, Tano SS, Deleyrolle LP (2018) Infiltrative and drug鈥恟esistant slowcling cells support metabolic heterogeneity in glioblastoma. The EMBO Journal 37: e98772. [crossref]
  333. Caren Duman, Kaneschka Yaqubi, Angelika Hoffmann, Acikgöz AA, Korshunov A, et al. (2019) AcylCoA-Binding protein drives glioblastoma tumorigenesis by sustaining fatty acid oxidation. Cell Metab 30: 274-289. [crossref]
  334. Antonella DR, Serena P, Marco R, Patrizia T, Letizia M, et al. (2012) A radial glia gene marker, fatty acid binding protein 7 (FABP7), is involved in proliferation and invasion of glioblastoma cells. Plos One 7: e52113. [crossref]
  335. Gupta K, Vuckovic I, Zhang S, Xiong Y, Carlson BL, et al. (2020) Radiation induced metabolic alterations associate with tumor aggressiveness and poor outcome in glioblastoma. Frontiers Media S. A 10: 535. [crossref]
  336. Son B, Lee S, Kim H, Kang H, Youn BH (2020) Decreased FBP1 expression rewires metabolic processes affecting aggressiveness of glioblastoma. Oncogene 39: 36-49. [crossref]
  337. Hua L, Shaan P, Affleck VS, Ian W, Turnbull DM, et al. (2017) Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells. Neuro-Oncology 43-54. [crossref]
  338. Hanna VS, Ebtisam A (2018) Synopsis of arachidonic acid metabolism: A review. J Adv Res 23-32. [crossref]
  339. Juric-Sekhar G, Zarkovic K, Waeg G, Cipak A, Zarkovic N (2009) Distribution of 4-hydroxynonenal-protein conjugates as a marker of lipid peroxidation and parameter of malignancy in astrocytic and ependymal tumors of the brain. Tumori 95: 762-768. [crossref]
  340. Zajdel A, Wilczok A, Slowinski J, Mazurek OU (2007) Aldehydic lipid peroxidation products in human brain astrocytomas. J Neuro-Oncol 84: 167-174. [crossref]
  341. Zarkovic K, Juric G, Waeg G, Kolenc D, Zarkovic N (2010) Immunohistochemical appearance of HNE-protein conjugates in human astrocytomas. Biofactors 24: 33-40. [crossref]
  342. Atilla-Gokcumen G, Muro E, Relat-Goberna J, Sasse S, Bedigian A, Coughlin M, et al. (2014) Dividing cells regulate their lipid composition and localization. Cell 156. [crossref]
  343. Lopez DH, Bestard-Escalas J, Garate J, Maim贸-Barcel贸 A, Fern谩ndez R, Reigada R, et al. (2018) Tissue-selective alteration of ethanolamine plasmalogen metabolism in dedifferentiated colon mucosa. Biochimica et Biophysica Acta (BBA) – Molecular and Cell Biology of Lipids:S263724189. [crossref]
  344. Sang TL, Lee JC, Kim JW, Cho SY, Seong JK, Moon MH (2016) Global changes in lipid profiles of mouse cortex, hippocampus, and hypothalamus upon p53 knockout. Sci Rep-Uk 6: 36510. [crossref]
  345. Baenke F, Peck B, Miess H, Schulze A (2013) Hooked on fat: The role of lipid synthesis in cancer metabolism and tumour development. Dis Model Mech 6: 1353-1363. [crossref]
  346. Gimple RC, Kidwell RL, Ljy K, Sun T, Gromovsky AD, et al. (2019) Glioma stem cell specific super enhancer promotes polyunsaturated fatty acid synthesis to support EGFR signaling. Cancer Discov. [crossref]
  347. Zou YK, Watters A, Cheng N, Perry CE, Chen Q (2019) Polyunsaturated fatty acids from astrocytes activate PPARγ signaling in cancer cells to promote brain metastasis. Cancer Discov 9: 1720-1735. [crossref]
  348. Cheng X, Geng F, Pan M, Wu X, Guo D (2020) Targeting DGAT1 ameliorates glioblastoma by increasing fat catabolism and oxidative stress. Cell Metab [crossref]
  349. Omuro A, Deangelis LM (2013) Glioblastoma and other malignant gliomas: A clinical review. Journal of the American Medical Association 310: 1842-1850. [crossref]
  350. Wen PY, Kesari S (2008) Malignant gliomas in adults. New Engl J Med 359: 492-507. [crossref]
  351. Accioly MT, Pacheco P, Maya-Monteiro CM, Carrossini N, Robbs BK, et al. (2008) Lipid bodies are reservoirs of cyclooxygenase-2 and sites of Prostaglandin-E2 synthesis in colon cancer cells. Cancer Res 68: 1732-1740. [crossref]
  352. Du W, Zhang L, Brett-Morris A, Aguila B, Kerner J, et al. (2017) HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism. Nat Commun 8: 1769. [crossref]
  353. Feng G, Xiang C, Wu X, Ji YY, Guo D (2017) Inhibition of SOAT1 suppresses glioblastoma growth via blocking SREBP-1-mediated lipogenesis. Clin Cancer Res 22. [crossref]
  354. Geng F, Guo D (2017) Lipid droplets, potential biomarker and metabolic target in glioblastoma. Intern Med Rev 3: 10. [crossref]
  355. Mitra R, Le TT, Gorjala P, Goodman OB (2017) Positive regulation of prostate cancer cell growth by lipid droplet forming and processing enzymes DGAT1 and ABHD5. Bmc Cancer 17: 631. [crossref]
  356. Pucer A, Brglez V, Payr C, Pungerar J, Petan T (2013) Group X secreted phospholipase A2 induces lipid droplet formation and prolongs breast cancer cell survival. Mol Cancer 12: 111. [crossref]
  357. Sevinsky CJ, Faiza K, Leila K, Anza D, Rao MK, et al. (2018) NDRG1 regulates neutral lipid metabolism in breast cancer cells. Breast Cancer Res 20: 55. [crossref]
  358. Sunami Y, Rebelo A, Kleeff J (2017) Lipid metabolism and lipid droplets in pancreatic cancer and stellate cells. Cancers 10: 3. [crossref]
  359. Tauchi-Sato K, Ozeki S, Houjou T, Taguchi R, Fujimoto T (2002) The surface of lipid droplets is a phospholipid monolayer with a unique fatty acid composition. J Biol Chem 277: 44507-44512. [crossref]
  360. Wilfling F, Haas JT, Walther TC, Jr R (2014) Lipid droplet biogenesis. Curr Opin Cell Biol [crossref]
  361. Olzmann James A, Carvalho Pedro (2019) Dynamics and functions of lipid droplets. 20: 137-155. [crossref]
  362. Paar M, Jungst C, Steiner NA, Magnes C, Sinner F, et al. (2012) Remodeling of lipid droplets during lipolysis and growth in adipocytes. J Biol Chem 287: 11164-11173. [crossref]
  363. Klionsky DJ, Emr SD (2000) Autophagy as a regulated pathway of cellular degradation. Science 290: 1717-1721. [crossref]
  364. Mizushima N, Komatsu M (2011) Autophagy: Renovation of cells and tissues. Cell 147: 728-741. [crossref]
  365. Mizushima N, Levine B, Cuervo AM, Klionsky DJ (2008) Autophagy fights disease through cellular self-digestion. Nature 451: 1069. [crossref]
  366. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, et al. (2009) Autophagy regulates lipid metabolism. Nature 458: 1131-1135. [crossref]
  367. Wu X, Geng F, Cheng X, Guo Q, Zhong Y, et al. (2020) Lipid droplets maintain energy homeostasis and glioblastoma growth via autophagic release of stored fatty acids. iScience 23: 101569. [crossref]
  368. Guo D, Reinitz F, Youssef M, Hong C, Nathanson D, Akhavan D, et al. (2011) An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov 1: 442-456. [crossref]
  369. Bjorkhem I (2004) Brain cholesterol: Long secret life behind a barrier. Arteriosclerosis, Thrombosis, and Vascular Biology 24: 806-815. [crossref]
  370. Dietschy JM, Turley SD (2001) Cholesterol metabolism in the brain. Curr Opin Lipidol 12: 105-112. [crossref]
  371. Hayashi H, Campenot RB, Vance DE, Vance JE (2004) Glial lipoproteins stimulate axon growth of central nervous system neurons in compartmented cultures. J Biol Chem 279: 14009-14015. [crossref]
  372. Karten B, Campenot RB, Vance DE, Vance JE (2006) Expression of ABCG1, but not ABCA1, correlates with cholesterol release by cerebellar astroglia. J Biol Chem 281: 4049-4057. [crossref]
  373. Wahrle SE, Jiang H, Parsadanian M, Legleiter J, Han X, et al. (2004) ABCA1 is required for normal central nervous system ApoE levels and for lipidation of astrocyte-secreted apoE. J Biol Chem 279: 40987-40993. [crossref]
  374. Orth M, Bellosta S (2012) Cholesterol: Its regulation and role in central nervous system disorders. Cholesterol 2012: 292598. [crossref]
  375. Jing C, Zhang X, Kusumo H, Costa LG, Guizzetti M (2013) Cholesterol efflux is differentially regulated in neurons and astrocytes: Implications for brain cholesterol homeostasis. Biochimica et Biophysica Acta (BBA) – Molecular and Cell Biology of Lipids 1831: 263-275. [crossref]
  376. Repa JJ (2000) Regulation of absorption and ABC1-Mediated efflux of cholesterol by RXR heterodimers. Science 289: 1524-1529. [crossref]
  377. Venkateswaran Asha, Laffitte Bryan A (2000) Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXR alpha. P Natl Acad Sci Usa 97: 12097-12102. [crossref]
  378. Zelcer N, Hong C, Boyadjian R, Tontonoz P (2009) LXR regulates cholesterol uptake through Idol-Dependent ubiquitination of the LDL receptor. Science 325: 100-104. [crossref]
  379. Villa G, Hulce J, Zanca C, Bi J, Mischel P (2016) An LXR-Cholesterol axis creates a metabolic Co-Dependency for brain cancers. Cancer Cell 30: 683-693. [crossref]
  380. Friedman HS, Kerby T, Calvert H (2000) Temozolomide and treatment of malignant glioma. Clinical cancer research: an official journal of the American Association for Cancer Research 6: 2585-2597. [crossref]
  381. Stupp R, Mason WP, Van D, Weller M, Fisher B, et al. (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma – Cancer/Radiothrapie 9: 196-197. [crossref]
  382. Massimo Aureli, Valentina Murdica, Nicoletta Loberto, Samarani M, Prinetti A, et al. (2014) Exploring the link between ceramide and ionizing radiation. Glycoconjugate J 31: 449-459. [crossref]
  383. Brady RO, Kanfer JN, Mock MB, Fredrickson DS (1966) The metabolism of sphingomyelin. II. Evidence of an enzymatic deficiency in Niemann-Pick diseae. Proceedings of the National Academy of Sciences 55: 366-369. [crossref]
  384. Haimovitz-Friedman A (1994) Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis. J Exp Med 180: 525-535. [crossref]
  385. Senchenkov A, Litvak DA, Cabot MC (2001) Targeting ceramide metabolism strategy for overcoming drug resistance. JNCI J Natl Cancer Inst 93: 347-357. [crossref]
  386. Mizushima N, Koike R, Kohsaka H, Kushi Y, Handa S, et al. (1996) Ceramide induces apoptosis via CPP32 activation. Febs Lett 395: 261-271. [crossref]
  387. Abuhusain HJ, Matin A, Qiao Q, Shen H, Kain N, et al. (2013) A metabolic shift favoring sphingosine 1-Phosphate at the expense of ceramide controls glioblastoma angiogenesis. J Biol Chem 288: 37355-37364. [crossref]
  388. Ji-Sun J, Young-Ho A, Byung-In M, Hee-Sun K (2016) Exogenous c2 ceramide suppresses matrix metalloproteinase gene expression by inhibiting ROS production and MAPK signaling pathways in PMA-Stimulated human astroglioma cells. Int J Mol Sci 17: 477. [crossref]
  389. Nganga R, Oleinik N, Ogretmen B (2018) Mechanisms of Ceramide-Dependent cancer cell death. Adv Cancer Res [crossref]
  390. Gault CR, Obeid LM, Hannun YA (2010) An overview of sphingolipid metabolism: From synthesis to breakdown. Oxygen Transport to Tissue XXXIII 688: 1-23. [Crossref]
  391. Buehrer BM, Bell RM (1992) Inhibition of sphingosine kinase in vitro and in platelets. Implications for signal transduction pathways. J Biol Chem 267: 3154-3159. [crossref]
  392. Brocklyn J, Young N, Roof R (2003) Sphingosine-1-phosphate stimulates motility and invasiveness of human glioblastoma multiforme cells. Cancer Lett 199: 53-60. [crossref]
  393. Bassi R, Anelli V, Giussani P, Tettamanti G, Viani P, et al. (2010) Sphingosine-1-phosphate is released by cerebellar astrocytes in response to bFGF and induces astrocyte proliferation through Gi-protein-coupled receptors. Glia 53: 621-630. [crossref]
  394. Anelli V, Gault CR, Cheng AB, Obeid LM (2008) Sphingosine kinase 1 is up-regulated during hypoxia in U87MG glioma cells. J Biol Chem 283: 3365-3375. [crossref]
  395. Strub GM, Maceyka M, Hait NC, Milstien S, Spiegel S (2010) Extracellular and intracellular actions of Sphingosine-1-Phosphate. Adv Exp Med Biol 688: 141-155. [crossref]
  396. Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Spiegel S (1996) Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature 381: 800-803. [crossref]
  397. Masayuki Nagahashi, Manabu Abe, Kenji Sakimura, Takabe K, Wakai T (2018) The role of sphingosine-1-phosphate in inflammation and cancer progression. Cancer Sci 109: 3671-3678. [crossref]
  398. Marfia G, Campanella R, Navone SE, Vito CD, Riccitelli E, et al. (2014) Autocrine/paracrine sphingosine phosphate fuels proliferative and stemness qualities of glioblastoma stem cells. Glia 62: 1968-1981. [crossref]
  399. Im DS, Clemens J, Macdonald TL, Lynch KR (2001) Characterization of the human and mouse sphingosine 1-phosphate receptor, S1P5 (Edg-8): Structure-activity relationship of sphingosine1-phosphate receptors. Biochemistry-Us 40: 14053-14060. [crossref]
  400. Hla T, Maciag T (1990) An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to Gprotein-coupled receptors. J Biol Chem 265: 9308-9313. [crossref]
  401. Masana MI, Brown RC, Pu H, Gurney ME, Dubocovich ML (1995) Cloning and characterization of a new member of the G-protein coupled receptor EDG family. Receptors Channels 3: 255-262. [crossref]
  402. Zondag GC, Postma FR, Etten IV, Verlaan I, Moolenaar WH (1998) Sphingosine 1-phosphate signalling through the G-protein-coupled receptor Edg-1. The Biochemical journal 330: 605-609. [crossref]
  403. Siehler S, Manning DR (2002) Pathways of transduction engaged by sphingosine 1-phosphate through G protein-coupled receptors. Biochimica et Biophysica Acta 1582: 94-99. [crossref]
  404. Sanchez T, Hla T (2004) Structural and functional characteristics of S1P receptors. J Cell Biochem 92. [crossref]
  405. Bien-Miller S, Lange S, Holm T, B枚hm A, Rauch BH (2016) Expression of S1P metabolizing enzymes and receptors correlate with survival time and regulate cell migration in glioblastoma multiforme. Oncotarget 7. [crossref]
  406. Yoshida Y, Nakada M, Sugimoto N, Harada T, Hayashi Y, et al. (2010) Sphingosine-1-phosphate receptor type 1 regulates glioma cell proliferation and correlates with patient survival. Int J Cancer 126: 2341-2352. [crossref]
  407. Bernhart E, Damm S, Wintersperger A, Nusshold C, Brunner AM, et al. (2015) Interference with distinct steps of sphingolipid synthesis and signaling attenuates proliferation of U87MG glioma cells. Biochem Pharmacol  [crossref]
  408. Young N, Brocklyn JRV (2007) Roles of Sphingosine-1-Phosphate (S1P) receptors in malignant behavior of glioma cells. Differential effects of S1P2 on cell migration and invasiveness. Exp Cell Res 313: 1615-1627. [crossref]
  409. Lepley D (2005) The g protein鈥揅oupled receptor S1P2 regulates Rho/Rho kinase pathway to inhibit tumor cell migration. Cancer Res 65: 3788. [crossref]
  410. Malchinkhuu E, Sato K, Maehama T, Mogi C, Tomura H, Ishiuchi S, et al. (2008) S1P(2) receptors mediate inhibition of glioma cell migration through Rho signaling pathways independent of PTEN. Biochem Biophys Res Commun 366(4):963-968 [crossref]
  411. Quint K, Stiel N, Neureiter D, Schlicker HU, Nimsky C, et al. (2014) The role of sphingosine kinase isoforms and receptors S1P1, S1P2, S1P3, and S1P5 in primary, secondary, and recurrent glioblastomas. Tumour Biol 35: 8979-8989. [crossref]
  412. Rothhammer V, Kenison JE, Tjon E, Takenaka MC, Lima KD, et al. (2017) Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation. Proceedings of the National Academy of ences 114: 201615413. [crossref]
  413. Sullivan SA, O Sullivan C, Healy LM, Dev KK, Sheridan GK (2018) Sphingosine 1鈥恜hosphate receptors regulate TLR4 innduced CXCL5 release from astrocytes and microglia. J Neurochem 144: 736-747. [crossref]
  414. Dai Z, Wu J, Chen F, Cheng Q, Zhang M, et al. (2019) Cxcl5 promotes the proliferation and migration of glioma cells in autocrine-and paracrinedependent manners. Oncol Rep 36: 3303-3310. [crossref]
  415. Windh RT, Lee MJ, Hla T, An S, Barr AJ, et al. (1999) Differential coupling of the sphingosine 1-Phosphate receptors edg-1, edg-3, and H218/Edg-5 to the gi, gq, and g12 families of heterotrimeric g proteins. J Biol Chem 274: 27351-27358. [crossref]
  416. Liu Y, Wang X, Li J, Tang J, Li B, et al. (2021) Sphingosine 1-Phosphate liposomes for targeted nitric oxide delivery to mediate anticancer effects against brain glioma tumors. Adv Mater.
  417. Bien-M枚ller S, Lange S, Holm T, B枚hm A, Rauch BH (2016) Expression of S1P metabolizing enzymes and receptors correlate with survival time and regulate cell migration in glioblastoma multiforme. Oncotarget 7. [crossref]
  418. Rekers NH, Sminia P, Peters GJ (2011) Towards tailored therapy of glioblastoma multiforme. J Chemotherapy 23: 187-199. [crossref]
  419. Bryan L, Paugh BS, Kapitonov D, Wilczynska KM, Alvarez SM, et al. (2008) Sphingosine-1-phosphate and interleukin-1 independently regulate plasminogen activator inhibitor-1 and urokinase-type plasminogen activator receptor expression in glioblastoma cells: Implications for invasiveness. Molecular Cancer Research Mcr 6: 1469. [crossref]
  420. Young N, Brocklyn JRV (2007) Roles of Sphingosine-1-Phosphate (S1P) receptors in malignant behavior of glioma cells. Differential effects of S1P2 on cell migration and invasiveness. Exp Cell Res 313: 1615-1627. [crossref]
  421. Sato K, Tomura H, Igarashi Y, Ui M, Okajima F (1999) Possible involvement of cell surface receptors in sphingosine 1-phosphate-induced activation of extracellular signal-regulated kinase in C6 glioma cells. Mol Pharmacol 55: 126-133. [crossref]
  422. Malchinkhuu E, Sato K, Horiuchi Y, Mogi C, Ohwada S, et al. (2005) Role of p38 mitogen-activated kinase and c-Jun terminal kinase in migration response to lysophosphatidic acid and sphingosine-1-phosphate in glioma cells. Oncogene 24: 6676-6688. [crossref]
  423. Brocklyn JRV, Young N, Roof R (2003) Sphingosine-1-phosphate stimulates motility and invasiveness of human glioblastoma multiforme cells. Cancer Lett 199: 53-60. [crossref]
  424. Yoshida Y, Nakada M, Harada T, Tanaka S, Furuta T, et al. (2010) The expression level of sphingosine-1-phosphate receptor type 1 is related to MIB-1 labeling index and predicts survival of glioblastoma patients. J Neuro-Oncol 98: 41-47. [crossref]
  425. Yoshida Y, Nakada M, Sugimoto N, Harada T, Hayashi Y, et al. (2010) Sphingosine-1-phosphate receptor type 1 regulates glioma cell proliferation and correlates with patient survival. Int J Cancer 126: 2341-2352. [crossref]
  426. Shimizu F, Watanabe TK, Shinomiya H, Nakamura Y, Fujiwara T (1997) Isolation and expression of a cDNA for human brain fatty acid-binding protein (BFABP). Biochimica et Biophysica Acta (BBA) – Gene Structure and Expression 1354: 24-28. [crossref]
  427. Xu LZ, Sanchez R, Sali A, Heintz N (1996) Ligand specificity of brain lipidbinding protein. J Biol Chem 271: 24711. [crossref]
  428. Kurtz A, Zimmer A, Schngen F, Brning G, Miller T (1994) The expression pattern of a novel gene encoding brain-fatty acid binding protein correlates with neuronal and glial cell development. Development 120: 2637-2649. [crossref]
  429. Owada Y, Yoshimoto T, Kondo H (1996) Spatio-temporally differential expression of genes for three members of fatty acid binding proteins in developing and mature rat brains. J Chem Neuroanat 12: 113-122. [crossref]
  430. Yun SW, Leong C, Zhai D, Tan YL, Lim L, et al. (2012) Neural stem cell specific fluorescent chemical probe binding to FABP7. P Natl Acad Sci Usa 109: 10214-10217.
  431. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, et al. (2003) Identification of a cancer stem cell in human brain tumors. Cancer Res 63: 5821. [crossref]
  432. Llaguno SA, Chen J, Parada LF (2009) Signalling in malignant astrocytomas: Role of neural stem cells and its therapeutic implications. Clinical Cancer Research An Official Journal of the American Association for Cancer Research 15: 7124-7129. [crossref]
  433. Kaloshi G, Mokhtari K, Carpentier C, Sophie T, Lejeune J, et al. (2007) FABP7 expression in glioblastomas: Relation to prognosis, invasion and EGFR status. J Neuro-Oncol 84: 245-248. [crossref]
  434. Liang YU, Maximilian D, Nathan W, Bollen AW, Aldape KD, et al. (2005) Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. Proc Natl Acad Sci USA 102: 5814-5819. [crossref]
  435. Liang Y, Bollen AW, Aldape KD, Gupta N (2006) Nuclear FABP7 immunoreactivity is preferentially expressed in infiltrative glioma and is associated with poor prognosis in EGFR-overexpressing glioblastoma. Bmc Cancer 6: 97. [crossref]
  436. Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, et al. (2014) Fatty acid uptake and lipid storage induced by HIF-1伪 contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep 9: 349-365.
  437. Hoangnh LB, Siebzehnrubl FA, Yang C, Tano SS, Deleyrolle LP (2018) Infiltrative and drug resistant slowcling cells support metabolic heterogeneity in glioblastoma. The EMBO Journal 37: e98772. [crossref]
  438. Xu X, Wang Y, Choi WS, Sun X, Godbout R (2021) Super resolution microscopy reveals DHA-dependent alterations in glioblastoma membrane remodelling and cell migration. Nanoscale 21.
  439. Paton CM, Ntambi JM (2008) Biochemical and physiological function of stearoyl-CoA desaturase. The American journal of physiology 297: E28-E37. [crossref]
  440. Matthew T Flowers, James M Ntambi (2008) Role of stearoyl-coenzyme a desaturase in regulating lipid metabolism. Curr Opin Lipidol 19: 248-256. [crossref]
  441. Wu X, Zou X, Chang Q, Zhang Y, Li Y, et al. (2013) The evolutionary pattern and the regulation of Stearoyl-CoA desaturase genes. Biomed Res Int 2013: 856521. [crossref]
  442. Ecker J, Liebisch G, Grandl M, Schmitz G (2010) Lower SCD expression in dendritic cells compared to macrophages leads to membrane lipids with less monounsaturated fatty acids. Immunobiology 215: 748-755. [crossref]
  443. Zhang L, Ge L, Parimoo S, Stenn K, Prouty SM (1999) Human stearoyl-CoA desaturase: Alternative transcripts generated from a single gene by usage of tandem polyadenylation sites. Biochem J 340: 255-264. [crossref]
  444. Noushmehr H, Weisenberger D, Diefes K, Phillips H, Pujara K, et al. (2010) The cancer genome atlas research network. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17: 510-522. [crossref]
  445. Menendez J, Lupu R (2007) Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 7: 763-777. [Crossref]
  446. Peck, Barrie, Schulze, Almut. (2016) Lipid desaturation – the next step in targeting lipogenesis in cancer? Febs J 283: 2767-2778. [crossref]
  447. Hilvo M, Denkert C, Lehtinen L, Muller B, Brockmoller S, et al. (2011) Novel theranostic opportunities offered by characterization of altered membrane lipid metabolism in breast cancer progression. Cancer Res 71: 3236-3245. [crossref]
  448. Scaglia N, Caviglia JM, Igal RA (2005) High stearoyl-CoA desaturase protein and activity levels in simian virus 40 transformed-human lung fibroblasts. Biochim Biophys Acta 1687: 141-151. [crossref]
  449. Brennan CW, Verhaak R, Mckenna A, Campos B, Noushmehr H, et al. (2013) The somatic genomic landscape of glioblastoma. Cell 155: 462-477. [crossref]
  450. Oatman N, Dasgupta N, Arora P, Choi K, Dasgupta B (2021) Mechanisms of stearoyl CoA desaturase inhibitor sensitivity and acquired resistance in cancer. Sci Adv 7: d7459. [crossref]
fig 7

Identification of Biomarkers in Colorectal Cancer Using a Multiplex Immunohistochemistry Technology

DOI: 10.31038/JCRM.2022554

Abstract

Colorectal Cancer (CRC) is a common malignant tumor with high mortality arising from adenomatous polyps of the large intestine. The rapid development of multiple immunofluorescence has led to the widespread application of a newly advanced technology called multiplex immunohistochemistry (mIHC), which enables the detection of multiple fluorescent proteins on a tumor tissue microarray (TMA) within the same temporal and spatial organization. Using this mIHC technology, we detected six tumor-associated proteins, including cluster of differentiation 4 (CD4), cluster of differentiation 8 (CD8), Pan-cytokeratin (P-CK), forkhead box P3 (FOXP3), programmed cell death 1 (PD1) as well as programmed death ligand-1 (PDL1) in cancer tissues and para-carcinomatous normal tissues from a cohort of 79 colorectal cancer patients. Results showed that, in CRC tissues, expression levels of P-CK and FOXP3 were upregulated while CD4 expression decreased significantly in comparison with adjacent normal tissues. What’s more, no significantly differential expression of CD8, PD1 or PDL1 was observed between cancer and normal tissues. FOXP3 expression was found to be correlated with tumor size (FOXP3 expression in tumor with volume >10 cm3 was significantly lower than that in tumor with volume ≤ 10 cm3), and reduced FOXP3 expression was associated with worse prognosis. P-CK expression in low-grade (Grade I-II) CRC patients was higher than that in advanced grade (Grade III-IV) patients, while association of P-CK expression with CRC prognosis was of no significance. In conclusion, FOXP3 and P-CK could be utilized as biopredictors of CRC (FOXP3 as a diagnostic and prognostic biomarker; P-CK as a diagnostic biomarker) for their differential expression patterns and clinicopathological correlation, while CD4, CD8, PD1 and PDL1 are more suitable for combined use.

Keywords

Colorectal cancer (CRC), Multiplex immunohistochemistry (mIHC), Tumor tissue microarray (TMA), Diagnosis, Prognosis, Biomarker

Introduction

Colorectal Cancer (CRC), one of the major causes of morbidity and mortality worldwide, is the second most common type of cancer in women and the third most common type of cancer in men, accounting for over 9% of all cancer incidence and causing death for more than 600,000 cases all over the world per year [1-4]. CRC is widely believed to develop in a multi-step process from Aberrant Crypt Foci (ACF), through benign and precancerous lesions (adenomas), to malignant tumors (adenocarcinomas) over an extended period of time [5]. Treatment of CRC usually comprises surgical resection of the primary tumors in patients followed by chemotherapy, radiotherapy and/or immunotherapy for advanced stages (stage III and IV) [6]. Despite advances in detection and available therapeutic strategies, the clinical outcomes for CRC remain poor due to tumor recurrence, metastasis, and resistance to radio-/chemo-therapy [7,8].

Early diagnosis of CRC is of importance for its significant impacts on cancer management, prognosis, recurrence and survival [9-11]. The 5-year survival rate could rise up to 90% in CRC patients who were diagnosed in the early stage, but unfortunately, the great majority of CRC cases had developed to an advanced stage at the time of diagnosis with a low survival rate around 8-9% [12,13]. Invasive techniques used for CRC diagnosis including endoscopic and radiological imaging suffered from poor patient compliance [14]. In addition, tumor markers such as carbohydrate antigen 19-9 (CA 19-9) and Carcinoembryonic Antigen (CEA) commonly used in clinical circumstance have the problems of unsatisfactory sensitivity and specificity, resulting in limited clinical application in CRC diagnosis, prognosis and survival [15]. Thus, the development of noninvasive and accurate screening tools for early detection and precise staging of CRC are of great importance and significance.

Conventional Immunohistochemistry (IHC) is a diagnostic technique widely used in the field of tissue pathology. However, IHC suffers from a number of limitations such as relatively high interobserver variability and limited labelling of a single marker per tissue section, resulting in missed opportunities of important diagnostic and prognostic information [16-18]. By contrast, multiplex Immunohistochemistry (mIHC), allowing simultaneous detection of multiple markers on a single tissue section, has emerged as a promising technology for its capability of provision of high throughput multiplex immunohistochemical staining and standardized quantitative analysis of highly reproducible and efficient tissue studies, as well as comprehensive study of cellular component, marker expression patterns, relative spatial distribution of multiple cell types and cell‐cell interactions, which are of benefit to diagnostic accuracy [19-21].

In light of this, we analyzed the expression levels and potential clinicopathological prognosis values of six tumor-associated proteins including cluster of differentiation 4 (CD4), cluster of differentiation 8 (CD8), Pan-cytokeratin (P-CK), forkhead box P3 (FOXP3), programmed cell death 1 (PD1) and programmed death ligand-1 (PDL1) in colorectal cancer, relying on 7-color fluorescent multiplex immunostaining of tumor tissue microarray (TMA) from a cohort of 79 cancer patients.

Materials and Methods

Information for Patients

The HColA180Su17 tumor Tissue Microarray (TMA) (Outdo, Shanghai, China) consisted of paired colorectal adenocarcinoma tissues and adjacent normal tissues derived from 79 colorectal cancer patients. These patients underwent surgery from Jun. 2006 to Apr. 2007, and the follow-up information was available from Sep. 2007 to Jul. 2015. The study was conducted under the approval of the Institutional Ethics Committee and all procedures were performed according to relevant guidelines and regulations for research. The clinicopathological characteristics of 79 cancer patients were summarized in Table 1.

Table 1: Clinicopathological characteristics of a cohort of 79 colorectal cancer patients

Clinicopathological characteristics (N=79)

Number

Proportion (%)

Gender
Male

38

48.10%

Female

41

51.90%

Age (years)

≤65

36

45.57%

>65

43

54.43%

T stage

T1

1

1.27%

T2

5

6.33%

T3

58

73.42%

T4

15

18.98%

Lymph node (N stage)
Negative (N0)

48

60.76%

Positive (N1a, b-N2a, b)

31

39.24%

Metastasis (M stage)
Negative (M0)

78

98.73%

Positive (M1a, b)

1

1.27%

TNM stage
I

5

6.33%

II A

34

43.04%

II B

6

7.59%

II C

3

3.80%

III A

0

0.00%

III B

28

35.44%

III C

3

3.80%

IV A

0

0.00%

IV B

0

0.00%

Pathological grade
I

16

20.25%

II

50

63.29%

III

12

15.19%

IV

1

1.27%

Histology
Adenocarcinoma

31

39.24%

Canalicular adenoma

41

51.90%

Mucinous adenocarcinoma

6

7.59%

Signet-ring cell carcinoma

1

1.27%

Disease status at last follow-up
Survival

42

53.16%

Death

37

46.84%

Preparation of Tissue Microarray (TMA)

Tissue Microarray (TMA) was made on basis of pathological diagnosis of each tissue. Briefly, formalin-fixed and paraffin-embedded samples were identified as well as the specimens were reviewed with hematoxylin and eosin stain by an independent surgical pathologist in order to confirm the presence of colorectal cancer and adjacent normal tissues [22]. For the formation of TMA, core cylinders (1 mm) were punched from each of circled areas and stored in a recipient paraffin block after circling of at least two representative tumor areas from each block by the pathologist. At last, consecutive TMA sections (6 mm thick) were cut and placed onto poly-L-lysinecoated slides for subsequent analysis [23].

Fluorescent mIHC of TMA

For multiplex Immunohistochemistry (mIHC) staining, antibodies for CD4, CD8, PCK, FOXP3, PD1 and PDL1were optimized by concentration and application order, meanwhile, a spectral library was built based on the single-stained slides [24]. The multiplex immunofluorescence staining and multispectral imaging of these six proteins were obtained on a TMA slide using Opal Polaris 7 Color Manual IHC Detection Kit (cat NEL861001KT, Akoya, US). In brief, the slide was deparaffinized by xylene for 10 min for three times, followed by 100% ethanol, 95% ethanol, 85% ethanol, and 75% ethanol for 5 min, respectively. After rinsing in distilled water for 3 min, slide was pretreated with 100 ml citric acid solution (pH6.0/pH9.0) for antigen retrieval with microwaving (15 min on 20% power after 45 s on 100% power) and transferred to a slide jar containing 1xTBST to mix well. Afterwards, the slide was blocked in 10% blocking solution for 10 min, stained respectively with primary antibody against CD4, CD8, P-CK, FOXP3, PD1 or PDL1 for 1 h at room temperature, washed with 1xTBST for 3 min twice and incubated with polymer HRPanti-mouse/rabbit IgG secondary antibody for 10 min at room temperature. The slide was covered by Tyramide (TSA)-conjugated fluorophore (TSA Fluorescence Kits, Panovue, Beijing, China) at 1:100 dilution and incubated for 10 min at room temperature, washed with 1xTBST for 3 min twice  for next staining procedure. Furthermore, the process was repeated by microwave heat-treating the slide for antigen retrieval for every additional marker in mIHC assay, followed by one primary antibody staining during each cycle ordered as CD4, CD8, P-CK, FOXP3, PD1 and PDL1, respectively, and then downstream procedures as mentioned above. After labelling of all these human antigens, cell nucleus were counterstained with 4′,6diamidino-2-phenylindole (DAPI) (Sigma-Aldrich, US). Detailed information about primary antibodies was summarized in Table 2.

Table 2: Primary antibodies used for mIHC staining

Antibodies

Dilution Antibody Type Catalogue#

Vender

CD4

1:200

Rabbit monoclonal ab133616

Abcam

CD8

1:100

Mouse monoclonal NBP2-34039

NOVUS

P-CK

1:100

Mouse monoclonal GM351529

Gene Tech

FOXP3

1:200

Mouse monoclonal 14-4777-83

Thermo

PD1

1:200

Mouse monoclonal GT228129

Gene Tech

PDL1

1:200

Rabbit monoclonal ab213524

Abcam

Multispectral Imaging

The stained slide was scanned using the Vectra Polaris (Akoya, US) to obtain multispectral images, which precisely captures the fluorescent spectra from 420 to 720 nm (at 20-nm wavelength intervals) with identical exposure time. Next, the scans were combined into a single stack image with high contrast and accuracy

Scoring Multispectral Images

InForm Tissue Analysis Software (Akoya, US) was used in batch analysis of experimental multispectral images [25]. Firstly, images of single-stained and unstained sections were used to respectively extract the fluorescent spectrum of each fluorescein and autofluorescence of tissues. Secondly, the extracted images were used in establishment of a spectral library for multispectral unmixing by InForm image analysis software. Finally, using this established spectral library, gain of reconstructed images of sections with removed autofluorescence was fulfilled. In order to score multispectral images, three to six representative regions of interest for imaging (200×) from each case were selected. A few representative multispectral images were then loaded into analysis software to build an algorithm for segmenting tissues and cells. Next, two tissue categories of STROMA and TUMOR were trained in accordance with intensity of DAPI signals, these detected tissue compartments were selected and quantified for each stained target proteins, and corresponding number of positive and total cells were counted as well. 4-bin (0, 1+, 2+, 3+) scoring system was used for quantification of expression levels of target proteins by calculating H-score (a score which was calculated using the percentage in each bin and ranges from 0 to 300) with cell stains. Results of H-score were shown by the positive rate of cells in each bin, including four levels (0~1, 1~2, 2~3, 3~) so as to measure and categorize protein expression levels into negative, low, medium and high levels, respectively. Generally, H-score with 0~1 and 1~2 (0, 1+) were considered as low expression level, while score with 2~3 and 3~ (2+, 3+) were considered as high expression level.

Statistical Analysis

The significance of experimental data from patient specimens was determined by the Mann-Whitney U test. The Kaplan-Meier test was used to assess overall survival (OS) rates, and survival curves were plotted by the log-rank test. *P<0.05 was considered as statistically significant, **P<0.01 and ***P<0.0001 were considered as strongly significant. Statistics software GraphPad Prism version 8 was used for all statistical analyses.

Results

Demographics

A following-up for the cohort of 79 CRC patients was performed from 2008 to 2015 for the evaluation of a seven-year survival. Among these eight clinicopathological characteristics including gender, age, tumor size, T stage, N stage, M stage, TNM stage and pathological grade, the survival was associated with three of them, namely N stage, TNM stage as well as pathological grade. The results showed that prognosis of patients with negative lymph nodes (N0), early TNM stage (TNM I-II) and low pathological grade (Grade I-II) were significantly better than those with positive lymph nodes (N1-2), late TNM stage (TNM 3-4) and advanced pathological grade (Grade III-IV) (P<0.05, Figure 1 and Table 3).

fig 1

Figure 1: Overall survival (OS) rates of clinicopathological characteristics analyzed by Kaplan-Meier test. A. Lymph Node (N Stage), B. TNM Stage, C. Pathological Grade as clinical prognostic factors in cancer tissues in a cohort of 79 CRC patients. Orange dotted line: Overall survival (OS) rates as 50%.

Table 3: Prognostic clinicopathological characteristics of a cohort of 79 colorecta cancer patients

Clinicopathological characteristics

HR (95%CI)

P Value

Gender (male vs. female)

0.806 (0.410-1.584)

0.532

Age (yeas≤65 vs. yeas>65)

0.755 (0.384-1.484)

0.415

Tumor size (V≤10 cm3 vs. V>10 cm3)

0.783 (0.327-1.876)

0.584

T stage (T1-3 vs. T4)

0.812 (0.333-1.978)

0.646

N stage (Negative vs. Positive)

0.3709 (0.179-0.768)

<0.01

M stage (Negative vs. Positive)

2.787 (0.179-43.36)

0.464

TNM (TNM I-II vs. TNM III-IV)

0.413 (0.201-0.852)

<0.01

Pathological grade (I-II vs. III-IV)

0.209 (0.071-0.611)

<0.01

Fluorescent mIHC Profile on TMA Slides Derived from Colorectal Cancer Patients

In order to obtain multiple fluorescent images, the TMA slides were trained according to intensity of DAPI signals before the selection of detected tissue compartments for each stained target proteins on slides. All six antibodies of CD4, CD8, P-CK, FOXP3, PD1 and PDL1 were then performed ahead of the quantification of protein expression level by scoring system to calculate H-score based on cell fluorescence. In detected tissue compartments and cells, images of monochromatic proteins were shown in the upper row ordered as DAPI, CD4, CD8, P-CK, FOXP3, PD1 as well as PDL1 (Figure 2). In addition, merged images of the multispectral fluorescence of these target proteins and DAPI were displayed at the bottom of the figure. The selected images displayed tumor (Figure 2A) and adjacent normal (Figure 2B) tissues, respectively.

fig 2

Figure 2: Mono- and multi-chromatic mIHC profile of colorectal cancer and adjacent normal tissues. A, B. Representative images of monochromatic and multispectral fluorescence in tissues from colorectal cancer and adjacent normal areas. Small images in the upper row displayed selected tissue compartments stained by DAPI, CD4, CD8, P-CK, FOXP3, PD1 and PDL1. Large images at the bottom showed a merged multispectral fluorescence from DAPI, CD4, CD8, P-CK, FOXP3, PD1 and PDL1.

Determination of Significant Markers by Fluorescent mIHC in Colorectal Cancer Patients

In a cohort of 79 colorectal patients, comparison of the expression levels of CD4, CD8, P-CK, FOXP3, PD1 and PDL1 were performed between tumor and paracarcinomatous normal tissues for the exploration of cancer associated potential biomarker. As shown in Figure 3 for monochromatic proteins, expressions of P-CK and FOXP3 were upregulated while CD4 expression decreased significantly in cancer tissues compared with adjacent normal tissues (Figure 3A, P<0.05; Figure 3C, P<0.001; Figure 3D, P<0.01). As shown in Figure 4 for bi- as well as multi-chromatic combinations, except that the expression levels of bichromatic CD4/P-CK, CD8/P-CK and P-CK/FOXP3, trichromatic CD4/CD8/P-CK, multichromatic CD4/CD8/P-CK/FOXP3 and CD4/CD8/P-CK/FOXP3/PD1/PDL1 were of significant differences (Figure 4A to 4F, P<0.001), differential expressions were not observed in other combinations in cancer tissues. As to the compared expression levels of single, double or multiple stained combinations of these six target proteins, all data were analyzed by Mann-Whitney U test and the P values were shown in Table 4.

fig 3

Figure 3: Comparing expression levels of monochromatic target proteins based on H-scores by mIHC from tumor versus normal tissues in a cohort of 79 colorectal cancer patients. A to F. Differential expression patterns of single stained proteins including CD4 (3A. P<0.05), CD8 (3B, P=0.915), P-CK (3C, P<0.001), FOXP3 (3D, P<0.01), PD1 (3E, ns. P=0.231) and PDL1 (3F, P=0.511).

fig 4

Figure 4: Comparing expression levels of di- and multi-chromatic target proteins based on H-scores by mIHC from tumor versus normal tissues in a cohort of 79 colorectal cancer patients. A to F. Comparing expression patterns of combination of double and multiple stained proteins including CD4/P-CK (3A. P<0.001), CD8/PC-K (3B, P<0.001), P-CK/FOXP3 (3C, P<0.05), CD4/CD8/P-CK (3D, P<0.001), CD4/CD8/P-CK/FOXP3 (3E, P<0.001), and CD4/CD8/P-CK/FOXP3/PD1/PDL1 (3F, P<0.001).

Table 4: Differential expression of mIHC markers in cancer vs. normal tissues

mIHC target proteins

Cancer vs. Normal (N=79)

CD4

P < 0.05

CD8

P=0.915

P-CK

P < 0.001

FOXP3

P < 0.01

PD1

P=0.231

PDL1

P=0.511

CD4/CD8

P=0.340

CD4/P-CK

P < 0.001

CD8/P-CK

P < 0.001

CD4/FOXP3

P=0.058

CD8/FOXP3

P=0.781

P-CK/FOXP3

P < 0.001

PD1/PDL1

P=0.859

CD4/CD8/P-CK

P < 0.001

CD4/CD8/FOXP3

P=0.402

FOXP3/PD1/PDL1

P=0.975

CD4/CD8/P-CK/FOXP3

P < 0.001

CD4/CD8/PD1/PDL1

P=0.392

CD4/CD8/FOXP3/PD1/PDL1

P=0.481

CD4/CD8/P-CK/FOXP3/PD1/PDL1

P < 0.001

Correlation between Six Proteins and Clinicopathological Characteristics

Statistic analyses were performed by Mann-Whitney U test to explore the correlation between six proteins (CD4, CD8, P-CK, FOXP3, PD1 and PDL1) and eight cancer related clinicopathological factors (gender, age, tumor size, T stage, lymph node, metastasis, TNM stage, pathological grade). FOXP3 and P-CK were found to be correlated with tumor size and pathological grade, respectively , even though most of the correlations were of no significance (Table 5). Among which, expression of FOXP3 in tumor with volume>10 cm3 (N=65) was significantly lower than that in tumor with volume≤10 cm3 (N=14) (Figure 5A, P<0.01), and expression of P-CK in low pathological grade (Grade I-II) (N=66) was higher than that in advanced grade (Grade III-IV) (N=13) (Figure 5B, P<0.05).

Table 5: Correlation between mIHC target proteins and clinicopathological characteristics

Clinicopathological characteristics

P value

CD4

CD8 P-CK FOXP3 PD1

PDL1

Gender (male vs. female)

0.901

0.232 0.540 0.351 0.375

0.656

Age (yeas ≤65 vs. yeas >65)

0.805

0.419 0.503 0.491 0.768

0.730

Tumor size (V≤10 cm3 vs. V>10 cm3)

0.843

0.673 0.817 <0.01 0.695

0.912

T stage (T1-3 vs. T4)

0.587

0.780 0.995 0.527 0.350

0.231

N stage (negative vs. positive)

0.778

0.432 0.877 0.253 0.784

0.194

M stage (negative vs. positive)

TNM (TNM I-II vs. TNM III-IV)

0.775

0.424 0.866 0.533 0.835

0.939

Pathological grade (I-II vs. III-IV)

0.417

0.423 <0.05 0.924 0.526

0.537

fig 5

Figure 5: Significant correlations between two target proteins and clinicopathological characteristics in colorectal cancer tissues. A. The expression level of FOXP3 significantly declined in larger tumor (V>10 cm3) in comparison with smaller tumor (V≤10 cm3). B. The expression level of P-CK in pathological grade I and II was significantly higher than that in advanced grade III and IV. Data on the graph was displayed as mean ± SD (*P<0.05, **P<0.01).

Association of Prognosis Markers with Clinical Outcomes

For purpose of prognosis potential of these six proteins in CRC, expression of each protein was divided into low-expressed group (H-score 0 to 1+) and high-expressed group (H-score 2+ to 3+) on the basis of H-score representation calculated by the fluorescence intensity from three to six representative regions of each sample. Association of low-/high-level protein expression with the seven-year overall survival (OS) status of 79 CRC patients were analyzed by Kaplan-Meier test. Compared with CD4 (Figure 6A, P=0.122), CD8 (Figure 6B, P=0.905), P-CK (Figure 6C, P=0.406), PD1 (Figure 6E, P=0.582) and PDL1 (Figure 6F, P=0.156), FOXP3 was the only one with statistically significant association with CRC prognosis (Figure 6D, P<0.05). CRC patients with high-level FOXP3 expression (N=6) seemed to have a longer OS time than those with low-level expression (N=73) (Figure 6D, P<0.05), which supported the tumor-growth potential of low-expressed FOXP3 observed in our study (Figure 5A, P<0.01).

fig 6

Figure 6: Overall survival (OS) rates with differential expression levels of CD4 and FOXP3 analyzed by Kaplan-Meier test. The low- and high-expression of A. CD4 and B. FOXP3 were associated with a status of seven-year survival in cancer tissues in a cohort of 79 CRC patients. Orange dotted line: half of OS rates as 50%.

Discussion

In this study, we performed multiplex immunohistochemistry analysis on six target proteins to explore the correlation of these molecules with colorectal cancer. FOXP3 (forkhead box P3, also named IPEX, PIDX) is a member of the forkhead box (FOX) family of transcription factors consisting of an evolutionarily conserved group of transcriptional regulators whose dysfunction has been associated with human malignant neoplasias [26-35]. FOXP3 is mainly expressed in regulatory T (Treg) cells, and it has also been found in other cells such as B lymphocytes [36-42]. FOXP3 was described as an important molecular actor involved in the development and function of Treg cells playing essential roles in the regulation of autoimmunity, infection and tumor environment [36-38]. FOXP3 is considered as a molecule at the crossroads of tumorigenesis and immunity for its bilateral role of cancer promotor or suppressor [42-46]. Furthermore, the role of FOXP3 in the biogenesis, development as well as clinical prognosis of colorectal cancer is still not completely understood, thereinto, infiltration of FOXP3+ Treg cells indicated favorable prognosis in some but not all studies [47-57]. Our results showed that the expression level of FOXP3 was not only significantly upregulated in tumor tissues (Figure 3D, P<0.01), but also associated with tumor size (Table 5, P<0.01). Additionally, FOXP3 expression in CRC patients with tumor volume>10 cm3 was significantly lower than that in patients with tumor volume≤10 cm3 (Figure 5A, P<0.01), indicating a tumor growth potential of FOXP3 with low expression in CRC, consistently, reduced expression of FOXP3 was associated with worse prognosis (Figure 6D, P<0.05). All these results showed that FOXP3 could be applied as a potential biomarker for CRC diagnosis and prognosis.

CD4, a membrane glycoprotein of T lymphocytes, is expressed not only in T lymphocytes, but also in B cells, macrophages as well as granulocytes. CD4 acts as a coreceptor with the T-cell receptor on T lymphocytes in recognition of antigens displayed by antigen-presenting cells in the context of class II major histocompatibility complex (MHC) molecules, and functions to initiate or augment the early phase of T-cell activation. Similarly, CD8, a cell surface glycoprotein found on most cytotoxic T lymphocytes mediating immune cell-cell interactions, acts as a coreceptor with the T-cell receptor on T lymphocytes to recognize antigens displayed by antigen-presenting cells in the context of class I MHC molecules. In general, CD4+ and CD8+ T cells identify antigens related to cancer cells and play significant roles in cancer immunology and immunotherapy [58,59]. Additionally, various studies have demonstrated that CD4+ and CD8+ T cells may also control tumor growth [60,61]. Increased levels of CD4+ and CD8+ T cells in colorectal tumor microenvironment were shown to correlate with improved response to chemoradiotherapy [62]. Results of prevenient studies suggested that levels of tumor infiltration by CD4+ and CD8+ T cells may be good predictive factors for patient clinical prognosis of various tumors including colorectal cancer, melanoma, oesophageal squamous cell carcinoma, ovarian cancer, pancreatic cancer and renal cancer [63-71]. Differently, compared with adjacent normal tissues, the expression levels of CD4 and CD8 in colorectal cancer tissues here decreased significantly and had no difference, respectively (Figure 3A and 3B), suggesting reduced immune infiltration of CD4+ and CD8+ T cells. What’s more, association of differential expression levels of CD4 or CD8 with clinical outcomes of CRC patients was of no significance (Figure 6A and 6B), and they had no significant association with tumor size or other clinicopathological characteristics (Table 5). Malignant tumors like CRC can cause the functional loss of antigen recognition, cell proliferation and activation of effector T cells, which is known as T cell exhaustion accompanied by the activation of multiple inhibitory receptors such as CTLA4 and PD1/PDL1 [72-74]. The decreased infiltration and effects on prognosis of CD4+ and CD8+ T cells observed here may be related to T cell exhaustion. Generally, the biological behaviors of cancers are influenced by the functional status of tumor-infiltrating immune cells whose roles in response to cancer are component- and stage-dependent. The CD4+ T cells consist of multiple morphologically and functionally distinctive subpopulations such as T regulatory (Treg) cells, T helper 1 (Th1), Th2, Th9, Th17 and follicular helper T (Tfh) cells, whose roles related to proinflammation and/or antiinflammation, activation and infiltration of CD8+ T cells in tumor microenvironment display tumor-promoting or tumor-suppressing effects in a case-dependent manner [75-87]. The limited tumor infiltration and functional potential of CD8+ T cells over served in this study might be partially due to a lack of CD4+ Th cell-mediated function.

PD1 (programmed cell death 1, also named PDCD1, CD279), an immunoinhibitory receptor belonging to the CD28 family that is expressed on activated T cells, is involved in T cell proliferation and functional regulation including those of effector CD8+ T cells, and also able to promote the differentiation of CD4+ T cells into T regulatory cells [88-90]. PD1 is expressed in many types of tumors and has demonstrated to play roles in anti-tumor immunity, safeguarding against autoimmunity as well as the inhibition of effective anti-tumor and anti-microbial immunity. Furthermore, PD1 interacts with ligand PDL1 (also named B7-H1, CD274) to form the PD1/PDL1 axis, an immune checkpoint which is usually up-regulated to help tumor cells avoid immune destruction in an immunosuppressive tumor microenvironment [91]. Overexpressed PDL1 can protect tumor cells by inhibiting the activity of PD1 expressing adjoining tumor-infiltrating effector CD4+/CD8+ T cells [92]. PDL1 is mainly expressed on the surface of antigen-representing and tumor cells in various types of cancer such as carcinomas of the adrenal cortex, bladder, brain, breast, colorectum, esophagus, gastrointestinal tract, kidney, liver, lung, ovary, pancreas, thymus, thyroid and urothelium [93]. Contradictory correlations of expression of PD1 and/or PDL1 with prognosis results and clinicopathological characteristics of colorectal cancer were observed (some investigations showed that overexpression of PD1 and/or PDL1 forecasted better prognosis, while others presented opposite results), even if PD1 and PDL1 may play oncogenic roles in colon cancer carcinogenesis [94-98]. Again, in our study, no significant difference was observed between expression of either PD1 or PDL1 in colorectal cancer tissues and those in normal tissues (Figure 3E and 3F), they also had no significant association with pathological grade or other characteristics (Table 5). Differential expression of PD1 or PDL1 was not statistically associated with CRC prognosis (Figure 6E and 6F). All these results indicated that PD1 and PDL1, in comparison to using alone, are more suitable for combination with other proteins for the application as potential biopredictors of CRC diagnosis.

References

  1. McGuire S Geneva (2016) Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Advances in Nutrition An International Review Journal 7: 418-419. [crossref]
  2. Jahani-Sherafat S, Alebouyeh M, Moghim S, et al. (2018) Role of gut microbiota in the pathogenesis of colorectal cancer; a review article. Gastroenterology and Hepatology from Bed to Bench 11: 101-109. [crossref]
  3. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, et al. (2015) Global cancer statistics, 2012. CA: A Cancer Journal for Clinicians 65: 87-108. [crossref]
  4. Favoriti P, Carbone G, Greco M, et al. (2016) Worldwide burden of colorectal cancer: a review. Updates in Surgery 68: 7-11. [crossref]
  5. Fearon ER, Vogelstein B (1990) A genetic model for colorectal tumorigenesis. Cell 61: 759-767. [crossref]
  6. Graham JS, Cassidy J (2012) Adjuvant therapy in colon cancer. Expert Review of Anticancer Therapy 12: 99-109. [crossref]
  7. Bandar MA, Kim N (2017) Current status and future perspectives on treatment of liver metastasis in colorectal cancer (Review). Oncology Reports 37: 2553-2564. [crossref]
  8. Soon-Chan K, Young-Kyoung S, Ye-Ah K, et al. (2018) Identification of genes inducing resistance to ionizing radiation in human rectal cancer cell lines: re-sensitization of radio-resistant rectal cancer cells through down regulating NDRG1. BMC Cancer 18: 594. [crossref]
  9. Duffy MJ, Dalen AV, Haglund C, et al. (2007) Tumour markers in colorectal cancer: European Group on Tumour Markers (EGTM) guidelines for clinical use. European Journal of Cancer 43: 1348-1360. [crossref]
  10. Duffy MJ, Dalen AV, Haglund C, et al. (2003) Clinical utility of biochemical markers in colorectal cancer: European Group on Tumour Markers (EGTM) guidelines. European Journal of Cancer 39: 718-727. [crossref]
  11. Zhou Y, Abel G A, Hamilton W, et al. (2017) Diagnosis of cancer as an emergency: a critical review of current evidence. Nature Reviews Clinical Oncology 14: 45-56. [crossref]
  12. Meyerhardt JA, Mayer RJ (2005) Systemic therapy for colorectal cancer. New England Journal of Medicine 352: 476-487. [crossref]
  13. Mazzanti R, Solazzo M, Ornella Fantappié, et al. (2006) Differential expression proteomics of human colon cancer. American Journal of Physiology Gastrointestinal & Liver Physiology 290: G1329.
  14. Wang H, Tso VK, Slupsky CM, Fedorak RN (2010) Metabolomics and detection of colorectal cancer in humans: a systematic review. Future Oncology 6: 1395-1406. [crossref]
  15. Ni Y, Xie G, Jia W (2014) Metabonomics of human colorectal cancer: new approaches for early diagnosis and biomarker discovery. Journal of Proteome Research 13: 3857-3870. [crossref]
  16. Polley M, Leung S, Mcshane LM, Gao D, Hugh JC, et al. (2013) An International Ki67 Reproducibility Study. Journal of the National Cancer Institute 105: 1897-1906. [crossref]
  17. Varga Z, Diebold J, Dommann-Scherrer C, Frick H, Kaup D, et al. (2015) How Reliable Is Ki-67 Immunohistochemistry in Grade 2 Breast Carcinomas? A QA Study of the Swiss Working Group of Breast- and Gynecopathologists. Plos One 7: e37379. [crossref]
  18. Cheng CL, Thike AA, Tan S, Chua PJ, Bay BH, et al. (2015) Expression of FGFR1 is an independent prognostic factor in triple-negative breast cancer. Breast Cancer Research & Treatment 151: 99-111. [crossref]
  19. Stack Edward C, Wang C, Roman KA, Hoyt CC (2014) Multiplexed immunohistochemistry, imaging, and quantitation: A review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods: A Companion to Methods in Enzymology 70: 46-58.
  20. Abel EJ, Bauman TM, Weiker M, Shi F, Downs TM, et al. (2014) Analysis and validation of tissue biomarkers for renal cell carcinoma using automated high-throughput evaluation of protein expression. Human Pathology 45: 1092-1099. [crossref]
  21. Lim JCT, Yeong JPS, Lim CJ, Ong CCH, Wong SC, et al. (2018) An automated staining protocol for 7colour immunofluorescence of human tissue sections for diagnostic and prognostic use. Pathology 50: 333. [crossref]
  22. Ring Kari L, Frumovitz MM, Yemelyanova AV, Soliman PT, Jazaeri AA (2017) Potential immunotherapy targets in recurrent cervical cancer. Gynecologic Oncology: An International Journal 145: 462-468. [crossref]
  23. F Min, Li Y, Kai H, Qi S, Zhang J, et al. (2017) IL33 Promotes Colon Cancer Cell Stemness via JNK Activation and Macrophage Recruitment. Cancer Research 77: 2735. [crossref]
  24. Ying L, Yan F, Meng Q, Yu L, Yuan X, et al. (2018) PD-L1 expression is a prognostic factor in subgroups of gastric cancer patients stratified according to their levels ofCD8 and FOXP3 immune markers. Oncoimmunology 7: e1433520. [crossref]
  25. Yang L, Liu Z, Tan J, Dong H, Zhang X (2018) Multispectral imaging reveals hyper active TGF-β signaling in colorectal cancer. Cancer biology & therapy 19: 1-8. [crossref]
  26. Darnell James E (2002) Transcription factors as targets for cancer therapy. Nature Reviews Cancer 2: 740-749. [crossref]
  27. Tong IL, Young RA (2013) Transcriptional Regulation and Its Misregulation in Disease. Cell 152: 1237-1251. [crossref]
  28. Bhagwat AS, Vakoc CR (2015) Targeting Transcription Factors in Cancer. Trends in Cancer 1: 53-65.
  29. Myatt SS, Lam WF (2007) The emerging roles of forkhead box (Fox) proteins in cancer. Nature Reviews Cancer 7: 847-859. [crossref]
  30. Hannenhalli S, Kaestner KH (2009) The evolution of Fox genes and their role in development and disease. Nature Reviews Genetics 10: 233-240. [crossref]
  31. Benayoun BA, Caburet S, Veitia RA (2011) Forkhead transcription factors: key players in health and disease. Trends in Genetics 27: 224-232. [crossref]
  32. Lam EF, Brosens JJ, Gomes AR, Koo CY (2013) Forkhead box proteins: tuning forks for transcriptional harmony. Nature Reviews Cancer 13: 482-495. [crossref]
  33. Katoh M, Igarashi M, Fukuda H, Nakagama H, Katoh M (2013) Cancer genetics and genomics of human FOX family genes. Cancer Letters 328: 198-206. [crossref]
  34. Lam EW, Gomes AR (2014) Forkhead box transcription factors in cancer initiation, progression and chemotherapeutic drug response. Frontiers Research Topic. Molecular & Cellular Oncology 4: 305. [crossref]
  35. Golson ML, Kaestner KH (2016) Fox transcription factors: from development to disease. Development 143: 4558-4570. [crossref]
  36. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunology 4: 330336. [crossref]
  37. Khattri R, Cox T, Yasayko SA, Ramsdell F (2003) An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nature Immunology 4: 337-342. [crossref]
  38. Campbell DJ, Ziegler SF (2007) FOXP3 modifies the phenotypic and functional properties of regulatory T cells. Nature Reviews Immunology 7: 305. [crossref]
  39. Triulzi T, Tagliabue E, Balsari A, Casalini P (2013) FOXP3 expression in tumor cells and implications for cancer progression. Journal of Cellular Physiology 228: 30-35. [crossref]
  40. Mercer F, Unutmaz D (2009) The Biology of FoxP3: A Key Player in Immune Suppression during Infections, Autoimmune Diseases and Cancer. Oxygen Transport to Tissue 665: 47-59. [crossref]
  41. Martin F, Ladoire S, Mignot G, Apetoh L, Ghiringhelli F (2010) Human FOXP3 and cancer. Oncogene 29: 4121-4129. [crossref]
  42. Redpath M, Xu B, Kempen L, Spatz A (2011) The dual role of the X-linked FoxP3 gene in human cancers. Molecular Oncology 5: 156-163. [crossref]
  43. Tao Z, Wang L, Morrison C, Chang X, Zhang H, et al. (2007) FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ErbB2 oncogene. Cell 129: 1275-1286. [crossref]
  44. Katoh H, Zheng P, Liu Y (2010) Signalling through FOXP3 as an X-linked tumor suppressor. International Journal of Biochemistry & Cell Biology 42: 1784-1787. [crossref]
  45. Wang L, Liu R, Ribick M, et al. (2010) FOXP3 as an X-linked tumor suppressor. Discovery medicine 10: 322-328.
  46. Redpath M, Xu B, Kempen LC, Spatz A (2011) The dual role of the X-linked FoxP3 gene in human cancers. Molecular Oncology 5: 156-163. [crossref]
  47. Xu P, Wei F, Zheng Z, Wang J, Wang P, et al. (2017) The Clinicopathological and Prognostic Implications of FoxP3+ Regulatory T Cells in Patients with Colorectal Cancer: A Meta-Analysis. Frontiers in Physiology 8: 950. [crossref]
  48. Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, et al. (2009) Intraepithelial Effector (CD3+)/Regulatory (FoxP3+) T-Cell Ratio Predicts a Clinical Outcome of Human Colon Carcinoma. Gastroenterology 137: 1270-1279. [crossref]
  49. Fridman WH, Pagès F, Sautès-Fridman C, Galon J (2012) The immune contexture in human tumours: impact on clinical outcome. Nature Reviews Cancer 12: 298-306. [crossref]
  50. Suzuki, H, Onishi, Morisaki T, Tanaka M, Katano M (2013) Intratumoral FOXP3+VEGFR2+ regulatory T cells are predictive markers for recurrence and survival in patients with colorectal cancer. Clinical Immunology San Diego Academic Press 146: 26-33. [crossref]
  51. Yong H, Huaiwei L, Yong Z, Yuan R, Wang F, et al. (2014) Prognostic Value of Tumor-Infiltrating FoxP3+ T Cells in Gastrointestinal Cancers: A Meta Analysis. Plos One 9: e94376. [crossref]
  52. Shang B, Liu Y, Jiang SJ, Liu Y (2015) Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Scientific Reports 5: 15179. [crossref]
  53. Yoon HH, Orrock JM, Foster NR, Sargent DJ, Smyrk TC, et al. (2012) Prognostic Impact of FoxP3+ Regulatory T Cells in Relation to CD8+ T Lymphocyte Density in Human Colon Carcinomas. PLoS ONE 7: e42274. [crossref]
  54. Salama P, Phillips M, Grieu F, Morris M, Zeps N, et al. (2009) Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. Journal of Clinical Oncology 27: 186-192. [crossref]
  55. Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, et al. (2009) Intraepithelial Effector (CD3+)/Regulatory (FoxP3+) T-Cell Ratio Predicts a Clinical Outcome of Human Colon Carcinoma. Gastroenterology 137: 1270-1279. [crossref]
  56. Deleeuw RJ, Kost SE, Kakal JA, Nelson BH (2012) The Prognostic Value of FoxP3+ TumorInfiltrating Lymphocytes in Cancer: A Critical Review of the Literature. Clinical Cancer Research 18: 3022-3029. [crossref]
  57. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, et al. (2016) Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nature Medicine 22: 679-684. [crossref]
  58. Jannie B, Tomasz A, Nikolina B, Melief CJM, Kastenmüller W (2018) CD4+ T cell help in cancer immunology and immunotherapy. Nature Reviews Immunology 18: 635-647. [crossref]
  59. Bagher Farhood, Masoud N, Mortezaee K (2019) CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. Journal of cellular physiology 234: 8509-8521. [crossref]
  60. Flynn S, Stockinger B (2003) Tumor and CD4 T-cell interactions: tumor escape as result of reciprocal inactivation. Blood 101: 4472-4478. [crossref]
  61. Boer D Th A, van Mierlo GJ, Fransen MF, Melief CJ, Offringa R, et al. (2005) CD4+ T Cells Are Able to Promote Tumor Growth through Inhibition of Tumor-Specific CD8+ T-Cell Responses in Tumor-Bearing Hosts. Cancer Research 65: 6984. [crossref]
  62. Yasuda K, Nirei T, Sunami E, Nagawa H, Kitayama J (2011) Density of CD4(+) and CD8(+) T lymphocytes in biopsy samples can be a predictor of pathological response to chemoradiotherapy (CRT) for rectal cancer. Radiation Oncology 6: 49. [crossref]
  63. Olino K, Park T, Ahuja N (2020) Exposing Hidden Targets: Combining epigenetic and immunotherapy to overcome cancer resistance – Science Direct. Seminars in Cancer Biology 65: 114-122.
  64. Mlecnik B, Bindea G, Angell HK, Maby P, Angelova M, et al. (2016) Integrative Analyses of Colorectal Cancer Show Immunoscore Is a Stronger Predictor of Patient Survival Than Microsatellite Instability. Immunity 44: 698-711. [crossref]
  65. Fukunaga A, Miyamoto M, Cho Y, Murakami S, Kawarada Y, et al. (2004) CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 28: 26-31. [crossref]
  66. Cho Y, Miyamoto M, Kato K, Fukunaga A, Shichinohe T, et al. (2003) CD4+ and CD8+ T cells cooperate to improve prognosis of patients with esophageal squamous cell carcinoma. Cancer Research 63: 1555-1559. [crossref]
  67. Zhang L, Conejogarcia J R, Katsaros D, Gimotty PA, Massobrio M, et al. (2003) Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348: 203-213. [crossref]
  68. Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, et al. (2001) Proliferative activity of intratumoral CD8 (+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res 61: 5132-5136. [crossref]
  69. Bromwich EJ, Mcardle PA, Canna K, McMillan DC, McNicol AM, et al. (2003) The relationship between Tlymphocyte infiltration, stage, tumour grade and survival in patients undergoing curative surgery for renal cell cancer. British Journal of Cancer 89: 1906-1908. [crossref]
  70. Giraldo NA, Sanchez-Salas R, Peske JD, Vano Y, Becht E, et al. (2018) The clinical role of the TME in solid cancer. British Journal of Cancer 120: 45-53. [crossref]
  71. Wang B, Li F, Guo L, et al. (2020) Loss of survival advantage for deficient mismatch repair in patients with advanced colorectal cancer may be caused by changes in prognostic value of CD8+T cell expression. World J Surg Oncol 18: 196. [crossref]
  72. Wherry E J, Kurachi M (2015) Molecular and cellular insights into T cell exhaustion. Nature Reviews Immunology 15: 486-499. [crossref]
  73. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, et al. (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nature Immunology 10: 29-37. [crossref]
  74. Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, et al. (2016) The epigenetic landscape of T cell exhaustion. Science 354: 1165-1169. [crossref]
  75. Grivennikov SI, Greten FR, Karin M (2010) Immunity, Inflammation, and Cancer. Cell 140: 883-899. [crossref]
  76. Baitsch L, Baumgaertner P, Estelle Devêvre, Raghav SK, Legat A, et al. (2011) Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. Journal of Clinical Investigation 3: 23-25. [crossref]
  77. Zarour HM (2016) Reversing T-cell Dysfunction and Exhaustion in Cancer. Clinical Cancer Research 22: 1856-1864. [crossref]
  78. Bos R, Sherman LA (2010) CD4+ T-Cell Help in the Tumor Milieu Is Required for Recruitment and Cytolytic Function of CD8+ T Lymphocytes. Cancer Research 70: 8368. [crossref]
  79. Ruffell B, De Nardo DG, Affara NI, Coussens LM (2010) Lymphocytes in cancer development: Polarization towards pro-tumor immunity. Cytokine & Growth Factor Reviews 21: 3-10. [crossref]
  80. Zanetti Maurizio (2015) Tapping CD4 T Cells for Cancer Immunotherapy: The Choice of Personalized Genomics. The Journal of Immunology: Official Journal of the American Association of Immunologists 194: 2049-2056. [crossref]
  81. Vegran F, Apetoh L, Ghiringhelli F (2015) Th9 Cells: A Novel CD4 T-cell Subset in the Immune War against Cancer. Cancer Research 75: 475-479. [crossref]
  82. Vegran F, Berger H, Boidot R, Mignot G, Bruchard M, et al. (2014) The transcription factor IRF1 dictates the IL21-dependent anticancer functions of [T.sub.H] 9 cells. Nature Immunology 15: 758-766. [crossref]
  83. Bailey SR, Nelson MH, Himes RA, Li Z, Mehrotra S, et al. (2014) Th17 cells in cancer: the ultimate identity crisis. Frontiers in Immunology 5: 276. [crossref]
  84. Sara O, Pizarro TT (2015) The Treg/Th17 Axis: A Dynamic Balance Regulated by the Gut Microbiome. Frontiers in Immunology 6: 639. [crossref]
  85. El-Omar E M, Rabkin C S, MD Gammon, et al. (2003) Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology 124: 1193-1201.
  86. Grivennikov Sergei I (2013) Inflammation and colorectal cancer: colitis-associated neoplasia. Seminars in Immunopathology 35: 229-244. [crossref]
  87. Huang J, Shen F, Huang H, Ling C, Zhang G (2017) Th1 high in tumor microenvironment is an indicator of poor prognosis for patients with NSCLC. Oncotarget 8: 13116-13125. [crossref]
  88. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, et al. (2000) Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation. The Journal of Experimental Medicine 192: 1027-1034. [crossref]
  89. Nishimura H, Nose M, Hiai H, et al. (1999) Development of Lupus-like Autoimmune Diseases by Disruption of the PD-1 Gene Encoding an ITIM Motif-Carrying Immunoreceptor. Immunity 11: 141-151. [crossref]
  90. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, et al. (2001) Autoimmune Dilated Cardiomyopathy in PD-1 Receptor-Deficient Mice. Science 291: 319-322. [crossref]
  91. Fusi A, Festino L, Botti G, Masucci G, Melero I, et al. (2015) PD-L1 expression as a potential predictive biomarker. Lancet Oncology 16: 1285-1287. [crossref]
  92. Dong Haidong, Strome SE, Salomao DR, Tamura H, Hirano F, et al. (2002) Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nature Medicine 8: 793-800. [crossref]
  93. Yu J, Wang X, Teng F, Kong L (2016) PD-L1 expression in human cancers and its association with clinical outcomes. Oncotargets & Therapy 9: 5023-5039. [crossref]
  94. Shi SJ, Wang LJ, Wang GD, Guo ZY, Wei M, et al. (2013) B7-H1 Expression Is Associated with Poor Prognosis in Colorectal Carcinoma and Regulates the Proliferation and Invasion of HCT116 Colorectal Cancer Cells. Plos One 8: e76012. [crossref]
  95. Angelica C M, Cristina I, David V, Wang X, Peltier HJ, et al. (2016) PDL1 Regulation by p53 via miR-34. Journal of the National Cancer Institute 108: 303. [crossref]
  96. Yang L, Xue R, Pan C (2019) Prognostic and clinicopathological value of PD-L1 in colorectal cancer: a systematic review and meta-analysis. OncoTargets and therapy 12: 3671-3682. [crossref]
  97. Masugi Y, Nishihara R, Yang J, Mima K, da Silva A, et al. (2016) Tumour CD274 (PD-L1) expression and T cells in colorectal cancer. Gut 1463. [crossref]
  98. Li Y, Liang L, Dai W, Cai G, Xu Y, et al. (2016) Prognostic impact of programed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor infiltrating lymphocytes in colorectal cancer. Molecular Cancer 15: 55. [crossref]

Implications of the Strong Black Woman Stereotype for Maternal and Perinatal Health: A Short Note

DOI: 10.31038/IGOJ.2022521

Short Commentary

Operationalizing diverse forms of racism is essential to dismantling inequities in maternal and perinatal health and is a necessary step toward reproductive health justice for Black women in the United States (U.S.). Despite the well-known negative association between racism and health outcomes among U.S. minority racial groups [1,2], scant research exists examining the associations between internalized racism and stress and their impact on maternal mental health and birth outcomes [3]. This limitation is problematic. Among non-Hispanic Black Americans living in the U.S., exposure to racism significantly correlates to poor mental health, including psychological stress, anxiety, and depression, which have a positive relationship to poor birth outcomes among Black women [4].

Research has found that the prevalence of Low Birthweight (LBW) babies among African American populations is approximately two times higher (13.9%) than in White, non-Hispanic populations (7.0%) [5]. While Preterm Births (PTB) were found to have generally declined in 2020, this rate continues to be much higher among Black non-Hispanic women than in White women at 14.39% and 9.10%, respectively [5]. The health implications for LBW and PTB infants are substantive and can lead to a life course of poor health. Adding to this concern, the infant mortality rate among Black births was 10.6 per 1,000 deaths, which is nearly 2.5 times higher than that of White infants (4.5 times per 1,000 deaths) [6].

Structural racism remains a constant threat to Black women’s reproductive health. Manifesting in personally-mediated discrimination and inequitable policies, racism is often based on historical and sociocultural tropes or stereotypes, which characterize Black Americans as inadequate and inferior. One response of stigmatized racial populations to pervasive negative racial stereotypes is to internalize this racism with significant repercussions to maternal and perinatal health.

Internalized racism is the unconscious appropriation of the dominant White culture’s actions, beliefs, and stereotypes about racialized peoples [3]. Not to be mistaken for individual pathology, it takes shape through frequent and enduring exposure to multiple layers of racial oppression in the U.S. [7,8]. One mechanism by which internalized racism can cause mental and physical harm is through an understudied specific internalized representation of racism, the Strong Black Woman (SBW). Intergenerationally, Black women perceive the Strong Black Woman as a natural and normal aspect of identity as it characterizes Black women’s pride, persistence, and imperviousness to everyday occurrences of racism, allowing for their survival and that of their families and communities within an adversarial social context [9]. As such, the SBW presents with certain normative behaviors, such as enduring strength, the suppression of emotions, resistance to vulnerability or dependence, persistence to succeed despite limited resources, and a responsibility to help others. While the SBW has been touted as a coping mechanism encouraging self-efficacy and perseverance, this caricature of Black women’s strength is rooted in attitudes and beliefs that justified their enslavement during chattel slavery in the U.S. to maintain White power and privilege [10].

Complicit with racist ideology, the SBW schema harms self-image with far-reaching implications for Black mothers [11-13]. The SBW is a norm to which Black women’s behavior is compared and modulated, leading to maladaptive perfectionism, affect, and coping. With few opportunities for expressing emotions or vulnerabilities, unrealistic expectations allow shame, guilt, and low self-esteem to surface when women perceive themselves as not meeting the standards. These factors are associated with strained interpersonal relationships, stress-related health behaviors, the embodiment of stress, delayed self-care, decreased help-seeking behaviors, and a lack of social or emotional support [12-16], which erodes resilience and compounds psychological stress, depression, and anxiety [2,14,17]. Further research demonstrates that health practitioners often dismiss Black mothers’ concerns using perceptions informed by a skewed understanding of Black women’s strengths [18]. The SBW schema reinforces a longstanding stereotype that Black women can “naturally endure” pain, affecting how their pain is perceived and managed in the healthcare setting, especially during labor and delivery [18]. As a form of internalized racism, the SBW stereotype threatens Black women’s health and well-being at individual and health system levels with severe implications for maternal and perinatal health.

There is an urgent need for health practitioners to mitigate these adverse maternal and perinatal outcomes [19]. One way is moving away from a physician-centered model of care toward a reproductive justice (RJ) framework of healthcare delivery, which addresses social and structural determinants of health, such as access to quality care, housing, nutrition, education, and diverse forms of racism [20]. RJ seeks to increase access to just and equitable care, improving adverse health outcomes and disparities. In the context of the SBW stereotype, health practitioners work to understand racism and its internalization. They encourage self-determination in perinatal health care experiences [20]. Furthermore, within an RJ framework, health practitioners make timely and appropriate recommendations for therapy while considering factors like racial or cultural concordance [21]. Removing language and policies within healthcare systems that rely on harmful stereotypes, such as the SBW schema, is necessary to improve Black perinatal and infant health outcomes.

Until harmful narratives surrounding Black women’s strength are disassembled, emotional dysregulation, poor mental health, and medical racism will likely continue, allowing for the persistence of poor maternal and perinatal outcomes. RJ seeks to understand better and mitigate the impacts of racism on perinatal and infant health outcomes. Clinical research examining internalized racism and its association with stress, maternal mental health, and birth outcomes are imperative for improving perinatal health care inequities.

Keywords

Perinatal health, Internalized racism, Structural racism, Reproductive justice, Maternal health care, Health communication

References

  1. Gale MM, Pieterse AL, Le DL, Huynh K, Powell S, et al. (2020) A meta-analysis of the relationship between internalized racial oppression and health-related outcomes. The Counseling Psychologist 48: 498-525.
  2. Jefferies K (2020) The strong black woman: insights and implications for nursing. Journal of the American Psychiatric Nurses Association 28: 332-338. [crossref]
  3. Treder K, White KO, Woodhams E, Pancholi R, Yinusa-Nyahkoon L (2022) Racism and the reproductive health experiences of U.S.-born black women. Obstetrics & Gynecology 139: 407-416. [crossref]
  4. Paradies Y, Ben J, Denson N, Elias A, Priest N, et al. (2015) Racism as a determinant of health: A systematic review and meta-analysis. PLOS ONE 10: e0138511. [crossref]
  5. Bridgeman-Bunyoli AM, Cheyney M, Monroe SM, Wiggins N, Vedam S (2022) Preterm and low birthweight birth in the United States: Black midwives speak of causality, prevention, and healing. Birth 49: 526-539. [crossref]
  6. Hoyert D (2022, February). Maternal mortality rates in the United States, 2020. Centers for Disease Control and Prevention.
  7. Brown DL, Rosnick CB, Segrist DJ (2017) Internalized racial oppression and higher education values: The mediational role of academic locus of control among college African American men and women. Journal of Black Psychology 43: 358-380.
  8. David EJR, Schroeder TM, Fernandez J (2019) Internalized racial oppression: A systematic review of the psychological literature on racism’s most insidious consequence. Journal of Social Issues 75: 1057-1086.
  9. Woods-Giscombe CL, Allen AM, Black AR, Stee TC, Li Y, et al. (2019) The Giscombe Superwoman schema questionnaire: Psychometric Properties and associations with mental health and health behaviors in African American women. Issues in Mental Health Nursing 40: 672-681. [crossref]
  10. Collins PH (2000) Gender, black feminism, and Black Political Economy. The ANNALS of the American Academy of Political and Social Science 568: 41-53.
  11. Evans SY, Bell K, Burton NK (2017) Black Women’s Mental Health: Balancing Strength and Vulnerability. SUNY Press.
  12. Nelson T, Cardemil EV, Overstreet NM, Hunter CD, Woods-Giscombé CL (2022) Association between superwoman schema, depression, and resilience: The mediating role of social isolation and gendered racial centrality. Cultural Diversity and Ethnic Minority Psychology. [crossref]
  13. Harrington EF, Crowther JH, Shipherd JC (2010) Trauma, binge eating, and the “strong Black woman”. Journal of Consulting and Clinical Psychology 78: 469. [crossref]
  14. Silva PH, Aiquoc KM, Silva Nunes AD, Medeiros WR, Souza TA, et al. (2022) Prevalence of access to prenatal care in the first trimester of pregnancy among black women compared to other races/ethnicities: A systematic review and meta-analysis. Public Health Reviews 43. [crossref]
  15. Jhingoeri N, Tarini BA, Barber J, Parikh K (2022) Elevated parental stress is associated with lower self-efficacy in provider communication during a pandemic. Hospital Pediatrics 12: 673-679. [crossref]
  16. Liao KYH, Wei M, Yin M (2020) The misunderstood schema of the strong Black woman: Exploring its mental health consequences and coping responses among African American women. Psychology of Women Quarterly 44: 84-104.
  17. Danieli Y, Norris FH, Engdahl B (2016) Multigenerational legacies of trauma: Modeling the what and how of transmission. American Journal of Orthopsychiatry 86: 639-651. [crossref]
  18. Adebayo CT, Parcell ES, Mkandawire-Valhmu L, Olukotun O (2022) African American Women’s maternal healthcare experiences: a Critical Race Theory perspective. Health Communication 37: 1135-1146. [crossref]
  19. Essien U, Molina R, Lasser K (2019) Strengthening the postpartum transition of care to address racial disparities in maternal health. Journal of the National Medical Association 111: 349-351. [crossref]
  20. Julian Z, Robles D, Whetstone S, Perritt JB, Jackson AV, et al. (2020) Community-informed models of perinatal and reproductive health services provision: A justice-centered paradigm toward equity among black birthing communities. Seminars in Perinatology 44: 151267.
  21. Donovan R, West L (2015) Stress and mental health: Moderating role of the strong Black woman stereotype. Journal of Black Psychology 41: 384-296.
fig 4

Vipsyana Meditation May Alter Medication for Mental Health of Older Adults

DOI: 10.31038/ASMHS.2022663

Abstract

Stress cognitions are important for survival, but if they are based on distorted perceptions, they may promote excessive stress arousal, creating a harmful milieu for cellular longevity. While in contrast, emotions based on ‘false projections’ or fear-based beliefs are harmful to longevity of your life. We speculate that certain types of meditation can increase awareness of present moment experience leading to positive cognitions, primarily by increasing meta-cognitive awareness of thought, a sense of control (and decreased need to control), and increased acceptance of emotional experience. These cognitive states and skills reduce cognitive stress and thus ability for more accurate appraisals, reducing exaggerated threat appraisals and rumination, and distress about distress. These positive states are thus stress-buffering. Increasing positive states and decreasing stress cognitions may in turn slow the rate of cellular aging. There is some indirect support of aspects of this hypothesis involving stress cognitions. In our previous study, perceived life stress – primarily an inability to cope with demands and feeling a lack of control, and higher nocturnal stress hormones (cortisol and catecholamines) were related to shorter telomere length. Trait negative mood was related to lower telomerase activity, a precursor of telomere shortening. Here we presented preliminary data from the same sample linking telomere length to higher proportions of challenge appraisals relative to threat appraisals in response to a standardized stressor. The results suggest that the relative balance of threat to challenge cognitions may be important in buffering against the long term wear and tear effects of stressors. To the extent that meditation mitigates stress-related cognitions and propagation of negative emotions and negative stress arousal, a longstanding practice of mindfulness or other forms of meditation may indeed decelerate cellular aging. We also speculate about the physiological mechanisms. Above we have reviewed data linking stress arousal and oxidative stress to telomere shortness. Meditative practices appear to improve the endocrine balance toward positive arousal (high DHEA, lower cortisol) and decrease oxidative stress. Thus, meditation practices may promote mitotic cell longevity both through decreasing stress hormones and oxidative stress and increasing hormones that may protect the telomere. There is much evidence of neuroendocrine and physical health benefits from TM, which has a longer history of study than MBSR. The newer studies of mindfulness meditation are promising, and offer insight into specific cognitive processes of how it may serve as an antidote to cognitive stress states. This field of stress induced cell aging is young, our model is highly speculative, and there are considerable gaps in our knowledge of the potential effects of meditation on cell aging. Several laboratories are working on diverse aspects of this model, which will soon allow it to be evaluated in light of the empirical data.

Older adults, those aged 60 or above, make important contributions to society as family members, volunteers and as active participants in the workforce. While most have good mental health, many older adults are at risk of developing mental disorders, neurological disorders or substance use problems as well as other health conditions such as diabetes, hearing loss, and osteoarthritis. Furthermore, as people age, they are more likely to experience several conditions at the same time. Globally, the population is ageing rapidly. Between 2015 and 2050, the proportion of the world’s population over 60 years will nearly double, from 12% to 22%. Mental health and well-being are as important in older age as at any other time of life. Mental and neurological disorders among older adults account for 6.6% of the total disability (DALYs) for this age group. Approximately 15% of adults aged 60 and over suffer from a mental disorder. The world’s population is ageing rapidly. Between 2015 and 2050, the proportion of the world’s older adults is estimated to almost double from about 12% to 22%. In absolute terms, this is an expected increase from 900 million to 2 billion people over the age of 60. Older people face special physical and mental health challenges which need to be recognized. Over 20% of adults aged 60 and over suffer from a mental or neurological disorder (excluding headache disorders) and 6.6% of all disability (disability adjusted life years-DALYs) among people over 60 years is attributed to mental and neurological disorders. These disorders in older people account for 17.4% of Years Lived with Disability (YLDs). The most common mental and neurological disorders in this age group are dementia and depression, which affect approximately 5% and 7% of the world’s older population, respectively. Anxiety disorders affect 3.8% of the older population, substance use problems affect almost 1% and around a quarter of deaths from self-harm are among people aged 60 or above. Substance abuse problems among older people are often overlooked or misdiagnosed. Mental health problems are under-identified by health-care professionals and older people themselves, and the stigma surrounding these conditions makes people reluctant to seek help.

Risk Factors for Mental Health Problems among Older Adults

There may be multiple risk factors for mental health problems at any point in life. Older people may experience life stressors common to all people, but also stressors that are more common in later life, like a significant ongoing loss in capacities and a decline in functional ability. For example, older adults may experience reduced mobility, chronic pain, frailty or other health problems, for which they require some form of long-term care. In addition, older people are more likely to experience events such as bereavement, or a drop in socioeconomic status with retirement. All of these stressors can result in isolation, loneliness or psychological distress in older people, for which they may require long-term care. Mental health has an impact on physical health and vice versa. For example, older adults with physical health conditions such as heart disease have higher rates of depression than those who are healthy. Additionally, untreated depression in an older person with heart disease can negatively affect its outcome. Older adults are also vulnerable to elder abuse – including physical, verbal, psychological, financial and sexual abuse; abandonment; neglect; and serious losses of dignity and respect. Current evidence suggests that 1 in 6 older people experience elder abuse. Elder abuse can lead not only to physical injuries, but also to serious, sometimes long-lasting psychological consequences, including depression and anxiety (Figure 1).

fig 1

Figure 1

Dementia and Depression among Older People as Public Health Issues

Dementia is the loss of cognitive functioning — thinking, remembering, and reasoning — to such an extent that it interferes with a person’s daily life and activities. Some people with dementia cannot control their emotions, and their personalities may change. It is a syndrome, usually of a chronic or progressive nature, in which there is deterioration in memory, thinking, behaviour and the ability to perform everyday activities. It mainly affects older people, although it is not a normal part of ageing. It is estimated that 50 million people worldwide are living with dementia with nearly 60% living in low- and middle-income countries (Figure 2).

fig 2

Figure 2

The total number of people with dementia is projected to increase to 82 million in 2030 and 152 million in 2050. There are significant social and economic issues in terms of the direct costs of medical, social and informal care associated with dementia. Moreover, physical, emotional and economic pressures can cause great stress to families and carers. Support is needed from the health, social, financial and legal systems for both people with dementia and their carers (Figure 3).

fig 3

Figure 3

Depression

Depression can cause great suffering and leads to impaired functioning in daily life. Unipolar depression occurs in 7% of the general older population and it accounts for 5.7% of YLDs among those over 60 years old. Depression is both underdiagnosed and undertreated in primary care settings. Symptoms are often overlooked and untreated because they co-occur with other problems encountered by older adults. Older people with depressive symptoms have poorer functioning compared to those with chronic medical conditions such as lung disease, hypertension or diabetes. Depression also increases the perception of poor health, the utilization of health care services and costs.

Treatment and Care Strategies to Address Mental Health Needs of Older People

It is important to prepare health providers and societies to meet the specific needs of older populations, including:

  • Training for health professionals in providing care for older people;
  • Preventing and managing age-associated chronic diseases including mental, neurological and substance use disorders;
  • Designing sustainable policies on long-term and palliative care; and
  • Developing age-friendly services and settings.

Health promotion – The mental health of older adults can be improved through promoting Active and Healthy Ageing. Mental health-specific health promotion for older adults involves creating living conditions and environments that support wellbeing and allow people to lead a healthy life. Promoting mental health depends largely on strategies to ensure that older people have the necessary resources to meet their needs, such as:

  • Providing security and freedom;
  • Adequate housing through supportive housing policy;
  • Social support for older people and their caregivers;
  • Health and social programmes targeted at vulnerable groups such as those who live alone and rural populations or who suffer from a chronic or relapsing mental or physical illness;
  • Programmes to prevent and deal with elder abuse; and
  • Community development programmes.

Interventions – Prompt recognition and treatment of mental, neurological and substance use disorders in older adults is essential. Both psychosocial interventions and medicines are recommended. There is no medication currently available to cure dementia but much can be done to support and improve the lives of people with dementia and their caregivers and families, such as:

  • Early diagnosis, in order to promote early and optimal management;
  • Optimizing physical and mental health, functional ability and well-being;
  • Identifying and treating accompanying physical illness;
  • Detecting and managing challenging behaviour; and
  • Providing information and long-term support to careers.

Mental health care in the community -Good general health and social care is important for promoting older people’s health, preventing disease and managing chronic illnesses. Training all health providers in working with issues and disorders related to ageing is therefore important. Effective, community-level primary mental health care for older people is crucial. It is equally important to focus on the long-term care of older adults suffering from mental disorders, as well as to provide caregivers with education, training and support. An appropriate and supportive legislative environment based on internationally accepted human rights standards is required to ensure the highest quality of services to people with mental illness and their caregivers.

Get Your Finances in Order

Organise your money so you can work out what you’ll have to live on. Gradually reducing your spending in the lead up to retirement will make it easier to adjust. Track down any old pensions, claim your state pension and check what other benefits you can claim (Figure 4).

fig 4

Figure 4

Wind Down Gently

Ensure a smoother transition by retiring in stages. By easing off your workload over several years, you’ll be able to get used to the idea of not working and fill your time in other ways. Ask your employer if you can cut back your working hours.

Prepare for Ups and Downs

There may be times when you feel lonely or a bit lost, which is normal. If ill health or changes in your relationships temporarily scupper your plans, accept that this has happened and get your back-up plan in action. Think positively and share any concerns with others. Use your free time to continue to challenge yourself mentally, whether it’s learning an instrument or a language or getting a qualification.

Eat well-Make sure you eat regular meals, especially if your previous pattern, while at work, was to snack. Take advantage of the extra time on your hands and explore healthy cooking options. Fruits and vegetables contain many vitamins and minerals that are good for your health. These include vitamins A (beta-carotene), C and E, magnesium, zinc, phosphorous and folic acid. Folic acid may reduce blood levels of homocysteine, a substance that may be a risk factor for coronary heart disease. Homocysteine is a type of amino acid, a chemical your body uses to make proteins. Normally, vitamin B12, vitamin B6, and folic acid break down homocysteine and change it into other substances your body needs. There should be very little homocysteine left in the bloodstream (Figure 5).

fig 5

Figure 5

Develop a Routine

You may find it feels more normal to continue getting up, eating and going to bed at roughly the same time every day. Plan in regular activities such as voluntary work, exercise and hobbies. This will keep things interesting and give you a purpose.

Exercise Your Mind

Government studies have shown that learning in later years can help people stay independent, so use your free time to continue to challenge yourself mentally, whether it’s learning an instrument or a language or getting a qualification (Figure 6).

fig 6

Figure 6

Keep Physically Active

We should all aim to do at least 150 minutes of moderate-intensity physical activity a week, so build up to this if you haven’t made exercise a normal part of your life previously. Why not sign up for a charity event to give you a goal to work towards ? WHO defines physical activity as any bodily movement produced by skeletal muscles that requires energy expenditure. Physical activity refers to all movement including during leisure time, for transport to get to and from places, or as part of a person’s work.

Make a List

Writing down your aims may help you focus on what you really want to achieve – like a ‘to do’ list. Work out what you can afford to do and schedule time to make it happen, so you experience a sense of accomplishment, as you would have done at work.

Seek Social Support

For many people, work can form a big part of their social life and it’s common to feel at a bit of a loose end once you retire. Fill the gaps by joining clubs and groups. Find out about the social and physical benefits of walking groups.

Make Peace and Move On

Don’t spend your retirement dwelling on your working days. Accept that you’ve done all you can in that job and focus on your next challenge. You’ve still got lots to achieve.

Go for a Health Check

Prevention is better than cure, and now is the perfect time to get your free midlife MOT. The NHS Health Check programme aims to help prevent heart disease, stroke, diabetes, kidney disease and certain types of dementia. Everyone between the ages of 40 and 74, who has not already been diagnosed with one of these conditions or have certain risk factors, will be invited once every five years to have a check to assess their risk of these age-related illnesses and will be given support and advice to help them reduce or manage that risk. If you’re in this category but haven’t had a check in the last five years, you can ask your GP for one.

Keep in Touch with Your Friends from Work

Just because you are retiring doesn’t mean you have to lose touch with the group of friends you made in your workplace. Why not make arrangements for regular catch-ups ? Or, you might want to use some of your new leisure time to catch up with old friends that you haven’t seen for a while. If you enjoy party planning, find an excuse to get everyone together and have fun arranging the perfect garden or dinner party, anniversary celebration or other special occasion. You could even raise funds for our life saving work at the same time through our “Give in Celebration” funds.

Pamper Yourself

After decades of hard work, you are due some ‘me time’. Whether your idea of indulgence is a city break, a day trip to a spa or a small pleasure like dining out or going to the cinema, schedule some time for a well-deserved treat.

Practise Mindfulness

Practising mindfulness has become more popular than ever in the last decade as a strategy to relieve stress, anxiety and depression. Fresh air and exercise is an instant mood booster and instrumental in maintaining your wellbeing. Research, such as a 2009 study from Goethe University in Germany, has shown that meditation strengthens the hippocampus, the area of the brain that is important for memory, and slows the decline of brain areas responsible for sustaining attention. There are no set guidelines for how often you should meditate for optimal result, but a handful of experiments suggest that a mere 10 to 20 minutes of mindfulness a day can be beneficial—if people stick with it.

Give Back to the Community

Ever thought of volunteering? Perhaps you’d enjoy getting involved with your local youth club, animal rescue centre, environmental organisation or elderly support group. There are plenty of charities that would welcome a helping hand, not least the BHF, of course! We offer the opportunity to help out in our shops, in a furniture or electrical store, with fundraising and at lots of different types of events.

Be One with Nature

Fresh air and exercise is an instant mood booster and instrumental in maintaining your wellbeing. Why not incorporate a walk in the woods or a nearby park into your daily routine? This is an ideal way of achieving the recommended minimum of 150 minutes of physical activity per week.

Travel More

Always dreamt of going on an around-the-world cruise, a wine-tasting trip through Italy, or a simple camping expedition in the Welsh valleys? Now you can finally make those long-held plans a reality, depending on your health and budget limitations. If longer trips aren’t practical, mini breaks may be a good alternative – or even days out to places you’ve never visited before.

Get a New Pet or Partner

Could you house a rescue cat or dog in need of a new home? Research has shown that our furry friends have a positive effect on our health and wellbeing.

Push Your Boundaries

It’s easy to get stuck in a rut, both health-wise and in general, and doing something different can be a refreshing change. Some people have found that simple changes, such as trying a tasty new recipe, finding a different hairdresser or joining an exercise class they haven’t done before gives them a new zest for life.

Take Up a New Project

Finally you have time to get stuck into all those things you’ve been meaning to do but never got round to. Mapping your family tree, building a shed, planting a veg patch… the list goes on, but now you can actually do what you’ve always wanted to. Need inspiration? Have a look at our features on gardening, healthy baking, and cycling groups. Read our feature about retirement. Read how volunteering can help you beat loneliness.

A very recent study tested whether an acute bout of exercise would induce a different response on telomerase activity in older vs. young individuals and whether this response would be gender-specific [1]. To test this hypothesis, age- and gender-related differences in telomerase and shelterin responses at 30, 60, and 90 min after a high intensity interval cycling exercise were determined in PBMC of 11 young (22 years) and 8 older (60 years) men and women. A larger increase in telomerase activity, as assessed by TERT mRNA levels, was found in the young compared to the older group after exercise. The second main finding of that study was the higher TERT response to the acute endurance exercise in men compared to women, in whom the response was negligible, independently of age (Figure 7).

fig 7

Figure 7

Those results showed that aging is associated with reduced telomerase activation in response to high-intensity cycling exercise in men [1]. Another study showed that a 30-min treadmill running session was long enough to increase PBMC telomerase activity in 22 young healthy subjects including 11 women and 11 men [2]. Altogether, those recent studies confirm that the increasing telomerase activity after a single bout of exercise could be one of the mechanisms by which physical activity protects against aging [2].

We propose that engaging in a healthy diet and regular physical activity could be both promising strategies to protect telomere maintenance and improve health span at old age (Figure 8 and Table 1).

  • Find a quiet, comfortable place to sit, with your back upright.
  • Put on headphones (this will help block outside distractions).
  • Select the meditation length that’s ideal for you.
  • Press play and close your eyes. Focus your attention on your breath, breathing in and out.

fig 8

Figure 8

Table 1: Professional Advantage of Vipassana

Sl. No.

 Professional Advantage of Vipassana

Students

N

%

1

Developed balanced mind,

29

21.8

2

Control over Tension angry frustration, agitation anxiety, impatience, Reduce stress

56

42.2

4

More empathetic, organized, confidant, orderly and disciplined

18

13.5

5

Objective perception

11

8.3

6

Build good relationship with peers, relatives, and colleague

7

5.3

7

Handle conflict situation

5

3.8

8

Make better decision making

13

9.8

9

Enhance my productivity

1

.8

10

No benefit, Not convinced. Only a spiritual process.

3

2.3

11

Better concentration

14

10.5

  Total

133

100.0

Meditation Reduce depression, tiredness, and fatigue, improve attention, emotion regulation, and mental flexibility. Meditation goes beyond simple relaxation techniques, although that is definitely one of the main benefits. I develop the system called Ven Dr Sumedh Thero system of ordination/Meditation i.e. based on my own experience in India, Sri Lanka, Thailand, Vietnam by meditating as brain exercise and mental energy conservation [3-5]. The study indicates that the Vipassana Meditation process enhanced their professional skills and approaches. Majority students reported that (42.2%) the awareness process helped them to control over their tensions, anxiety and impatience and reduce their anxiety to perceive things professionally than personally [4].

References

  1. Cluckey TG, Nieto NC, Rodoni BM, Traustadottir T (2017) Preliminary evidence that age and sex affect exercise-induced hTERT expression. Gerontol 96: 7-11. [crossref]
  2. Zietzer A, Buschmann EE, Janke D, Li L, Brix M, et al., (2017) Acute physical exercise and long-term individual shear rate therapy increase telomerase activity in human peripheral blood mononuclear cells. Acta Physiol 220: 251-262. [crossref]
  3. Ven Sumedh Thero, Kataria HB, Aditya Suman (2022) Meditation for Skin Aging, Reduces Wrinkles and Change Your Appearance? International Journal of Clinical & Experimental Dermatology 7: 8-12.
  4. Ven Sumedh Thero, Kataria HB, Aditya Suman (2021) How Running Give Us a High Expectations to Overcome Neurological Disorders . Journal of Neurology Research Review & Reports. SRC/JNRRR-157 Volume 3(3): 1-6.
  5. Ven Sumedh Thero (2021) Family Dynamics and Health in Post Covid-19. Clinical Research and Clinical Case Reports 1.

Review on Studies on Genetic Variability of Chickpea (Cicer arietinum L.) Genotypes for Future Breeding Program in Ethiopia

DOI: 10.31038/AFS.2022443

Abstract

Genetic variability studies provide basic information concerning genetic properties of population following which breeding methods could be formulated for future improvement of the crop. Components of genetic parameters such as genotypic coefficient of variation and phenotypic coefficient of variation have an immense importance in detecting the amount of genetic variation exist in the genotypes. Genetic variability study for agronomic traits is a key component of the breeding program for boarding the genetic pool of crop. Once genetic variability of certain crops has been successfully determined crop improvement is easy through the use of appropriate selection methods on yield components hence they are easily inherited than total yield itself. Thus, in this review, studies of genetic variability of chickpea have discussed to help different researchers on their variability studies by providing some important information that will help chickpea improvements.

Key words

Variability, GCV, PCV, Genotypes, Crop Improvement

Introduction

Chickpea (Cicer arietinum L.) is self-pollinated diploid (2n=2×=16) annual leguminous plant belongs to family Fabacea, with a genome size of 738.09 Mbp (Varshney et al., 2013). Chickpea is the third most important pulse crop in the world after faba bean and field pea [1]. Chickpea is one of the first pulse crops domesticated in the Fertile Crescent about 7400 years ago and most probably originated in an area of South-eastern Turkey adjoining Syria [2]. Ethiopia is designated as a secondary center of origin while South-west Asia and the Mediterranean are the two primary center of origin of chickpea according to Vavilov [3].

The achievement of crop improvement through breeding program largely relies on the extent of genetic and phenotypic variability existed among individuals in the population. Selection and development of new variety depends upon the extent of genetic variability in the base population [4] and breeders require existence and extent of interrelationship among important characters for the selection and development of varieties from populations comprising diversified genotypes [5]. The low yields have been attributed to several factors among which include low genetic diversity of cultivated chickpea and several biotic and abiotic stresses [6]. Evaluation and assessment of genetic resources is a pre-requisite for which the future breeding work depends.

Objective

To review Studies on Genetic Variability of Chickpea (Cicer arietinum L.) crop for its future Breeding Program in Ethiopia.

Literature Review

Origin and Distribution

Chickpea (Cicer arietinum L.) belongs to the family Leguminoseae, sub-family Papilionaceae and tribe Cicereae. Chickpea is one of the first pulse crops domesticated in Old World and most probably originated in an area of South-eastern, Turkey adjoining Syria [2,7]. This crop was gradually introduced to the west Mediterranean region, to Eastern and Southern Asia and East Africa. It reached the Indian sub-continent before 2000 BC [8]. Ethiopia is a second center of diversity for chickpea [9]. Chickpea is the only cultivated species within genus Cicer and grown in relatively well-drained black soils, in the cool semi-arid areas of the tropics, sub-tropics as well as the temperate areas [10].

Desi and Kabuli are the two chickpea types produced globally. Kabuli types have a larger cream-colored seed with a thin seed coat whereas the Desi types have a smaller, reddish brown-colored seed with a thick seed coat. Their content also vary in carbohydrates content which ranged from 54 – 71% for Kabuli and 51 – 65 % for Desi type; protein from 12.6 – 29% for Kabuli and from16.7 – 30.6 % for Desi; lipid from 3.4 ¬- 8.8% for Kabuli and from 2.9 – 7.4% for Desi; and energy from 357 – 447 kcal/100g and from 334 – 437 kcal/100g for Kabuli and Desi, respectively [11]. On an average, world production consists of about 75% of Desi and 25% of Kabuli types (EARO, 2004). Although Kabuli types can be profitably adapted in the country, Ethiopia traditionally produces largely the Desi types of chickpea.

Ecology of Chickpea

Chickpea is traditionally grown in the northern hemisphere, mostly at relatively high elevations in India and Ethiopia. However, most of the Desi type chickpea is grown between 20° and 30° N while Kabuli type is grown above 30°N. These environmental conditions give significance difference in photoperiod, temperature and precipitation, all of which have a profound effect on growth and development of the crop. Chickpea requires fertile soil with good drainage system. Any water-logged conditions can severely damage the crop. Chickpeas generally grow on black or red soils and require a soil pH of 6.0 to 7.0. The crop prefers soil with good residual soil moisture content. Chickpeas can be grown on a wide range of soil types provided that the drainage is good and they cannot withstand water logging. For optimum results, clay loams are required. In Ethiopia, chickpea is best adapted to the areas having Vertisols [8].

Economic Importance of Chickpea

Chickpea production has many benefits; first, it fixes atmospheric nitrogen in soils and thus improves soil fertility and saves fertilizer costs in subsequent crops. Second, it improves more intensive and productive use of land, particularly in areas where land is scarce and the crop can be grown as a second crop using residual moisture. Third, it reduces malnutrition and improves human health especially for the poor who cannot afford livestock products. It is an excellent source of protein, fiber, complex carbohydrates, vitamins, and minerals. Fourth, the growing demand in both the domestic and export markets provides a source of cash for small holder producers. Fifth, it increases livestock productivity as the residue is rich in digestible crude protein content compared to cereals [12].

Chickpea Breeding Efforts and Major Achievements in Ethiopia Specifically

According to Asnake et al. [13], the national chickpea and lentil research program came up with 17 superior varieties of chickpea during the decade (2005-2016). The new chickpea varieties have comparative advantages in terms of earliness, Aschochyta blight tolerance, seed size, grain yield, suitability for mechanization and rust resistance among others. The advance in release of chickpea variety for the last decade revealed that 9 Kabili type and 8 Desi type chickpea varieties have been released for production. The release of the chickpea varieties so far was also based on product concepts and market oriented. Despite the release of several improved varieties, however, the variety replacement rate of chickpea is reasonably low. The genetic gains from breeding are also low as compared to the expectation. This calls for improving breeding progress for economic attributes on one hand and effective promotion of the available technologies on the other [13].

In the early phases of chickpea breeding, selections from local landraces were used to develop new varieties. Later the national chickpea improvement program created significant genetic variability for major agro-morphological traits desired by the breeding program through the introduction of diverse germplasm lines from different sources [14]. The major agro-morphological traits prioritized by the breeding program includes productivity, seed size, plant phenology and resistance to key biotic and abiotic stresses prevalent in the country; particularly resistance to wilt/root rot diseases complex, ascochyta blight, major insect pests, drought, moisture and heat stresses.

In breeding programs, combination of bulk and pedigree methods are mainly used in handling several segregating generation developed from different crossing schemes. In early segregating generations, selection is done for simple traits such as disease resistance and seed traits. Screening of several segregating populations and local germplasm genotypes for resistance/tolerance to Fusarium wilt using an aggressive wilt sick plots at Debre Zeit Agricultural Research Center allowed the identification of sources of disease resistance [14].

Precision in selection for different biotic and abiotic stresses such as disease resistance, drought and heat tolerance can be greatly improved by screening several advanced germplasm line/segregating generation under controlled environmental conditions or at hot spot locations. Genomics assisted breeding (GAB) techniques, particularly marker assisted backcross breeding, marker assisted selection/marker assisted recurrent selection have a great potential to enhance precision and efficiency of chickpea breeding program [6]. These days, several success stories of GAB to develop superior varieties are reported in different pulse and cereal crops [15,16]. Therefore, integration of genomics tools in Ethiopia chickpea breeding program has a great potential to speed up the efficiency of selections in the segregating generations for higher and rapid genetic gains.

Moreover, single seed descent and speed breeding/rapid generation advancement methods are already in use at present and needs to be further strengthen to reduce the time required to reach the desired level of homozygosity and to speed up the release of appropriate varieties with desired traits. Adoption of speed breeding technology by generating 4-6 generations per year will be contributing to accelerate genetic gain in legumes breeding program [17].

Genetic Variation, Heritability and Genetic Advance in Genetic Variability Studies

Genetic variability studies provide basic information concerning genetic properties of population following which breeding methods could be formulated for future improvement of the crop [18]. Genetic variability study for agronomic traits is a key component of the breeding program for boarding the genetic pool of crop [4]. Component of genetic parameters such as genotypic coefficient of variation and phenotypic coefficient of variation have an immense important in detecting the amount of genetic variation exist in the genotypes.

Scholars like [19,20] reported high PCV and moderate GCV for number of pods per plant. Another scholars, [21] recorded highest phenotypic and genotypic coefficient of variation (PCV and GCV) for number of pods per plant followed by biological yield per plant and 100-grain weight. In relation to this result [22] reported moderate genotypic coefficient of variations were for grain yield per plant (19.73), number of pods per plant (18.90 %), biological yield per plant (13.56%), number of primary branches per plant (12.75%) and 100-grain weight (11.60%).

Ali and Ahsan [23] reported the presence of greatest genotypic and phenotypic coefficient of variation in chickpea for number of seed per plant, number of pods per plant and plant height. Tesfamichael et al. [1] observed the presence of large variation for days to 50% flowering, plant height, days to maturity, number of pod per plant, 100 seed weight, and seed yield. Johnson et al. [24] conducted an experiment to determine genetic variability of thirty-one chickpea genotypes. His study indicated that the mean sum of squares due to genotypes were significant for all characters studied and suggested the existence of sufficient variability among the genotypes for the traits. This report also showed high values of genotypic coefficients of variation for secondary branches per plant, pod per plant, seed yield per plant, biological yield and primary branches per plant.

Chopdar et al. [25] found the highest genotypic coefficients variations for seed yield and 100-seed weight and moderate coefficient of variation for harvest index, number of pods per plant and biomass per plant and plant height in chickpea. And also genotypic coefficients of variations were low for days to maturity, days to 50 per cent flowering, primary branches per plant and number of seeds per pod. Fasil Hailu [20] reported highest phenotypic and genotypic variance for biological yield, harvest index and number of pods per plant while lowest value was recorded for number of seed per pod, primary branches and secondary branches and said selection is effective for high genotypic and phenotypic variability characters. In addition, Awol et al. [26] reported the lowest PCV and GCV (4.2% and 3.91%) for days to maturity, while the highest PCV and GCV values of 28.64% and 27%; respectively, were obtained for grain yield.

Other scholars [27] reported High GCV was recorded for hundred seed weight (36.01), number of secondary branches (20) and harvest index (22.4) and high PCV for hundred seed weight (36.02), number of secondary branches (27.53), harvest index (23.37), grain yield (22.89) and biological yield. And also Moderate GCV and PCV were noted for traits such as grain yield, biological yield, number of primary branches, days to emergence and number of pods per plant, and number of primary branches, days to emergence and number of pods per plant, respectively. Higher phenotypic and genotypic coefficient of variability indicates the existence of wide genetic variation among the genotypes under the study so that genetic improvement could be possible through selection.

Hussain et al. [22] also reported low GCV for days to 50% flowering (2.10%).  Traits with low GCV and PCV indicate presence of narrow genetic variability. Traits such as physiological maturity, days to flowering, number of seed per pod, grain filling period and plant height was showed low GCV and PCV value. Thus, improvement for such traits could be possible through hybridization followed by selection.

Dubey and Srivastava [28] also reported high heritability (broad sense) for plant height, number of pods per plant, 100-grain weight and grain yield per plant. Malik et al (2010) reported high broad sense heritability estimates for number of secondary branches, harvest index, hundred seed weight and number of pods per plant and grain yield.

Chand et al. [29] reported that the highest broad sense heritability estimate was obtained for 100 seed weight (80%), number of seeds per pod (77%) and number of primary branch (62%) in chickpea genotypes. Hussain et al. [22] studied genetic variability and mode of inheritance in eight quantitative traits of chickpea and reported that high broad sense heritability for grain yield (96.40%), number of pods per plant (93.19%), 100-grain weight (89.67%), biological yield (83.83%) and plant height (78.83). According to Chopdar et al. [25], 100-seed weight had the highest heritability followed by days to 50% flowering (85.31), seed yield per plant (84.29), days to maturity (78.39), biomass per plant (69.06), number of pods per plant (68.06) and harvest index (67.51) in chickpea.

Parameshwarappa et al. [30] reported high heritability with high genetic advance as percent of mean for pods per plant, 100-seed weight and seed yield per plant, suggesting that these traits could be improved through simple selection. High heritability with high genetic advance as percent of means is the indication for the presence of additive gene action. He also found that high heritability with moderate genetic advance as percent of means for days to 50 percent flowering, and high heritability with low genetic advance as percent of means for plant height, primary branches per plant and secondary branches per plant.

According to Biru et al. [31] hundred seed weight, number of pods per plant, number of seed per pod and grain yield showed high heritability combined with high genetic advance as a percentage of mean. Moreover, low heritability and expected genetic advance were observed for days to maturity and branches per plant.

Joshi et al. [32] observed that high estimates of heritability in broad sense for days to 50% flowering, days to maturity, plant height, biological yield per plant, seed yield, harvest index and 100-seed weight, indicating that these characters were less affected by the environment and the plant breeder may use these characters for selection on the basis of phenotypic expression in the individual material.

Awol et al. [26] reported high broad sense heritability estimate for grain yield, hundred seed weight, biological yield, number of pods per plant, days to flowering, plant height, number of primary branches and number of secondary branches. Also Alemayo et al. [27] in their study on variability of 56 chickpea genotypes, estimate for all thirteen traits showed high (>60%) broad sense heritability. The highest heritability was obtained for hundred seed weight (99.96%) while the lowest was for number of seed per pod (62.96%). Presence of high broad sense heritability indicates selection based on phenotypic expression of individual genotypes for such characters might be easy due to relatively small effects of environment on phenotype.

But Arora and Jeena [33] recorded low value of heritability for days to 50% flowering; Singh and Rao (1991) for number of primary branches per plant. According to Johnson et al. [24] heritability alone might not be effective unless coupled with higher genetic advance as percent of the mean in predicting the effectiveness of selecting the best performing genotypes and he classified estimates of genetic advances as a percentage of the mean excelling 20% as high, ranging from 10 to 20% as moderate and those which showed below 10 as low.

Zali et al. [19] reported that the genetic advance (5% selection intensity) was the highest for number of secondary branches, number of seeds per plant and seed yield. This implies that progress on improving seed yield could be achieved through simple selection of the number of secondary branches and number of seeds per plant. Another scholar [22] reported high heritability coupled with high genetic advance as percent of mean for grain yield per plant (39.91), number of pods per plant (37.59), biological yield per plant (25.58) and 100-grain weight (22.62).

In their research, Alemayo et al. (2021) reported high heritability coupled with high genetic advance as percent of the mean were recorded for hundred seed weight (74.26%), number of secondary branches (54.65%), harvest index (44.34%), grain yield (33.38%), biological yield (32.6%), number of primary branches (28.35%), days to emergence (27.3%) and number of pods per plant (25.8%). High estimate of genetic advance as percent of the mean for these traits indicates that whenever we select the best 5% genotypes as parent for a given trait, genotypic value of the new population for the traits will be improved highly and they are governed by additive gene [34].

In addition, Alemayo et al. [27] reported, for traits like plant height, grain filling period and days to flowering showed high heritability with moderate genetic advance as percent of mean. This shows that, such traits are primarily under genetic control and their selection can achieved based on their phenotypic performance. In another hand they reported as, Low genetic advance as percent of mean were observed for number of seed per pod and days to physiological maturity. This suggested that the expressions of such traits are controlled by non-additive gene action and their selection might not be satisfactory. So, the appropriate usage of pure line selection may be valuable for improving these characters with moderate or high heritability characters [35].

Conclusion and Future Prospects

For better improvement of a crop study on genetic variability is the crucial that help in getting sufficient information regarding per se performance, variability, heritability, genetic advance and association that may exist among yield and its components – agronomic and phonological traits. Because, without presence of genetic variability among genotypes (in our case, among chickpea genotypes) there is no success for its improvement. So, for successful breeding program [for chickpea] researchers should focus on creating variability among chickpea genotypes.

References

  1. Tesfamichael SM, Githiri SM, Nyende AB, Rao NVPRG (2015) Variation for agro morphological traits among Kabuli chickpea (Cicer arietinum L.) genotypes. Journal of Agricultural Science 7: 75.
  2. Van der Maesen LJG (1987) Origin, history and taxonomy of chickpea 11-34.
  3. Vavilov NI (1926) The origin of the cultivation of ‘primary ‘crops, in particular cultivated hemp. Studies on the origin of cultivated plants 221-233.
  4. Singh BD (2001) Plant breeding: Principles and Methods. Kalyani Publishers, New Delhi.
  5. Falconer DS, Mackay FC (1996) Introduction to Quantitative Genetics. Longman, New York.
  6. Gaur PM, Jukanti AK, Varshney RK (2012) Impact of genomic technologies on chickpea breeding strategies. Agronomy 2: 199-221.
  7. Toker C, Cagirgan IC (2004) The use of phenotypic correlations and factor analysis in determining characters for grain yield selection in chickpea (Cicer arietinum L.). Hereditas 140: 226-228. [crossref]
  8. Martin JH, Waldren RP, Stamp DL (2000) Principles of Field Crop Production. Fourth ed., Macmillan Publishing Corporation: USA.
  9. Pundir RPS, Hailemariam Mengesha (1983) Collection of chickpea in Ethiopia. Inter. Chickpea Newsletter, 8: 6-7.
  10. Atta BM, Shah TM (2009) Stability analysis of elite chickpea genotypes tested under diverse environments. Australia Journal of Crop Science 3: 249-256.
  11. Wood JA, Grusak MA (2007) Nutritional value of chickpea. EARO (Ethiopian Agricultural Research Organization), 2004. Directory of Released Crop Varieties and their Recommended Cultural Practices. EARO: Addis Ababa, Ethiopia 101-142.
  12. FDRE (Federal Democratic Republic of Ethiopia) (2010) Extension Package for Pulse Production and Improved Management Practices. Agricultural Extension Directorate. Amharic version. FDRE: Addis Ababa, Ethiopia.
  13. Asnake F, Lijalem K, Million E, Dagnachew B, Niguse G, et al., (2018) A Decade of Research Progress in Chickpea and Lentil Breeding and Genetics. Crop Sci 6: 101-113.
  14. Asnake F, Dagnachew B (2020) Chickpea Breeding and Crop Improvement in Ethiopia: Past, Present and the Future. Universal Journal of Agricultural Research 8: 33-40.
  15. Varshney RK, Hoisington DA, Tyagi AK (2006) Advances in cereal genomics and applications in crop breeding. Trends Biotechnol 24: 490-499. [crossref]
  16. Varshney RK, Thudi M, May GD, Jackson SA (2010) Legume Genomics and Breeding. Plant Breeding Rev 33: 257-304.
  17. Varshney RK, Pandey MK, Bohra A, Singh VK, Thudi M (2018) Toward the sequence based breeding in legumes in the post genome sequencing era. Theoretical and Applied Genetics 132: 797-816.
  18. Khlestkina EK, Huang XQ, Quenum FB, Chebotar S, Röder MS, et al. (2004b) Genetic diversity in cultivated plants – loss or stability? Theoretical and Applied Genetics 108: 1466-1472. [crossref]
  19. Zali H, Farshadfar E, Sabaghpour SH (2011) Genetic variability and inter relationships among agronomic traits in chickpea (Cicer arietinum L.) genotypes. Crop breeding journal 1: 127-132.
  20. Fasil Hailu Gebremichael M.Sc Thesis (2019) Genetic variability and character association of kabuli chickpea (Cicer arietinum L.) Genotypes for grain yield and related traits at Debre zeit and akaki, central Ethiopia.
  21. Dwevedi KK, Gaibriyal ML (2009) Assessment of genetic diversity of cultivated chickpea (Cicer arietinum L.). Asian J. Agri. Sci 1: 7-8.
  22. Hussain N, Ghaffar A, Aslam M, Hussain K (2016) Assessment of genetic variation and mode of inheritance of some quantitative traits in chickpea (Cicer arietinum L.). JAPS: Journal of Animal & Plant Sciences 26: 1334-1338.
  23. Ali Q, Ahsan M (2012) Estimation of genetic variability and correlation analysis for quantitative traits in chickpea (Cicer arietinum L.). International Journal for Agro Veterinary and Medical Sciences 6: 241-249.
  24. Johnson PL, Sharma RN, Nanda HC (2015) Genetic diversity and association analysis for yield traits chickpea (Cicer arietinum L.) under rice based cropping system. The Bioscan 10: 879-884.
  25. Chopdar DK, Bharti B, Sharma PP, Dubey RB, Meena BB (2017) Studies on genetic variability, character association and path analysis for yield and its contributing traits in chickpea [Cicer arietinum (L.)]. Legume Research-An International Journal 40: 824-829.
  26. Awol Mohammed, Bulti Tesso, Chris Ojiewo, Seid Ahmed (2019) Assessment of genetic variability and heritability of agronomic traits in Ethiopian chickpea (Cicer arietinum L.) landraces. Black Sea Journal of Agriculture 2: 10-15.
  27. Alemayo GT, Gurmu GN, Singh BCS, Getachew Z (2021) Genetic Variability of Chickpea (Cicer Arietinum L.) Genotypes for Yield and Yield Components in West Shewa, Ethiopia. Plant Cell Biotechnology And Molecular Biology 307-331.
  28. Dubey KK, Srivastava SBL (2007) Study of direct selection in chickpea (Cicer arietinum L.). Plant Archives 7: 211-212.
  29. Chand JU, Singh DP, Roopa LG (2012) Assessment of genetic variability and correlation of important yield related traits in chickpea (Cicer arietinum L.). Legume Research: An International Journal 35.
  30. Parameshwarappa SG, Salimath PM, Upadhyaya HD, Patil SS, Kajjidoni ST (2012) Genetic variability studies in mini core collection of chickpea (Cicer arietinum L.) under different environments. Karnataka Journal of Agricultural Sciences 25: 305-308.
  31. Biru A, Kassahun T, Teklehaimanot H, Dagnachew L (2017) Broad sense heritability and genetic advance for grain yield and yield components of chickpea (Cicer arietinum L.) genotypes in western Ethiopia. International Journal of Genetics and Molecular Biology 9: 21-25.
  32. Joshi P, Yasin M, Sundaram P (2018) Genetic variability, heritability and genetic advance study for seed yield and yield component traits in a chickpea recombinant inbred line (RIL) Population. International Journal of Pure and Applied Bioscience 6: 136-141.
  33. Arora PP, Jeena AS (2000) Variability in relation to response to selection in chickpea. Sci. Dig 20: 267-298.
  34. Noor F, Ashaf M, Ghafoor A (2003) Path analysis and relationship among quantitative traits in chickpea (Cicer arietinum L.). Pakistan J. Biol. Sci 6: 551-555.
  35. Arshad M, Bakhsh A, Bashir M, Haqqani AM (2002). Determining the heritability and relationship between yield and yield components in chickpea (Cicer arietinum L.). Pakistan J. Bot 34: 237-245.
fig 1a & 1b

Developing an Inner Psychophysics for Social Issues: Reflections, Experiments, and Futures

DOI: 10.31038/PSYJ.2022444

Abstract

The objective of Inner Psychophysics is to provide a number or a metric, on ideas, with the number showing the magnitude of the idea on a specific dimension of meaning. We introduce a new approach to measuring the values of ideas, applying the approach to the study of responses to 27 different types of social problems. The approach to create this Inner Psychophysics comes from the research system known as Mind Genomics. Mind Genomics presents the respondent with the social problem, and a unique set 24 vignettes, viz., combinations of ‘answers’ to that social problem, these vignettes created by an underlying experimental design. The respondent rates each test vignette using a scale of solvability. The pattern of responses to the vignettes is deconstructed into the contribution of each ‘answer’, through OLS (ordinary least squares) regression. The OLS regression across a group of respondents provides the psychological magnitude of the solution offered as judged so to solve the particular problem. The approach opens up the potential of a ‘metric for the social consensus,’ measuring the value of ideas relevant to society as a whole, and to the person in particular.

Introduction

Psychophysics is the oldest branch of experimental psychology, dealing with the relation between the physical world (thus ‘physics’) and the subjective world of our own consciousness (thus ‘psycho’). The question might well be asked what is this presumably arcane psychological science dealing with up to date, indeed new approaches to science? The question is relevant, and indeed, as the paper and data will show. The evolution of an ‘inner psychophysics’ provides today’s researcher with a new set of tools to think about the problems of the world. The founder of today’s ‘modern psychophysics,’ encapsulated the opportunity in his posthumous book, ‘Psychophysics: An Introduction to its Perceptual, Neural and Social Prospects. This paper presents the application of psychophysical thinking and disciplined rigor to the study of how people ‘think’ about problems. Stevens also introduced the phrase ‘a metric for the social consensus,’ in his discussions about the prospects of psychophysics in the world of social issues [1-3].

The original efforts in psychophysics began about 200 years ago, with the world of physiologists and with the effort to understand how people distinguish different levels of the same stimulus, for example, different levels of sugar in water, or today, different levels of sweetener in cola. Just how small of a difference can we perceive? Or, to push things even more, what the is lowest physical level of a stimulus that we can detect? [4]. These are the difference and the detection threshold, respectively, both of interest to scientists, but of relatively little interest to the social scientist and research, unless we are dealing in psychology, food science, or perhaps loss of sensory function due to accident or disease.

The important thing to come out of psychophysics is the notion of ‘man as a measuring instrument,’ the notion that there is a metric of perception. Is there a way to assign numbers to objects or better to experiences of objects, so that one can understand what happens in the mind of people, when these objects are mixed, changed, masked, etc.? In simpler terms, think of a cup of coffee. If we can measure the subjective perception of aspects of that coffee, such as its coffeeness’, then what happens when we add milk. Or add sugar. Or change coffee roast, and so forth. At a mundane level, can we measure how much perceived coffeeness changes?

Steven’s ‘Outer’ and ‘Inner’ Psychophysics

By way of full disclosure, author HRM was one of the last PhD students of the SS Stevens, receiving his PhD in the early days of 1969. Some 16 months before, Stevens had suggested that HRM ‘try his hand’ at something such as taste or political scaling, rather than pursuing research dealing with topics requiring sophistication in electronics, such as hearing and seeing. That suggestion would become a guide through a 54-year future, now a 54-year history. The notion of measuring taste forced thinking about the mind, the way people say things taste versus how much they like what they taste. This first suggestion, studying taste, focused attention on the inner world of the mind, one focused on what things taste like, why people differ in what they like, whether there are basic taste preference groups, and so forth. The well-behaved laws of psychophysics – ‘change this, you get that,’ working so well in loudness, seem to break down in taste. Change the sugar in cola or in coffee, and you get more/less coffee flavor, but you like the coffee more, and so forth. Here was the next level of exploration, a more ‘inner-focused world’.

If taste was to be the jumping off portion from this outer psychophysics to the measurement of feelings, such as liking, then the next efforts would be even more divergent. How does one deal with social problems which have many aspects to them? We are no longer dealing with simple ingredients, which when mixed create a food, and whose mixtures can be evaluated by a ‘taster’ to find out rules. We are dealing now with the desire to measure the perception of a compound situation, with many factors. Can the spirit of psychophysics add something, or we stop at sugar coffee, or salt in pickles?

Some years later, through ongoing studies of perception, it became obvious that one could deal with the inner world, using man as a measuring instrument. The slavish adherence of systematic change of the stimulus in degrees and the measurement, had to be discarded. It would be nice to say that a murder is six times more serious than a bank robbery with two people injured, but that type of slavish adherence would not create this new inner psychophysics. It would simply be adapting and changing the hallowed methods of psychophysics (systematically change, and then measure), moving from tones and lights to sugar and coffee, and now to statements about crimes. There would be some major efforts, such as the utility of money [5], efforts to maintain the numerical foundations of psychophysics because money has an intrinsic numerical feature. Another would be the relation between perceived seriousness of crime and the measurable magnitude punishment.

Enter Mathematics: The Contribution of Conjoint Measurement, and Axiomatic Measurement Theory

If psychophysics provided a strong link to the empirical world, indeed a link which presupposed real stimuli, then mathematical psychological provided a link to the world of philosophy and mathematics. The 1950’s saw the rise of interest in mathematics and psychology. The goal of mathematical psychological in the 1950’s and 1960’s was to put psychology on firm theoretical footing. Eugene Galanter became an active participant in this newly emerging, working at once with Stevens in psychophysics at Harvard, and later with famed mathematical psychologist R. Duncan Luce. Luce and his colleagues were interested in ‘fundamental measurement’ of psychological quantities, seeking to measure psychology with the same mathematical rigor that physicists measured the real world. That effort would bring to fruition the Handbook of Mathematical Psychology, and well as the efforts of psychologist coining the term ‘functional measurement [6-9].

The simple idea which is relevant to us is that one could mix test stimuli, ideas, not only food ingredients, instruct the respondent to evaluate these mixtures, and estimate the contribution of each component to the response assigned to the mixture suggested deeply mathematical, axiomatic approaches to do that. Anderson suggested simpler approaches, using regression. Finally, the pioneering academics at Wharton Business School, showed how the regression approach could be used to deal with simple business problems [10-12].

The history of psychophysics and the history of mathematical psychology met in the systematics promised by and delivered by Mind Genomics. The mathematical foundations had been laid down by axiomatic measurement theory. The objective, systematized measurement of experience, had been laid down by psychophysics at first, and afterwards by applied psychology and consumer research. What remained was to create a ‘system’ which could quantify experience in a systematic way, building databases, virtually ‘wikis of the mind’, rather than simply providing one or two papers on a topic which solved a problem with an interesting mathematics. It was time for the creation of a corpus of psychophysically motivated knowledge, an inner psychophysics of thought, rather than the traditional psychophysics of perception.

Reflections on the Journey from the Outer Psychophysics to an Inner Psychophysics

New thinking is difficult, not so much because of the problems as the necessity to break out of the paradigms which one ‘knows’ to work, even though the paradigm is not the best. The inertia to remain with the tried and true, the best practices, the papers confined to topics that are publishable, is endemic in the world of academics and thinking. At the same time, inertia seems to be a universal law, whether the issue is science and knowledge, or business. This is not the place to discuss the business aspect, but it is the place to shine a light on the subtle tendency to stay within the paradigms that one learned as a student, the tried and true, those paradigms which get one published.

The beginning of the journey to inner psychophysics occurred with a resounding NO, when author HRM asked permission to combine studies of how sweet an item tasted, and how much the item was liked. This effort was a direct step away from simple psychophysics, with the implicit notion of a ‘right answer’. This notion of a ‘right answer’ summarizes the world view espoused by Stevens and associates that psychophysics was searching for invariance, for ‘rules’ of perception. Departures from the invariances would be seen as the irritating contribution of random noise, such as the ‘regression effect’, wherein the tendency of research is to underestimate the pattern of the relation between physical stimulus and subjective, judged response. “Hedonics” was a complicating, ‘secondary factor’, which could only muddle the orderliness of nature, and not teach anything, at least to those imbued with exciting Harvard psychophysics of the 1950’s and 1960’s.

The notion of cognition, hedonics, experience as factors driving the perception of a stimulus, could not be handled easily in the new outer psychophysics except parametrically. That is, one could measure the relation between the physical stimulus and the subjective response, create an equation with parameters, and see how these parameters changed when the respondent was given different instructions, and so forth. An example would be judging the apparent size of a circle of known diameter versus judge the actual size. It would be this limitation, this refusal to accept ideas as subject to psychophysics, that author HRM, would end up attempting to overcome during the course of the 54-year journey.

The course of the 54-year journey would be marked by a variety of signal events, events leading to what is called in today’s business ‘pivoting.’ The early work on the journey dealt with judgments of likes and dislikes, as well as sensory intensity [13]. The spirit guiding the work was the same, search for lawful relations, change one parameter, and measure the change in a parameter of that lawful relation. The limited, disciplined approach of the outset psychophysics was too constraining. It was clear at the very beginning that the rigorous scientific approaches to measuring perceptual magnitudes using ‘ratio-scaling’ would be a non-starter. The effort of the 1950’s and 1960’s to create a valid scale of magnitude was relevant, but not productive in a world where the application of the method would drown out methodological differences and minor issues. In other words, squabbles about whether the ratings possessed ‘ratio scale’ properties might be interesting, but not particular productive in a world begging for measurement, for a yet-to-be sketched out inner psychophysics.

The movement away from simple studies of perceptual magnitudes was further occasioned by the effort to apply the psychophysical thinking to business issues, and the difficulties ensuing in the application of ratio scaling methods such as magnitude estimation. The focus was no longer on measurement, but on creating sufficient understanding about the stimulus, the food or cosmetic product, so that the effort would generate a winner in in the marketplace.

The path to understanding first comprises experiments with mixtures, first mixtures of ingredients, and then mixtures of ideas, steps needed to define the product, to optimize the product itself, and then to sell the product. Over time, the focus turned mainly to ideas, and the realization that one could mix ideas (statements), present these combinations to respondents, get the responses to the combinations, and then using statistics such as OLS (ordinary least-squares regression) one could estimate the contribution of each idea in the mixture to the total response.

Inner Psychophysics Propelled by the Vision of Industrial-scale Knowledge Creation

A great deal of what the author calls the “Inner Psychophysics” came about because of the desire to create knowledge at a far more rapid level than was being done, and especially the dream that the inevitable tedium of a psychophysical experiment could simply be eliminated. During the 20th century, especially until the 1980’s, researchers were content to work with one subject at a time, the subject being call the ‘O’, an abbreviation for the German term Beobachter. The fact that the respondent is an observer suggests a slow, well-disciplined process, during which the experimenter presents one stimulus to one observer, and measures the response, whether the response is to say when the stimulus is detected as ‘being there’, when the stimulus quality is recognized, or when the stimulus intensity is to be assigned a response to report its perceived intensity.

The psychophysics of the last century, especially the middle of the 20th century, focused on precision of stimulus, and precision of measurement, with the goal of discovering the relations between variables, on the one hand physical stimuli and on the other subjective responses. It is important to keep in mind the dramatic pivot or change in thinking. Whereas psychophysics of the Harvard format searched for lawful relations between variables (physical stimulus levels; ratings of perceived magnitude), the application of the same thinking to food and to ideas was to search for lawful, usable relation. The experiments need not reveal an ‘ultimate truth’, but rather needed to be ‘good enough,’ to identify a better pickle, salad dressing, orange juice or even features of a cash-back credit card.

The industrial-scale creation would be facilitated by two things. The first was a change in direction. Rather than focusing one’s effort on the laws relating physical stimulus and subjective response (outer psychophysics), a new, and far-less explored area would focus on measuring ideas, not actual physical things (inner psychophysics).

The second would focus on method, on working not with single ideas, but deliberately with mixtures of ideas, presented to the respondent in a controlled situation, and evaluated by the respondent. These mixtures would be created by experimental design, a systematic prescription of the composition of each mixture, viz., which phrases or elements would appear in each vignette. The experimental design ensured that the researcher would be able to link a measure of the respondent’s thinking to the specific elements. The rationale for mixtures was the realization that single ideas were not the typical ‘product’ of thought. We think of mixtures because our world comprises compound stimuli, mixtures of physical stimuli, and our thinking in turn comprises different impressions, different thoughts. Forcing the individual to focus on one thought, one impression, one message or idea, is more akin to meditation, whose goal is to shunt the mind away from the blooming, buzzing confusion of the typically disordered mind, filled with ideas flitting about.

The world view was thus psychophysics, search for relations and for laws. The world view was also controlled complexity, with the compound stimulus taking up the attention of the respondent and being judged. The structure of the mixtures appeared to be a ‘blooming, buzzing confusion’ in the words of Harvard psychologist William James. To create the inner psychophysics meant to prevent the respondent from taking active psychological control of the situation. Rather, the designed forced the respondent to pay attention to combinations of meaningful messages (vignettes), albeit messages somewhat garbled in structure to avoid revealing the underlying structure, and thus to prevent the respondent from ‘gaming’ the system.

As will be shown in the remainder of this paper, the output of this mechanized approach to research produced an understanding of how we think and make decisions, in the spirit of psychophysics, at pace and scope that can be only described as industrial scale. Some of the reasons for the term ‘industrial scale production of knowledge’ come from the manner that the approach was used, viz. evaluation of systematic mixture of ideas.

The Mind Genomics ‘Process’ for Creating an Experiment

The study presented here comes from a developing effort to understand the mind of ordinary people in term of what types of actions can solve well-known social problems. At a quite simple level, one can either ask respondents to tell the researcher what might solve the problems or present solutions to the respondent and instructed to scale each solution in terms of expected ability to solve the problem. The solutions are concrete actions, simple and relevant. The pattern of responses gives a sense of what the respondent may be thinking with respect to solving a problem.

The study highlighted here went several stages beyond that simple, straightforward approach. The inspiration came from traditional personality theory, and from cognitive psychology. In personality theory, psychologist Rorschach among many others believed that people were not often able to paint a picture of what was going on in their minds. Rorschach developed a set of ambiguous pictures, and instructed the respondent to say what they ‘saw’. The pattern of what the respondent reported ‘seeing’ suggested how the respondent organized her or his perceptions of the world. Could such an approach be generalized, so that the pictures would be replaced by metaphoric words, rich with meaning? And so was born the current study. The study combines a desire to understand the mind of the individual, the use of Mind Genomics to do the experiment, and the acceleration of knowledge development through a novel set of approaches to the underlying experimental design [14].

The process itself follows a series of well-choreographed steps, leading to statistical analyses and then to pattern recognition of possible underlying processes:

  1. The structure of the experimental design begins with a single topic (e.g., a social problem), continues with four questions dealing with the problem, and in turn uses four specific answers to each question. The three stages are easy to do, becoming a template. Good practice suggests that the 16 answers (henceforth elements) be simple declarative statements, each comprising 12 words or fewer, with no conjunctives. These declarative statements should be easily and quickly scanned, with as little ‘friction’ as possible.
  2. The specific combinations are prescribed by an underlying experimental design. The experimental design . The experimental design ensured that each element appeared exactly five times across the 24 vignettes, and that the pattern of appearances made each element statistically independent of the other 15 elements. A vignette could have at most one element or answer from a question. The actual design generates vignettes comprising a mixture of 4-element vignettes, 3-element vignettes, and 2-element vignettes, respectively, but never a 1-element vignette.
  3. The experimental design was set so that the data from each respondent, viz., the vignettes and their ratings, could be analyzed by ordinary least-squares (OLS) regression. That is, each respondent’s data comprised an entire experiment. The data could be analyzed in groups, or at the level of the individual. For this paper, the focus will be on the results emerging from the OLS regression at the level of each respondent.
  4. A key problem in experimental design is the focus on testing one specific set of combinations out of the large array of the underlying ‘design space’. The quality of knowledge suffers because only one small region of the design space is explored, usually that region believed to be the most promising, whether that belief is correct or not. . There is much more to the design space. The research resources are wasted minimizing the “noise” in this presumably promising region, either by eliminating noise (impossible in an Inner Psychophysics), or by averaging out the noise in this region by replication (a waste of resources).
  5. The solution of Mind Genomics is to permute the experimental design [15]. The permutation strategy maintains the structure of the experimental design but changes the specific combinations. The task of permuting requires that the four questions be treated separately, and that the elements within a question be juggled around but remain with the question. In this way, no element was left out, but rather its identification number changed. For example, A1 would become A3, A2 would become A4, A4 would become A2 and A3 would become or remain A3. At the initial creation of the permuted designs, each new design was tested to ensure that it ran with the OLS (ordinary least-squares) regression package. Each respondent ends up evaluating a different set of 24 combinations.
  6. The benefit to research is that research becomes once again exploratory as well as confirmatory, due to the wide variation in the combinations. It is no longer a situation of knowing the answer or guessing at the answer ahead of time. The answer emerge quickly. The data from the full range of combination tested quickly reveal what elements perform well versus what elements perform poorly.
  7. Continuing and finishing with an overview of the permuted design of Mind Genomics, it would be quickly obvious that studies need not be large and expensive. The ability to create equations or models with as few as 5-10 respondents, because of the ability to cover the design space, means that one can being to understand the ‘mind’ of people with so-called ‘demo studies’, virtually automatic studies, set up and implemented at low cost. The setup takes about 20 minutes once the ideas are concretized in the mind of the research. The time from launch (using a credit card to pay) to delivery of the finalized results in tabulated form, ready for presentation, is approximately 15-30 minutes.
  8. The final step, as of this writing (Fall, 2022), is to make the above-mentioned system work with a series of different studies of social problems, here, 27 studies. In the spirit of accelerated knowledge development, each study is a carbon copy of every other study, except for one item, the specific social issue addressed in the study. That is, the orientation, rating scale, and elements are identical. What differs is the problem. When everything else is held constant, only the topic being varied, we have then the makings of the database of the mind, done at industrial scale

Applying the Approach to the ‘Solution’ of Social Problems

We begin with a set of 27 social problems, and a set of solutions. The problems are ones which are simple to describe and are not further elaborated. In turn the 16 element or solutions are general approaches, such as the involvement of business, rather than more focused solutions comprising specific steps. The 27 problems are shown in Table 1, and the 16 solutions are shown in Table 2. For right now, it is just important to keep in mind that these problems and solutions represent a small number of the many possible problems one can encounter, and the solutions that might be applied. For this introductory study, using the Mind Genomics template, we are limited to four types of solutions for a problem, and four specific solutions each solution type. The number of problems is unlimited, however.

  1. Figure 1a and 1b shows two screen shots. Each problem is represented by a single phrase, describing the problem. That phrase is called ‘the SLUG’. In the figures, the words ‘ABORTION RIGHTS’ constitute the SLUG. The SLUG changes in each study, to present the topic of that study. There is no further elaboration of the topic as art of the introduction.
  2. The approach is templated, allowing the researcher to set up the study within 40 minutes, once the researcher identifies the social problem, creates the four questions, creates the four answers to each question (16 answers or elements), and then creates the rating scale. The researcher simply fills in the template as shown for one study, abortion, in Figure 1a and Figure 1b, respectively.
  3. Since the study is templated and of the precise same format except the topic, moving from one study to 26 copies becomes a straightforward task. The researcher copies the base study, but then changes the nature of the problem in the introduction, and the rating scale. This activity requires about 10 minutes per study. The activity simply requires the change of SLUGS. The total time for this ‘reproduction’ step is about two hours for the 26 remaining studies.
  4. Launch the 27 studies in rapid sequence. Each study requires about 1-2 minutes to launch, an effort accomplished in about one hour or less. The 27 studies run in parallel, each with about 50 respondents. With ‘easy-to-find’ respondents, the 27 steps take about two hours to run in the field, since they are running simultaneously, and require only a total of 1350 respondents.
  5. Using the ‘raw data’ files generated by the program, combine the raw data from the 27 studies into one comprehensive data file comprising all the data. Each respondent generates 24 rows of data. A study of one topic generates 24×50 or 1200 rows of data. The 27 studies generate 1200×27 rows of data. The effort to combine the data, ensuring that each study is properly incorporated into the large-scale database, requires about 2 hours.
  6. Convert the ratings so that ratings of 1-3 are converted to 0, to reflect the fact that the respondent does not feel that the combination of solutions will solve the social problem. Convert ratings of 4 and 5 to reflect the fact that the respondent does feel that the combination of solutions presented in the vignette will solve the social problem. Thus the ratings assigned by the respondent, a 5-point scale, are converted to a no/yes scale. To each converted value, viz., 24 binary values for each respondent, one per vignette, add a vanishingly small random number (~ 10-3). The small random number will not affect the results but will ensure variation in the newly created binary variable, (0=will not work, 100=will work). This type of conversion, viz., from a Likert Scale (multi-point category scale) to a binary scale, is a hallmark of Mind Genomics. The conversion comes from the history of consumer researchers and public opinion researchers working with YES/NO scales because managers do not understand what to do with averages of ratings. The averages have statistical meaning, of course, but have little built in meaning for a manager who has to make business decisions.
  7. Since the 24 vignettes evaluated by a respondent are created according to an underlying experimental design, we know that the 16 independent variables (viz., the 16 solutions) are statistically independent of each other. Create a model (equation) for each respondent relating the presence/absence of the 16 elements to the newly created binary variable ‘solve the problem.’ The equation does not have an additive constant, forcing all the information about the pattern to emerge from the coefficients. We express the equation as: Work (0/100) = k1(Solution A1) + k2(Solution A2) + …. K16(Solution D4). Each respondent thus generates 16 coefficients, the ‘model’ for that respondent. The coefficient shows the number of points on a 100-point scale for ‘working’ contributed by each of the 16 solutions.
  8. Array all the coefficients in a data matrix, each row corresponding to a respondent, and each column corresponding to one of the 16 solutions or elements. The data matrix is very large, comprising approximately 50 rows per study, one per respondent, and 27 blocks of rows, one block per study, to generate 1350 rows. Each row is unique, corresponding to a respondent, study, and comprises information about the respondent (age, gender, answer to classificaiton questions), and then the 16 coefficients.
  9. Cluster all respondents, independent of the problem topic, but simply based on the pattern of the 16 coefficients for the respondent. The clustering is called k-means [16]. The researcher has a choice of the measure of distance or dissimilarity. For these data we cluster using the so-called Pearson Model, where the distance between two respondents is based on the quantity (1-Pearson Correlation Coefficient computed across the 16 corresponding pairs of coefficients). Note that the clustering program ‘does not know’ that there are 27 studies. The structure of the data is the same from one study to another, from one respondent to another
  10. Each respondent is assigned to exactly one of the three large clusters (now called mind-sets), independent of WHO the person ‘is’, and the study in which the respondent participated. That is, the clustering program considers only the pattern of the coefficient. As a consequence, each of the three clusters can end up comprising respondents from each of the 27 studies. Finally, a respondent can be assigned to only one of the three clusters or mind-sets.
  11. Once the respondent is assigned to exactly one of the three mind-sets by the clustering program, the original raw data (24 rows of data for each respondent in each study) can now be augmented by an additional variable, namely the cluster membership of each respondent. The original raw data can be reanalyzed, first by total panel, then by mind-set, and then by mindset x study. With three mind-sets, there are now one grand equation with all the data, 27 equations for the 27 studies, and 81 equations for the 27 studies x 3 mind-sets.
  12. The analysis as outlined in Step 11 can be further strengthened by considering only those vignettes not rate ‘3’. Recall that ‘3’ corresponds to ‘cannot answer the question’. Eliminating all with ratings of ‘3’ eliminates these uncertain answers.
  13. The final data analytic step looks at the pattern of coefficients for the different groups (total, three mind-sets), considering the matrix of 16 elements (the solutions) x 27 studies. We will look only at strong performing elements, rather than trying to cope with a ‘wall of numbers’. For total panel, ‘strong’ is operationally defined as a coefficient of 25 or higher. For subgroups defined by the mind-sets, ‘strong’ is operationally defined as a coefficient of 30 or higher. These stringent criteria correspond to coefficients which are ‘statistically significant’ (P<0.05) through analysis of variance for OLS regression. All other coefficients will not be shown, in order to let the patterns emerge.
  14. The goal of the analysis is to get a sense of ‘what works’ for problems, solutions, and mind-sets. As we will see, most solutions fail to work for most problems. It is not that the solution is consciously thought to not work, but rather when the solution (an element) is combined with other elements, the patterns emerging suggest that the specific solution is simply irrelevant. As we will see, however, many solutions do work.
  15. The effort for one database, for one country, easy easily multiplied, either to the same database for different countries, or different topic databases for the country. From the point of view of cost, each database of 27 studies and 50 respondents per study can be created for $10,000 – $15,000, assuming that the respondents are easy to locate. That effort comes to about $400 – $500 per study. The time to create the database is equally impressive, days and weeks, not years.

Table 1: The 27 social problems. Each social problem was not further defined

table 1

Table 2: The 16 solutions and their abbreviation in the data tables

table 2

 
 

fig 1a & 1b

Figure 1a and 1b: Screen shots of the set up for one study (abortion rights)

Results for Total Panel and Three Emergent Mind-sets

Let us now look at the data from the total panel. In its full form, Table 3 would show 16 columns ( one per each of the 16 solution or elements), and 27 rows (one row for each of the 27 problems). Recall from #13 above that the strong performing combinations of problem (row) and solution (column) are those with coefficients of +20 or higher. The strong performing combination correspond to significant likelihood of the solution solving the problem, across all respondents, but excluding those vignettes assigned a rating of ‘3’ (cannot decide).

Table 3: Summary table of coefficients for coefficients emerging from the model relating presence/absence of 16 solutions (column) to the expected ability to solve the specific problem (row). Models were estimated after excluding all vignettes assigned the rating 3 (cannot decide). Only strong performing elements are shown (viz., coefficient of 25 or higher)

table 3

Only 20 of the possible 432 problem/solutions are perceived as likely to ‘work’. The strongest performing solutions come from business. The strongest performing problem is parenting. The rest of the combinations which ‘work’ are scattered. Finally, five of the 16 solutions never work with any problem, and 15 of the 27 problems are not amenable to any solution.

One of the key features of Mind Genomics is the search for mind-sets. The notion of mind-sets is that for each topic area, one can discovered different patterns of ‘weights’ applied by the respondent to the information. For example, when it comes to purchasing a product, one pattern of weights suggests that the respondent pays attention to product features, whereas another pattern of weights applied to the same elements suggests that the respondent pays attention to the experience of consuming the product, or the health benefits of the product, rather than paying attention to the features.

Our analysis proceeds by looking for ‘general’ mind-sets, across all 27 problems, and all 16 solutions. The coefficients for the three emergent mind-sets appear in Tables 4-6. Once again the only coefficients which appear in the tables are those coefficients deemed to be ‘very strong’ performers, this time with a value of +30 or higher. This increased stringency removes many coefficients. Yet, a casual inspection of Tables 4-6 shows that each table comprises more problems, more solutions, and more coefficients. The mind-sets do not believe that the key solutions will work everywhere, but just in some areas, in distinctly different areas, in fact. The mind-sets do not line up in an orderly fashion. That is, we do not have a simplistic set of psychophysical functions for the inner psychophysics. We do have patterns, and metrics for the social consensus.

  1. Mind-Set 1 (Table 4) appears to feel that business and education solutions will work more effectively than will solutions offered by government. Mind-Set 1 does not believe strongly in the public sector is able to provide workable solutions to many problems. Mind-Set 1 shows 46 problem/solution combinations of 30 or higher, and three problems/solutions combination with coefficients of 40 or higher. The 46 combinations are more than twice as many as the 20 combinations for strong performing elements from the Total Panel, even with the increased stringency applied to the mind-sets.
  2. Mind-Set 2 (Table 5) appears to feel that education and the law will solve many of the problems. Mind-Set 2 shows 50 problem/solution combinations of coefficient 30 or higher, and three combinations which show a coefficient of 40 or higher,
  3. Mind-Set 3 (Table 6) appears to feel that law and business will solve many of the problems. Mind-Set 3 shows 50 problem/solution combinations of coefficient 30 or higher, and five combinations which show a coefficient of 40 or higher,
  4. The increased richness of Tables 4-6 arises from the fact that the clustering isolates groups of individuals who think alike at the granular level of specific problems. By separating the mind-sets, the clustering program ensures that the individual coefficients have a less likely chance to cancel each other. We attribute the increased range to the hypothesis that people may be fundamentally different in their mental criteria. Inner Psychophysics reveals those differences, in a way that could not have been done before.

Table 4: Summary table of coefficients for model relating presence/absence of 16 solutions (column)to the expected ability to solve the specific problem (row). The data come from Mind-Set 1, which appears to focus on business and education, respectively, as the preferred solution to problems

table 4

Table 5: Summary table of coefficients for model relating presence/absence of 16 solutions (column) to the expected ability to solve the specific problem (row). The data come from Mind-Set 2, which appears to focus on education and the law, respectively, as the preferred solution to problems

table 5

Table 6: Summary table of coefficients for model relating presence/absence of 16 solutions (column)to the expected ability to solve the specific problem (row). The data come from Mind-Set 3, which appears to focus on law and business, respectively, as the preferred solution to problems

table 6

The Inner Psychophysics and Response Time

Response time is assumed to reflect processes which occur. Longer responses times are presumed to suggest the involvement of more processes. So attractive is the study of response time as an indicator of internal processes that response time has moved from a simply a non-cognitive measure in behavior to a world of its own. Responses times are presented, along with hypotheses of what might be occurring [17]. Indeed, an entire division of applied consumer researcher has emerged to test ideas, the field being called implicit researcher after the work of Harvard psychologist Mazharin Banaji and her associates [18].

Let us take the same approach as above, relating the presence/absence of the 16 elements, not however to ratings of ability to solve the problem, but rather to the response time. The Mind Genomics program measured the number of seconds between the appearance of the vignette and the response assigned. When the respondent ‘dawdled’ in the self-pace experiment, the response time became unnecessarily long, for reasons other than reading and reacting. An operationally defined limit of six seconds was assumed for a participant. All vignettes with response times of nine seconds or longer were eliminated from analysis, as were all vignettes assigned the rating ‘3’ (cannot decide).

One might argue that by selecting data with responses times of 9 seconds or less, one is deliberately reducing the discrimination power of the analysis, by eliminating vignettes which required deliberation. This is correct removed a number of suspiciously long response times.

The Mind Genomics program then estimates the response time for each element (viz., solution) by using OLS (ordinary least-squares) regression. The equation is: Response Time = k1(A1) + k2(A2)…k16(D4). The equation is the same as the equations above for problem solving, other than the fact that there is no additive constant. The rationale for the absence of an additive constant is that the response time should be ‘0’ in the absence of any elements.

Table 7 shows the average coefficients for response times for three problems across 16 solutions. These problems are college expenses, COVID vaccination, and police cruelty. The average coefficients are shown by Total Panel, and then by three mind-sets. ‘Long’ response times (viz, high coefficients) of 2.0 seconds for an element (viz., solution) are shown by shaded cells. To allow the patterns to emerge, Table 7 presents only those coefficients which are 1.0 (seconds) or more.

Table 7: Estimated response times for specific elements, for the total panel and for the respondents in a defined mind-set. Only those response times for vignettes rating 1, 2, 4 or 5, were used in the computation. Only response times 9 seconds or shorter were used in the computation, under the assumption that longer response times meant that the respondent was multi-tasking

table 7

The pattern is obvious at the most general level…. People think about solutions when confronted with the topic of paying college expenses. People ponder the offered solutions. In contrast, there are fewer long response times for COVID vaccinations, and very few for Police Cruelty. In other words, it’s not only the solution, but rather the unique combination of problem and solution. We have here evidence of how the topic ‘controls’ attention.

Based upon the array of response times for elements shown in Table 7, we are left with the Herculean task of discovering an interpreting a coherent pattern, for Total Panel and then for mind-set. The pattern is, paradoxically, a lack of a pattern across mind-sets. That is, respondents may differ in what they believe will solve a problem, but difference in mind-sets does not manifest itself in the pattern of response times.

It is important to realize that the response times do not necessarily mean right or wrong, agree or disagree, and so forth. When confronted with data about Mind Genomics and its measurement of response time, the novice in Mind Genomics often asks whether a short response time (or conversely a long response time) is means that the person likes the topic, dislikes the topic, and so forth. We are so accustomed to judgments of dislike/like, bad/good, etc., that it is difficult to accept the fact that the response time (or other such metric, such as pupil size or galvanic skin response, GSR), are simply measures without any inherent meaning., viz., cognitively ‘poor.’ It is we who search for the meaning, wanting to contextualize observations of human non-conscious responses as clue to judgment, such as the extremely popular notion [19] patterns of thinking; System 1 (Fast) and System 2 (Slow Deliberate).

Discussion and Conclusion

The early work in psychophysics focused on measurement, the assignment of numbers to perceptions. The search for lawful relations between these measured intensities of sensation and a physical correlate would come to the fore even during the early days of psychophysics, in the 1860’s, with founder [20]. It was Fechner who would trumpet the logarithmic ‘law of perception, viz., that the relation between physical stimuli and perceived intensity was a logarithmic relation. One consequence of that effort to seek regularity in nature using one’s measures was to focus on the relationships between external stimuli and internal perceptions. This ‘external psychophysics’ focused on the search for lawful relationships that could be expressed by simple equations. The effort would be continued and brought to far more depth and application by Harvard professor S.S. Stevens, known as the father of modern psychophysics.

This paper began with the desire to extend psychophysics to the measurement of internal ideas The contribution of this paper is the introduction of a simple method for presenting stimuli, doing so in a way which forces the respondent to act as a measuring instrument, prevents biases, and emerges with numbers representing metrics of the mind. There are undoubtedly improvement that can be had, but the key aspects of the objective to ‘measure ideas’ (viz., the ‘inner psychophysics).

If we were to summarize the effort, we would point out these features:

A. The notion of isolating a variable and studying it in depth simply does not work when the nature of people is to think about ideas which are compound and complex. Traditional psychophysical methods simply are too unrealistic in view of the fact that the researcher cannot control the stimulus, the mind.

B. True to the word ‘psycho-physics’ which links two realms, the stimuli must be controlled by the researcher, and capable of systematic variation. If not, we are not true to the vision of psychophysics, linking two domains. The approach presented here, evaluation of systematically created combinations of stimuli, is consistent with the methods espoused by psychophysics.

C. The response should be a metric of ‘intensity’. The Likert scale presented here is such a scale. For science, the Likert scale data suffice. For application, most people have trouble interpreting the ‘implication’ of values on the Likert scale. That is, the scale is adequate, but most managers don’t really know the ‘practical meaning’ of the scale values. The transformation of Likert values to a binary scale ensures that the user of the information can make sense of the data.

D. The same type of study can be used to assess the impact of a set of ideas evaluated against different contexts. In the world of psychophysics, this type of study reveals the influence of different ‘backgrounds’ to affect the response to the stimulus (e.g., the perception of various coffees when the amount of milk is systematically varied). The psychophysical ‘thinking’ re-emerges when the topic or general problem is systematically varied across the 27 experiments, and then the study executed with new respondents. The outcome generates parallel sets of measures for ideas, each set pertaining to a specific topic, but everything else remaining the same.

E. Psychophysicists often look for explainable differences in the pattern of reactions to stimuli, more so in the chemical senses than in other areas, perhaps because in the chemical senses it is well known that people differ in what they like. This notion of basic groups, mind-sets, developed for simple stimuli in psychophysics, transfers straightforward to studies in Mind Genomics. Using the coefficients emerging from the OLS deconstruction of the rating at the level of the individual, one can find these ‘mind-sets’ for any specific topic, or as in this people, search for mind-sets which transcend the particular problem. Ability to cluster the respondents en masse, to create groups of individuals who show similar patterns of coefficients, viz., similar ways of thinking about solutions to problems.

Ability to measure the response time and determine whether we can discover any relation between response time as a well-known variable in psychology, and importance in decision making. The data emerging from this study suggest that the use of response times will not be particularly productive, except as a general measure. That is, we learn a great deal from the pattern relating ratings (more correctly transformed ratings) to the solutions. We learn a lot less by using response time in place of ratings. It may be that these relations exist, but are covered up by the richness of data, so that the basic patterns between text and response times are too subtle to be revealed in a study of this type.

References

  1. Stevens SS, Greenbaum HB (1966) Regression effect in psychophysical judgment. Perception & Psychophysics 1: 439-446.
  2. Stevens SS (1975) Psychophysics: Introduction to its Perceptual, Neural, and Social Prospects, Psychophysics, New York, John Wiley.
  3. Stevens SS (1966) A metric for the social consensus. Science 151: 530-541. [crossref]
  4. Boring EG (1942) Sensation and Perception in the History of Experimental Psychology. Appleton-Century.
  5. Galanter, E (1962) The direct measurement of utility and subjective probability. The American Journal of Psychology 75: 208-220. [crossref]
  6. Miller GA (1964) Mathematics and Psychology, John Wiley, New York.
  7. Luce, R. D., Bush, R. R., Galanter, E. (Eds.) (1963) Handbook of Mathematical Psychology: Volume John Wiley
  8. Luce RD, Tukey JW (1964) Simultaneous conjoint measurement: A new type of fundamental measurement. Journal of Mathematical Psychology 1: 1-27.
  9. Anderson NH (1976) How functional measurement can yield validated interval scales of mental quantities. Journal of Applied Psychology 61-677.
  10. Green PE, Srinivasan, V (1978) Conjoint analysis in consumer research: issues and outlook. Journal of Consumer Research 5: 103-123.
  11. Green, Paul E, Wind Y (1975) New Way to Measure Consumers’ Judgments. Harvard Business Review 53: 107-117.
  12. Wind, Y (1978) Issues and advances in segmentation research. Journal of Marketing Research 15: 317-337.
  13. Moskowitz HR, Kluter RA, Westerling, J, Jacobs HL (1974) Sugar sweetness and pleasantness: Evidence for different psychological laws. Science 184: 583-585. [crossref]
  14. Goertz G, Mahoney J (2013) Methodological Rorschach tests: Contrasting interpretations in qualitative and quantitative research. Comparative Political Studies 46: 236-251.
  15. Gofman, A, Moskowitz, H. (2010) Isomorphic permuted experimental designs and their application in conjoint analysis. Journal of Sensory Studies 25: 127-145.
  16. Dubes, R, Jain AK (1980) Clustering methodologies in exploratory data analysis. Advances in Computers 19: 113-228.
  17. Walczyk JJ, Roper KS, Seemann, E, Humphrey AM (2003) Cognitive mechanisms underlying lying to questions: Response time as a cue to deception. Applied Cognitive Psychology: The Official Journal of the Society for Applied Research in Memory and Cognition 17: 755-774.
  18. Cunningham WA, Preacher KJ, Banaji MR (2001) Implicit attitude measures: Consistency, stability, and convergent validity. Psychological Science 12: 163-170. [crossref]
  19. Kahneman, D (2011) Thinking, Fast and Slow. Macmillan
  20. Fechner GT (1860) Elements of Psychophysics Leipzig, Germany.
fig 4

Hypothalmic Hamartoma – A Rare Cause of Central Precocious Puberty

DOI: 10.31038/JCRM.2022553

Abstract

Hypothalamic hamartoma is a well-known neurological rare cause of central precocious puberty and gelastic seizures and may be asymptomatic for long period. It is rare and non-progressive tumor like congenital malformation. Precocious puberty defined as children attained the puberty before age of 8 years in girls and 9 years in boys. We present such a case of precocious puberty due to hypothalamic hamartoma in 5 year old boy and its radiological imaging findings.

Keywords

Hamartoma, hypothalamic, precocious, puberty, central cause

Introduction

Hypothalamic hamartoma is a rare non progressive tumor like well-organized congenital malformation of tuber cinereum in the floor of third ventricle [1-4]. It causes the classical spectrum or trait of gelastic seizures, precocious puberty and developmental delay or behavioral disorder [1,3]. It is most usual neurological cause of central precocious puberty.2 This condition may be asymptomatic for long period, or may present with clinical appearance and symptoms of precocious puberty and complex partial seizures refectory to anticonvulsant drugs [1,2]. One third of patients with hypothalamic hamartoma can be present with precocious puberty and it is defined as children attained the puberty before age of 8 years in girls and 9 years in boys [1,4,5]. Furthermore precocious puberty is divided into gonadotropin releasing hormone (GnRH)-dependent/central and gonadotropin releasing hormone (GnRH)-independent / peripheral precocious puberty [4]. Contrast enhanced MRI is investigation of choice and play important role in diagnosis of hypothalamic hamartoma [2,3] It appears as well-defined non enhancing lesion in floor of third ventricle showing similar signal intensity to the grey matter of brain parenchyma [3,4,6]. Gold standard treatment option for isolated central precocious puberty due to hypothalamic hamartoma is long acting analogs agonists of GnRH (GnHas) with good efficacy and safety [4,7].

Case Report

5 Years old boy came in paediatric outpatient department of Liaquat National Hospital with complain of faster growth of child than expected according to his father. On further questioning child has also history of epileptic laughter (gelastic seizures) 1-2 times, 2-3 months back for which he has taken antiepileptic drugs from local doctor in periphery of Afghanistan.

On local examination there were axillary and pubic hair and enlargement of penis measured about 4-5 cm in length. This was classified as TANNER stage (sexual maturity rating) 04 (Figure 1). Neurological examination was normal with Glasgow coma scale (GCS) of 15/15. Electroencephalogram (EEG) was recommended to see the cause of previous history of epileptic seizures was negative. His baseline laboratory investigations turned out to be within normal limits. On the basis of clinical and examination diagnosis of precocious puberty was made. His endocrinology related laboratory investigations were deranged. Follicle Stimulating Hormone (FSH) 4.11 (N= 1.9 mIU/ml), Luteinizing Hormone (LH) 9.82 (N1.3 mIU/ml) Testosterone 1500 ng/dL and Testosterone/Estradiol T/E2 ratio 45 pg/ml. All these investigations are raised and not normally corresponding to the patient’s age. Thyroid Function Tests (TFT) were within the normal limit.

fig 1

Figure 1: Enlarged penis and appearance of pubic hair not corresponding to the patient’s actual age

He underwent the contrast enhanced MRI brain which showed abnormal signal intensity mass arising from the tuber cinereum in the region of hypothalamus. It appeared iso-intense to grey matter on T1 and T2 weighted images and showed no post contrast enhancement. It measured about 0.9 x 1.1 cm. The lesion was very small in size and has no mass effect or compression over the adjacent brain parenchyma. This was diagnosed as hypothalamic hamartoma. (Figures 2 and 3). Patient also underwent the X-ray right hand and elbow according to the radiological protocol to see the exact age of patient which showed 15 years reported by the experienced radiologist (Figure 4). During hospital stay course patient was asymptomatic and discharged on long acting analogs agonists of GnRH (GnHas). Follow-up was recommended in outpatient department of endocrinology.

fig 2

Figure 2: A Axial T2 weighted image and B sagittal T2 weighted image show abnormal signal intensity mass arising from the tuber cinereum in the region of hypothalamus. It appears iso-intense to the brain parenchyma (black arrow head)

fig 3

Figure 3: A sagittal plain T1 weighted image and B sagittal contrast enhanced T1 weighted image show abnormal signal intensity mass arising from the tuber cinereum in the region of hypothalamus. It appears iso-intense to brain parenchyma and show no post contrast enhancement (White arrow head)

fig 4

Figure 4: A anterior-posterior elbow, B lateral elbow and C anterior-posterior wrist X-rays show bone age of 15 years

Discussion

Previously Hypothalamic hamartoma used to be an uncommon finding and was estimated to occur in one person per one million population,1 however with timely advancement in the field of Radiology and better clinical recognition, the early diagnosis has now become possible and its incidence has significantly decreased [1,2].

Most patients usually present in the first or second decade of life [1-3]. Precocious puberty is the most common presentation, however larger hamartomas are less likely to produce precocious puberty [2,3]. Other clinical presentations include developmental delay, attention deficit or hyperactivity disorder and anxiety [1-4]. A very specific feature of hypothalamic hamartoma is Gelastic seizure [1-3]. In our case, hamartoma measured up to 1cm on MRI brain (Figure 2) and patient was presented with complaints of both precocious puberty and laughing fits (Gelastic seizures).

On MRI, hypothalamic hamartomas produce soft tissue intensity masses which are isointense to grey matter on T1WI and hyperintense on T2WI [2,3]. They are homogeneous and sharply marginated by the surrounding CSF with no post contrast enhancement [2,3,6]. Calcification is rare and hemorrhage is not described in these lesions [2,3]. This classical appearance of soft tissue intensity mass in the region of hypothalamus was also observed in our patient (Figure 2).

Patients with central precocious puberty are usually managed conservatively [2], for progressive central precocious puberty, treatment with a depot GnRH agonist is suggested and is generally continued for 11 years [2-4]. However, surgery including resection or disconnection through craniotomy or trans-spheroidal approach can be helpful in cases of poorly controlled epilepsy [2-6], to our patient, we advised long acting analogs agonists of GnRH (GnHas) with a follow up in OPD.

Conclusion

Hypothalamic hamartomas are rare congenital malformation seen in floor of third ventricle. Precocious puberty is most usual presenting feature. MRI brain play important role in diagnosis of these lesions. Radiologist should aware of imaging findings of hypothalamic hamartoma and should examine the hypothalamic region properly in suspected cases as they can be easily missed due to small size. Bone age can be assessed by using the wrist X-ray and we can also observe the slowing of bone growth after treatment by X-ray wrist recommendation in every 6th month.

References

  1. Qasim BA, Mohammed AA (2020) Hamartoma of hypothalamus presented as precocious puberty and epilepsy in a 10-year-old girl. International Journal of Surgery Case Reports 77: 170-173. [crossref]
  2. Mahmood R, Al Taei TH, Samah Al Obaidi MD (2019) Hamartoma of the Hypothalamus. Bahrain Medical Bulletin 41.
  3. Kalekar T (2015) Hypothalamic Hamartomas: Two Cases. Open Journal of Medical Imaging 5: 20.
  4. Sharma P, Acharya N, Guleria TC (2020) Hypothalamic hamartoma presenting as central precocious puberty: a rare case report. International Journal of Contemporary Pediatrics 7: 1634.
  5. Mahajan ZA, Mehta SR (2020) Central precocious puberty: A case report. Medical Journal of Dr. DY Patil Vidyapeeth 13: 413.
  6. Boyko OB, Curnes JT, Oakes WJ, Burger PC (1991) Hamartomas of the tuber cinereum: CT, MR, and pathologic findings. American Journal of Neuroradiology 12: 309-314. [crossref]
  7. Eugster EA (2019) Treatment of central precocious puberty. Journal of the Endocrine Society 3: 965-972. [crossref]
fig 5

Arizona Reopening Phase 3 and COVID-19: After 18 Months

DOI: 10.31038/JCRM.2022544

Abstract

There had been three Arizona COVID-19 Reopening Phases. On March 5, 2021, Arizona’s Reopening Phase 3 began. The state is the sixth largest in size of the United States 50 states and about the same size as Italy. There were four case surges — in the summer and fall 2021 with Delta variant, and the winter 2021-22 and summer 2022 with Omicron variants. This 18 months longitudinal study examined changes in the number of new COVID-19 cases, hospitalized cases, deaths, and vaccinations. There was an increase of more than 1.4 million cases during the study period. The data source used was from the Arizona Department of Health Services COVID-19 dashboard database. Even with the case surges, the new normal was low number of severe cases, manageable hospitalization numbers, and low number of deaths.

Keywords

COVID-19, Arizona returning to normal, Longitudinal study, Arizona and COVID-19

Introduction

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also known as COVID-19 (coronavirus). It is a respiratory disease (attacks primarily the lungs) that spreads by person to person through respiratory droplets (coughs, sneezes, and talks) and contaminated surfaces or objects. Since the virus first appears in Wuhan, China in December 2019, there has been more than 600 million cases in the world. On September 7, 2022, Johns Hopkins University [1] reports that there are 606,889,445 total COVID-19 cases and 6,507,958 deaths associated with the virus in the world. The United States has the highest total cases (95,020,855) and deaths (1,048,989) in the world [1].

A three prolong attack is used against the virus by encouraging the public to practice preventive health behaviors that reduces the risks of getting respiratory infections (e.g., coronavirus, flu, and cold), and using vaccines and therapeutics. The preventive health behaviors include, but not limited to, practicing physical and social distancing, washing hands frequently and thoroughly, and wearing face masks. Johns Hopkins reports that more than 12.18 billion vaccine doses have been administered in the world and the U.S. has administered more than 605 million vaccine doses (September 7, 2022) [1].

There has been three Arizona Reopening Phases. During Arizona’s Reopening Phase 2 winter surge in 2020, ABC and NBC News report that the state has the highest new cases per capital in the world [2,3]. On September 7, Arizona is ranked 12th in total COVID-19 cases (2,258,040) and 11th in total deaths (31,162) of the 50 U.S. states [1]. Arizona is the sixth largest in size (113,990 square miles / 295,233 square kilometers) of the U.S. 50 states and is about the same size as Italy (301,340 square kilometer) [4,5]. The state population estimate is 7,276,316 on July 1, 2021 [6].

A partnership between the U.S. federal government and each of the 50 states is required to address the COVID-19 pandemic [7,8]. The federal government provides the national guidance primarily through the Centers for Disease Control and Prevention (CDC) and needed logistical support (e.g., provide federal supplemental funding, needed medical personnel and resources, and other needed assistance). The states decide on what actions to take and when to carry out those actions; the state COVID-19 restrictions; when to carry out each reopening phase; and the state vaccination plan.

On March 5, 2021, Arizona Governor Douglas Ducey begin Reopening Phase 3 (final reopening phase) after the state had administered more than two million vaccine doses and several weeks of declining cases [9,10]. This eases more of the COVID-19 restrictions. As more people become vaccinated and those infected recovered and have immunity against the virus; the numbers of cases, hospitalizations, and deaths will be low; COVID-19 will be manageable; and the state returns to normal.

The remainder of the paper examines Arizona Reopening Phase 3 (March 5, 2021 to September 7, 2022) looking at changes in the number of new COVID-19 cases, hospitalizations, and deaths.

Methods

This was an 18-months longitudinal study. The Arizona Department of Health Services (the state health department) COVID-19 dashboard database was the data source used. The study examined the changes in the numbers of new COVID-19 cases, hospitalized cases, deaths, and vaccines administered.

There were several data limitations. The COVID-19 case numbers represented the numbers of positive tests reported. When more than one test given to the same person (e.g., during hospitalization, at work, and mandatory testing), there were individual case duplications. Aggressive testing resulted in increases in false positive and false negative testing results. The case numbers did not include positive home testing results.

Delays in the data submitted to the state health department affected the timeliness of data reported and caused fluctuations in the number of cases, hospitalizations, deaths, and vaccinations. The state health department continued to adjust the reported numbers that may take more than a month to correct the numbers. The deaths associated with the coronavirus may cause by more than one serious underlying medical conditions, and the virus may not be the primary cause of death.

Results

A case could be mild (no symptoms), moderate (sick, but can recover at home), and severe (require hospitalization and/or result in death). There were four case surges during the Reopening Phase 3: 2 summers, 1 fall, and 1 winter. The 2022 cases (882,671) had already exceeded the 2021 total case numbers (838,836). Figure 1 shows the Arizona weekly COVID-19 cases during January 1, 2020 to September 10, 2022.

fig 1

Figure 1: Arizona Weekly COVID-19 Cases: January 1, 2020 to September 10, 2022.
Source: Arizona Department of Health Services Arizona COVID-19 weekly Cases Graph*2022 cases as of September 14.

At the end of the 18 months of Arizona Reopening Phase (began March, 5, 2021), there were 1,432,921 COVID-19 cases, 59,091 case hospitalizations, and 14,839 deaths associated with the virus in Arizona (Table 1). There were higher percentages of hospitalizations, and deaths in the first 6 months of the first year than the following two 6-month periods.

Table 1: Arizona Reopening Phase 3 Total Numbers of COVID-19 Cases, Hospitalizations, and Deaths: March 7, 2021 to September 7, 2022

table 1

Source: Arizona Department of Health Services COVID-19 Dashboard.
Arizona 2021 population estimate is 7,276,316, July 1, 2021 – U.S. Census.

Table 2 tracks the weekly total and weekly numbers of COVID-19 cases, hospitalized cases, and deaths during the past 6 months (March 9 through September 7, 2022). The largest weekly numbers of cases (20,198) occurred on July 6, while hospitalizations (1,955) occurred on March 9. The largest weekly number of deaths was on March 16 (457).

Table 2: Arizona Total and Weekly Numbers of COVID-19 Cases, Hospitalizations, and Deaths

table 2

Source: Arizona Department of Health Services COVID-19 Dashboard.
Arizona 2021 population estimate is 7,285,370, July 1, 2021 – Arizona OEO.

Figures 2-4 compare the numbers of COVID-19 cases, hospitalized cases, and deaths by age groups for the three 6-month periods. A case could be mild, moderate, and severe. Most people recovered and did not require hospitalization. There was an increase of 1,432,921 cases during the 18 months. The 20-44 years age group had the largest number of cases (Figure 2). There were more females (52.8%) than males (47.2%) who got the virus on September 7, 2022.

fig 2

Figure 2: Arizona Reopening Phase 3 COVID-19 Cases by Age Groups for Three 6-Month Periods.
Source: Arizona Department of Health Services COVID-19 Cases by Age Groups Statistics.

The percentages of total hospitalized cases (severe cases) decreased from 7 percent on March 6, 2021 to 5 percent on September 7, 2022. The case hospitalizations had increased by 59,091 during the study period. Seniors had the highest percent of the total hospitalizations (43.7% on September 7) and those under 20 years of age had the lowest percent (4.5%). Eighteen percent (18.1%) of seniors diagnosed with COVID-19 hospitalized, while 1.1 percent of those under 20 years of age hospitalized. There were more males (52.2%) than females (47.8%) hospitalized. Figure 3 shows the hospitalization numbers for each age group with the virus for the three 6-month periods.

The numbers of deaths had increased by 14,839 during the 18 months. The rates of fatalities per 100,000 population increased 227.05 to 433.50. As expected, seniors had the highest percent of total deaths (71.3% on September 7) and those under 20 years of age had the lowest percent (0.2) Eight percent (7.9%) of the seniors diagnosed with COVID-19 died, while 0.01 percent of those under 20 years of age died. There were more males (59%) than females (41%) who died. Figure 4 shows the numbers of deaths for each age group with the virus for the three 6-month periods.

The first U.S. COVID-19 vaccine, Pfizer/BioNTech Comirnaty, approved for emergency use authorization on December 11, 2020. In late December, Arizona began to administer vaccines. During Reopening Phase 3 (March 5, 2021 to September 7, 2022), there were 10,451,502 vaccine doses administered, and 3,813,974 fully vaccinated against the virus. Figure 5 shows the numbers of COVID-19 vaccines that were given in Arizona (persons fully vaccinated, persons receiving at least one dose, and total doses given) during the 18 months.

Initially, there were three vaccines available (Pfizer/BioNTech Comirnaty, Moderna Spikevax, and Johnson&Johnson Jcovden). Novavax Nuvaxivud became the fourth vaccine available in July 2022. The vaccines provided different levels of protection against COVID-19 and its variants. Those 65 years and older had the highest vaccination percentage, while those under 20 years of age had the lowest (Figure 6). It was expected the vaccination rates for this age group will increase with the approval of younger children vaccines use.

fig 3

Figure 3: Arizona Reopening Phase 3 Hospitalized COVID-19 Cases by Age Groups for Three 6-Month Periods.
Source: Arizona Department of Health Services Hospitalized COVID-19 Cases by Age Groups Statistics.

fig 4

Figure 4: Arizona Reopening Phase 3 COVID-19 Deaths by Age Groups for Three 6-Month Periods.
Source: Arizona Department of Health Services COVID-19 Deaths by Age Groups Statistics.

fig 5

Figure 5: Arizona Reopening Phase 3 COVID-19 Vaccination Numbers: March 5, 2021 to September 7, 2022.
Source: Arizona Department of Health Services COVID-19 Vaccination Statistics.

fig 6

Figure 6: Arizona COVID-19 Vaccination Percentages (at least one dose) by Age Groups on September 7, 2022.
Source: Arizona Department of Health Services COVID-19 Vaccinations by Age Group Statistics.

Discussion

The Arizona Governor began Reopening Phase 3 (final phase of reopening) after the state had administered more than two million vaccine doses and several weeks of declining cases on March 5, 2021 [9,10]. The state continued its efforts to vaccinate its population. The number of vaccine dosages administered had increase from 2,016,512 on March 5, 2021 to 12,468,014 on September 7, 2022. Sixty-two percent (4,525,048) of the state population were fully vaccinated. The largest numbers of fully vaccinated persons occurred in the week of April 17 to 23, 2021 (249,755) [9,10]. The pace of vaccination began to slow down in June.

Arizona case numbers had decreased in the spring and early summer 2021. At the end of June, the Arizona State Legislature and Governor had rescinded many of the state COVID-19 restrictions. The state used a three-pronged attack against the virus: (1) encourage preventive health behaviors, (2) increase vaccination numbers, and (3) use therapeutics. During the month of July, the highly contagious Delta variant appeared in the state and began the summer surge. Even with the increase vaccination efforts and other actions, they were not enough to stop the Delta variant. This resulted in the fall surge.

In December, the more contagious Omicron variant appeared in the state and began to surge. The Omicron variant surge in January 2022, and the cases remained high into early March. For more than two months in the spring, the cases were low. The state cases rose at the end May as the Omicron variants moved westward in the U.S. and began the summer surge. By late August, the cases declined.

It has been more than 32 months since the first COVID-19 cases appeared in Arizona on January 22, 2020; the state has not returned to pre-pandemic normal of zero cases and no face mask wearing. Most health facilities require both medical staff and patients wear masks. Many businesses require their staff wear masks and masking wearing is optional for customers, and they still have their virus protective glass/plastic barriers. There are signs of the public experiencing COVID fatigue (e.g., significant numbers did not wear masks during summer 2022 case surge and did not pay attention to the daily/weekly number of case increases).

Many still have anxiety/depression/stress associated with the virus. The causes for the mental anguish are the uncertainty of the virus, constant emergent of new variants, vaccine limitations, the lack control of the situation, and no end to the virus. There are persons who have not adapt to the new normal and have limited their interactions with people.

Overtime, the vaccines are not as effective against the later variants (Delta and Omicron) and Omicron subvariants as the original Alpha – breakthrough infections and wane over time. Even though the vaccines and their boosters reduce the risks in getting a severe case, one can still can get the virus. There has been very little increase in the vaccination rate in the last six months — the number of fully vaccinated percent increased only by 2.9 percent.

The Food and Drug Administration has approved both Pfizer and Moderna new bivalent COVID-19 vaccines that are effective against both the original virus and Omicron BA.4 and BA.5 variants on August 31, 2022. With the new vaccines, it is expected the vaccination rates will rise; the population immunity level will be high enough to keep the winter case surge lower than last year; and the state will move the closer to returning back pre-pandemic normal.

Conclusion

The vaccines, most of the Omicron variant cases are mild or moderate, and therapeutics have kept the number of hospitalizations and deaths low. Even with the occasion case surges, the state new normal are low number of severe cases, manageable hospitalization numbers, and low number of deaths.

References

  1. Johns Hopkins University Coronavirus Resource Center, https://coronavirus.jhu.edu/.
  2. Deliso, Meredith. “Arizona ‘hottest hot spot’ for COVID-19 as health officials warn of hospital strain: The state has the highest infections per capita globally, based on JHU data, ABC News, January 7, 2021, https://abcnews.go.com/US/arizona-hottest-hot-spot-covid-19-health-officials/story?id=75062175.
  3. Chow, Denise and Joe Murphy. These three states have the worst Covid infection rates of anywhere in the world: Arizona currently has the highest per capita rate of new Covid-19 infections, with 785 cases per 100,000 people over the past seven days, followed closely by California and Rhode Island, NBC News, January 5, 2021 and updated on January 7, 2021, https://www.nbcnews.com/science/science-news/these-three-states-have-worst-covid-infection-rates-anywhere-world-n1252861.
  4. Britannica, Arizona state, United States, https://www.britannica.com/place/Arizona-state.
  5. My Life Elsewhere, Arizona is around the same size as Italy, https://www.mylifeelsewhere.com/country-size-comparison/arizona-usa/italy.
  6. United States Census Bureau, Quick Facts, https://www.census.gov/quickfacts/AZ.
  7. Eng H (2020) Arizona and COVID-19. Medical and Clinical Research 5: 175-178.
  8. Eng H (2021) Arizona Reopening Phase 2: Rise and Fall of COVID-19 Cases. Medical and Clinical Research 6: 114-118.
  9. Eng H (2021) Arizona Reopening Phase 3 and COVID-19: Returning to Normal. Medical and Clinical Research 6: 687-669.
  10. Eng H (2022) Arizona Reopening Phase 3 and COVID-19: One Year Later. Journal of Clinical Research and Medicine 5: 1-7.

Anxiety, Stress, Depression among School Going Adolescents in Bareilly City: A Cross Sectional Study

DOI: 10.31038/PSC.2022221

Abstract

Context: Depressive disorders often start at a young age. There is a need for early identification of depression, anxiety, and stress (DAS) and prevention. The present study was undertaken to find the magnitude of DAS among adolescents.

Aims: To find the mental health status of school going adolescents in Chandigarh. The objectives were (i) to study the prevalence of DAS among school going adolescents and (ii) to study the correlates of DAS. Settings and Design: A Cross‑sectional survey of students of four classes from 9th to 12th studying in government schools.

Subjects and Methods: Ten government schools in Bareilly City were randomly selected through lottery method. In each school, for each of the four classes, a section was randomly selected again by the lottery method. Forty students were selected from each school reaching sample size of 470. DAS scale 21 questionnaires were used.

Statistical Analysis Used: The data entry was done in MS Office Excel 2007. The analysis was done in the form of frequency tables, charts cross tables. For statistical significance, Chi‑square test and correlation was found between various factors.

Results: The prevalence of DAS was 65.53%, 80.85%, and 47.02%, respectively. Overall, comorbidity between depression and anxiety was 57.65%. Extremely severe depression was very less (3%). The prevalence of DAS was higher in females. For depression and anxiety, the peak age was 18 years. Conclusions: The prevalence of DAS was high among school going adolescents in Bareilly City. There is a need for early and effective identification of DAS that can prevent many psychiatric disorders at their nascent stage.

Keywords

Adolescent, Depression, Anxiety, Stress, Bareilly City

Introduction

Mental illnesses account for a significant share of the disease burden in all civilizations. Depression, anxiety, and stress are among the most common causes of illness and disability in children. Teenagers are predisposed to a range of mental health difficulties due to the physical, psychological, and behavioral variations during this time. Poor academic performance, lack of communication with friends and family members, substance misuse, feelings of abandonment, homicidal thoughts, and suicide ideation are all signs of these three diseases [1,2].

For at least two weeks, depression is defined by persistent unhappiness and a loss of interest in activities that you generally love, as well as incapacity to function in everyday life. Anxiety is a sensation of tension accompanied by concerned thoughts and bodily changes such as elevated blood pressure. Anxiety disorders are characterized by recurrent intrusive thoughts or concerns.

Nearly 20% of children suffer from a diagnosable mental illness. In addition, numerous mental health illnesses emerge during adolescence. Before reaching adulthood, between 20-30% of children will experience at least one significant depressive episode. For a quarter of people, mood disorders such as depression first appear throughout adolescence. Anxiety disorders and impulse control disorders (such as conduct disorder or attention deficit/hyperactivity disorder) affect 50-75 percent of adolescents during their adolescence. As children enter puberty, existing mental health issues become more complex and acute. Adolescents with untreated mental health issues are more likely to perform poorly in school, drop out, have strained family connections, abuse substances, and engage in risky sexual practices [3].

Being away from home, grade, stream of study, academic performance and examination-related concerns, and cyber bullying have all been connected to depression in previous studies. According to earlier studies, sex, grade level of pupils, and kind of school (public or private), family type, not living with parents, educational level of parents, and high academic stress were all factors of anxiety.

Globally, depression is one of the leading causes of illness and disability. Even in developed nation’s depression is a known health burden among children, adolescents, and adults. One in four children in the age group of 13-15 years in India suffers from depression, which affects 86 million people in the South-East Asia region, the World Health Organization. In adolescents, major depression is projected to rank second-most cause of human illness by the year 2020 [4,5].

To battle the burden of juvenile mental health concerns, researchers must investigate the extent and risk factors of symptoms of depression, anxiety, and stress. Nevertheless, anxiety and depression in early teens commonly go undetected and untreated, particularly in developing countries like India, due to limited access to psychological and psychiatric care as well as the enormous societal stigma regarding mental health concerns. Keeping these points in mind, an attempt will be made to assess the depression, anxiety and stress among school students in Bareilly City, Uttar Pradesh.

Materials and Methods

The A cross sectional research was carried out among school students in Bareilly city from February 2022 – April 2022. The study was approved by the Institutional Review Board, Institute of Dental Sciences, Bareilly. Prior beginning the research, the production lines head provided written approval for it to be carried out there. Sample size has been scientifically estimated using G Power V 3.1 Software which yielded a minimum sample size of 470 School students. School students who were aged 10-19 years and whose parents will give consent to allow their children to participate were included in the study. School going children whose parents are not willing to give consent and who are likely to take transfer during the study period and those who did not want to take part in this research were eliminated. Multistage sampling technique was used for all the study units until the required sample size is attained.

The complete information about the study will be informed to the participants as mentioned in the participant information sheet. Signed informed consent form will be obtained from all the participating subjects after explaining the complete procedure in their vernacular language. A pre-designed semi-structured, self-administered questionnaire was used to assess socio-demographic profile like age, gender, religion etc. and associated factors like pressure to perform, relations with parents etc. Depression anxiety stress scale (DASS)-21 was used to detect depression, anxiety and stress.

Statistical Analysis

Data was entered on Microsoft excel software and statistical analysis was done using a licensed version of SPSS 21. Descriptive analysis was done by calculating proportions, means and standard deviation.

Results

Out of 470 students, the maximum number of students participating in study was from 9th class (28.72%), and minimum number of students was from 12th class (22.76%). There were 257 male (54.68%) and 213 female (45.31%) participants in the study. The maximum number of students were from the age group of 16 years, i.e., 180 (38.29%) and minimum from the age group of 19 years, i.e., 3 (0.63%) students. Table 1 shows gender‑wise distribution of participants having DAS. Table 2 shows that overall comorbidity between all three disorders, i.e., DAS was 36.1%. Distribution of participant and 12th class having the DAS is shown in Table 3. On comparison of DAS among participants of 9th, 10th, 11th, 12th classes, it can be seen that depression was higher in 12th class, anxiety was higher in 10th class, and stress was higher in 9th class. While comparing DAS among participant of nonboard (9th + 11th) and board classes (10th + 12th), it was found that it was higher in board classes than in nonboard classes. It was found that all types of depression (75.59%) and stress (53.52%) were higher in board classes than of nonboard classes (57.2% and 41.63%,respectively).Among students of classes 11th and 12th, according to their stream, it was found that depression and anxiety were maximum in medical students (78.57%), and stress was more in commerce students (48.89%). It was found that extremely severe depression was highest among medical students (03.57%); mild depression was also more in them (28.57%). Moderate depression was more in arts students (43.42%). Extremely severe (17.10%) and moderate anxiety (27.63%) were higher in arts students. Mild anxiety was higher in medical students (42.86%). Severe anxiety was higher in commerce students (16.67%). Extremely severe stress was present only in commerce students (01.11%); severe stress was higher in nonmedical students (60.71%). Mild stress was higher in arts students (35.53%).

Table 1: Gender wise distribution of participants having depression, anxiety, and stress (n=470)

Gender

Number of students

Prevalence of DAS (%)

Males

257

85 (33.07)

Females

213

85 (39.9)

DAS: Depression, anxiety and stress

Table 2: Comorbidity between different disorders (n=470)

Comorbidity

n (%)

Overall

170 (36.17)

Depression and anxiety

271 (57.65)

Depression and stress

144 (40)

Stress and anxiety

200 (50)

Table 3: Distribution of participants of 9th, 10th, 11th and 12th class having the depression, anxiety, and stress (n=470)

Students of class

DAS Normal (%) Mild (%) Moderate (%) Severe (%)

Extremely severe (%)

Class 9th (n=135) Depression

55 (40.74)

21 (15.56) 40 (29.63) 14 (10.37)

5 (3.7)

  Anxiety

37 (27.41)

31 (22.96) 52 (38.52) 14 (10.37)

1 (0.74)

  Stress

32 (23.7)

37 (27.41) 51 (37.78) 9 (06.67)

6 (4.44)

Class 10th (n=106) Depression

28 (26.42)

27 (25.47) 32 (30.19) 14 (13.21)

5 (4.72)

  Anxiety

18 (16.96)

21 (19.81) 39 (36.79) 15 (14.15)

13 (12.26)

  Stress

50 (47.17)

30 (28.30) 10 (09.43) 15 (14.15)

1 (0.94)

Class 11th (n=122) Depression

55 (45.08)

20 (16.39) 34 (27.87) 12 (09.84)

1 (0.82)

  Anxiety

27 (22.13)

36 (29.51) 26 (21.31) 21 (17.21)

12 (09.84)

  Stress

72 (59.02)

29 (23.77) 11 (09.02) 9 (07.38)

1 (0.82)

Class 12th (n=107) Depression

25 (23.30)

23 (21.36) 43 (40.78) 12 (11.65)

4 (02.91)

  Anxiety

21 (19.42)

29 (27.18) 25 (23.30) 12 (11.65)

20 (18.45)

  Stress

49 (45.63)

40 (37.86) 6 (05.83) 12 (10.68)

0

DAS: Depression, anxiety and stress            

Self-satisfaction with academic performances in participants with DAS was 67.08%, 86.07%, and 40.5%, respectively whereas the parent’s satisfaction with academic performances of their wards with DAS was 66.86%, 80.47%, and 43.19%, respectively. Poor socioeconomic conditions and father’s occupation (nonworking) were directly related with higher level of DAS. With increase in the education level of parents, level of DAS in their children decreased. As the parents love decreased, level of depression and stress in the participants increased. DAS was found to be more among students whose mothers were not alive. The level of anxiety was found higher in the participants belonging to the joint families. Students staying away from home in hostels and paying guest accommodations had higher levels of depression and stress. It was found that the prevalence of depression and stress was more in students who were bullied by batch mates. It was also found that the prevalence of DAS was more in students who felt overburdened with test schedules. The level of stress was higher among the participants who were not self‑satisfied with their academic performance and whose parents were not satisfied. Participants, who took alcohol and smoked, showed higher prevalence of DAS.

Discussion

In our study, we found that the prevalence of DAS was more in students who feel overburdened with test schedules. The level of stress was higher among the participants who were not self‑satisfied with their academic performance and whose parents not satisfied. Similar results have been reported by other studies, namely, Kaur and Sharma, Moreira and Furegato, Liu and Lu and Gray-Stanley et al. [6-12].

A study done by Deb et al. revealed that 63.5% of the higher secondary students in Kolkata experience academic stress, and the parental pressure for better academic performance was found to be mostly responsible for academic stress as reported by 66.0% of the students. It was found that the prevalence of DAS was higher in females than in males. The study by Kaur and Sharma in Chandigarh also found that girls were more academically frustrated than the boys. The study conducted in Bengaluru by Sharma and Kirmani found that girls had higher scores on beck depression inventory than boys. A comprehensive review of almost all general population studies conducted to date in the United [13,14].

States of America, Puerto Rico, Canada, France, Iceland, Taiwan, Korea, Germany, and Hong Kong reported that young women predominated over men in lifetime prevalence rates of major depression. In India, similar findings were obtained by Verma et al. Academic stress is a type of stress that arises due to academic factors such as heavy school schedule, unrealistic expectation and demands of parents and teachers, low academic performance, poor study habits, and not having enough time to deal with school’s multiple priorities. Academic stress is recognized as a risk factor for depression and suicidal behavior. The experience of school-related stress such as poor academic performance, negative feedback from parents and teachers about school work; daily hassles in the school environment, stressful life events, and negative affect states during school work were all leads to increase in depression. Poor academic grades generally predict high educational stress; the discrepancy between expected and actual grades may play a more important role in the development of psychological distress and other mental health problems [15-20].

In our study, DAS increases as the intake of alcohol increases. Higher DAS was found among those who drink alcohol and those who were occasional smokers. Severe depression and extremely severe stress were more in males as compared to females. A study done in Chennai in 1986 revealed that 23.25% had contemplated suicide earlier and that 91.9% of them were aged 30 years or less. A strong association of suicidal tendency with alcohol was reported in 10.42% of the sample. The suicide rate was more in males as compared to females. It might be due to the reason that males are not emotionally very strong as compared to females and shared less of their problems as compared to female.

The prevalence of DAS increases as the parents love decreases, lack of parental affection takes toil on mental peace of children. In a review done by Zgambo, et al. in 2012, it was seen that children and adolescents who live without parents exhibit higher levels of depressive symptoms than those who live with parents around them. Depression is decreased by higher levels of parental care and lower levels of parental indifference. Greenberger et al. stipulate that strong positive family relationships lessen the symptoms of depression. Many other factors, such as loss of loved ones, conflicts with parents, teachers, and peers, and significant physical diseases may have important effects on adolescent suicidality.In our study, as the level of parent’s education increases, the level of prevalence DAS among adolescents decreases. There is a direct relation between the parents’ mental health and their children’s health.

A cross-sectional study by Olfson et al. in 2003, on parental depression and child mental health reported that children of parents with depression were approximately twice as likely as children of parents without depression to have a variety of mental health problem. The prevalence of depression and stress was more among the participants who were bullied by their batch mates or seniors. According to the study conducted by Khawaja et al. in 2015 in Pakistan showed that physical abuse (P = 0.05), verbal abuse (P = 0.003), injury (P = 0.02), and bullying (P < 0.001) were significantly associated with psychological stress.

As the age increases, the prevalence of DAS was also found to be increasing. The peak of the prevalence of depression was in the 18th year of age. Tepper et al. argue that depressive symptoms do not differ between boys and girls but intensify with age. This trend of increasing DAS may be due to different social and developmental challenges faced by teens. In our study, depression and stress were prevalent in participants who belong to poor families. Direct and indirect effects of relative poverty had bad effect on the development of emotional, behavior, and psychiatric problems. Poverty has multidimensional phenomenon, encompassing inability to satisfy basic needs, lack of control over resources, lack of education and poor health [21-28].

Conclusion

According to study, the overall prevalence of DAS among school going adolescents in Chandigarh was high. DAS in this population have been shown to be associated with increased risk of suicidal behavior, homicidal ideation, tobacco use, and other substance use. The burden of mental disorder is great as they are prevalent in all societies. They create a substantial burden for affected individuals and their families and produce significant economic and social hardships that affect society as a whole.

References

  1. Alvi T, Assad F, Ramzan M, Khan FA (2020) Depression, anxiety and their associated factors among medical students. J Coll Physicians Surg Pak 20: 122-6. [crossref]
  2. Lopez AD, Murray CC (1998) The global burden of disease, Nat Med 4: 1241-3.
  3. Rask K, Astedt Kurki P, Laippala P (2002) Adolescent subjective wellbeing and realized values. J Adv Nurs 38: 254-63. [crossref]
  4. The Global Burden of Disease: Generating Evidence, Guiding Policy – European Union and Free Trade Association Regional Edition.
  5. The World Health Report 2001 – Mental Health: New Understanding, New Hope. WHO.
  6. Depression Available from: nimh.nih.gov/health/publications/depression/index.shtml.
  7. Williams CA (1989) Empathy and burnout in male and female helping professionals. Res Nurs Health 12: 169-178. [crossref]
  8. Farby JJ (1980) Depression. In: Woody RH, editor. Encyclopaedia of Clinical Assessment. San Francisco: Jossey-Bas.1980.
  9. Kaur S, Sharma V (2014) Depression among adolescents in relation to their academic stress. Indian J Appl Res 4: 183-5. [crossref]
  10. Moreira DP, Furegato AR (2012) Stress and depression among students of the last semester in two nursing courses. Rev Lat Am Enfermagem 21: 155-62. [crossref]
  11. Liu Y, Lu Z (2012) Chinese high school students’ academic stress and depressive symptoms: Gender and school climate as moderators. Stress Health 28: 340-6. [crossref]
  12. Gray Stanley JA, Muramatsu N, Heller T, Hughes S, Johnson TP, et al. (2010) Work stress and depression among direct support professionals: The role of work support and locus of control. J Intellect Disabil Res 54: 749-61.
  13. Deb S, Strodl E, Sun J (2015) Academic stress, parental pressure, anxiety and mental health among Indian high school students. Int J Psychol Behav Sci 5: 26-34.
  14. Sharma P, Kirmani MN (2015) Exploring depression & anxiety among college going students. Indian J Sci Res 4: 528-32.
  15. Gomez HF, Piccinelli M (1997) Division of Mental Health and Prevention of Substance Abuse. Gender Differences in the Epidemiology of Affective Disorders and Schizophrenia.
  16. Verma N, Jain M, Roy P (2014) Assessment of magnitude and grades of depression among adolescents in Raipur city, Int Res J Med Sci 2: 10-3.
  17. Banerjee S (2011) Effect of Various Counseling Strategies on Academic Stress of Secondary Level Students. Punjab University, Chandigarh, India.
  18. Ang RP, Huan VS (2006) Relationship between academic stress and suicidal ideation: Testing for depression as a mediator using multiple regression. Child Psychiatry Hum Dev 37: 133-43. [crossref]
  19. Bjorkman SM (2007) Relationships among Academic Stress, Social Support, and Internalizing and Externalizing Behavior in Adolescence. Northern Illinois University, U.S.A.
  20. Lin HC, Tang TC, Yen JY, Ko CH, Huang CF, et al. (2008) Depression and its association with self esteem, family, peer and school factors in a population of 9586 adolescents in Southern Taiwan. Psychiatry Clin Neurosci 62: 412-20. [crossref]
  21. Zgambo M, Kalembo F, Guoping H, Honghong W (2012) Depression among chinese children and adolescents: A review of the literature. Int J Child Youth Family Stud 3: 442-57. [crossref]
  22. ZhangY, Li H, Zou S (2011) Association between cognitive distortion, type D personality, family environment, and depression in Chinese adolescents. Depress Res Treat 143045. [crossref]
  23. Liu YL (2003) Parent child interaction and children’s depression: The relationships between parent child interaction and children’s depressive symptoms in Taiwan. J Adolesc 26: 447-57. [crossref]
  24. Greenberger E, Chen C, Tally SR, Qi D (2000) Family, peer, and individual correlates of depressive symptomatology among U.S. and Chinese adolescents. J Consult Clin Psychol 68: 209-19. [crossref]
  25. Liu X, Tein JY, Sandler IN, Zhao Z (2005) Psychopathology associated with suicide attempts among rural adolescents of China. Suicide Life Threat Behav 35: 265-76. [crossref]
  26. Olfson M, Marcus SC, Druss B, Alan Pincus H, Weissman MM (2003) Parental depression, child mental health problems, and health care utilization. Med Care 41: 716-21. [crossref]
  27. Khawaja S, Khoja AA, Motwani K (2005) Abuse among school going adolescents in three major cities of Pakistan: Is it associated with school performances and mood disorders? J Pak Med Assoc 65: 142-7. [crossref]
  28. Tepper P, Liu X, Guo C, Zhai J, Liu T, Li C (2008) Depressive symptoms in Chinese children and adolescents: Parent, teacher, and self reports. J Affect Disord 111: 291-8. [crossref]

What is the Status of Female Adolescent Depression in India and How is it Affecting the Modern Society

DOI: 10.31038/ASMHS.2022662

 

Adolescence is marked by dramatic developmental changes in physical, cognitive, and social-emotional capacities [1]. However, this is also a period which is beset by a number of challenges. For instance, engagement risk behaviours are more common among adolescents. Engagement in risk behaviours may pose a significant threat to health if involvement spans multiple behaviours. The asset model suggests that contextual aspects of young people’s lives, such as factors related to family, school and community, serve as a protective function against health risk behaviours [2]. Even though most adolescents are able to cope with such dramatic changes a large number of them encounter problems and difficulties caused by such changes. If they are unable to cope with stress caused by these changes, they may develop mental health problems, especially depression [3].

Moreover, abnormalities of social adjustment are detectable in childhood in some people who develop psychotic illness. Sex and the rate of development of different components of the capacity for social interaction are important determinants of the risk of psychosis and other psychiatric disorders in adulthood [4].

While the link between positive ageing and the perception of loneliness has been well-established [5], it is also well-known that psychological distress during adolescent period of the life span is common experience that may be due to the innumerable changes adolescents face [6]. As early as age 11, young adolescents begin to form their self-concept and must cope with increasing expectations from parents, peers, school, and society [7]. The intersection of these experiences, couple with other environmental stressors, can result in elevated distress, such as anxiety and depression [8]. These stressors are sometimes manifested in academic performance, such as reading abilities [9].

A study [10] defines depression in cognitive terms. It is based on the underlying theoretical assumption that the affection and behaviour of an individual are determined in great measure by the way an individual structures the world. His cognitions are based on attitudes and assumptions developed from previous experiences. The cognitive model states that there are specific concepts to explain the psychological substrata of depression: cognitive triad, schemas, and cognitive errors.

The cognitive triad consists of three main cognitive patterns: (a) patients view themselves negatively; (b) they interrupt their experiences negatively; and, (c) they have a negative view of the future. The second component of the cognitive model is the structural organisation of thought which Beck called schema. Schemas are relatively stable cognitive patterns that constitute model. [10] argue that a schematic interpretation always mediates between experience and emotional responses. A person’s negative and distorted cognition in a concrete situation are considered errors in the processing of the information, which are also called “automatic thoughts”. [11] offers a complementary perspective by proposing that adverse interactions with primary caregivers lead children to form negative internal working models of themselves and others. These insecurely attached individuals are likely to believe that they are unworthy of care and that others are unavailable and unpredictable.

[12] argued that cognitive distortion, that is the tendency to construe or distort the significance of events in a way that is consistent with a negative view of the self, the environment, and the future, all play a central role in the development and maintenance of depression. In one study it has been argued that depressive symptoms were differentially related to eating concerns and depressive symptoms may give clues as to which aspects of shame are important in each of the two types of pathology [13]. This emphasise the notion that cognitive distortions are likely to lead to depression in adolescents and the chances are further maximised if they have faulty relationships with their parents.

Adolescence is a period when fitting and connecting with others are highly valued; thus, interpersonal conflicts in close relationships can lead to even greater anxiety and depression levels [14]. Vast amounts of literature highlight adolescents’ need for a sense of belongingness and the importance it plays in their daily relationship. As per the hierarchy attachment model, adolescent–mother attachment outweighed adolescent–father attachment to some extent in predicting adolescents’ perceived social interrelationship measures. As per the integration attachment model, significant differences emerged on most social interrelationship measures between the 4 distinct subgroups: secure attachment to both parents, neither, only father, only mother [15]. Adolescence is a time of change, growth and all too often, struggle. It is a period where an individual could potentially have a notable experience of self-compassion [16].

[17] found that depressed youths were subject to harsher and less consistent parenting, as reported by both the child and the parent, compared to youths who were not depressed. Using data collected as part of the National Longitudinal Survey of Youth, it has been revealed that mother’s use of physical punishment predicted children’s depressive symptoms [18]. Also, Domestic violence negatively impacted children’s behaviour with their mothers in interactions but did not influence maternal report of problem behaviours, suggesting that the impact of domestic violence begins very early and in the realm of relationships rather than in mental health [19].

While prior work has theorised that certain populations may be at increased psychological vulnerability from intimate partner violence (IPV), recent findings indicate that both perpetration and victimisation are associated with increases in depressive symptoms for both men and women, and irrespective of whether IPV exposure occurred in adolescence or young adulthood. Cumulative exposure to IPV does not appear to increase depressive symptoms beyond the effect observed for the most recent IPV exposure, but physical maltreatment by a parent does appear to diminish the association between IPV perpetration and depressive symptoms for a small subset of the sample [20].

Interaction between parents and children are determining the quality of parent-child relationship. Negative interactions in a family can lead to blame game. Adolescents may blame their aggressive and depressive behaviours on their parents’ rejecting attitudes, and parents may excuse their rejecting attitudes on their children’s behaviours. But instead of blame, perhaps it is more a question of dysfunctional interactions that are self-perpetuating, negativity begetting negativity as it were.

Few studies have examined both maternal and paternal parenting practices in the prediction of child outcomes despite evidence that underscores the salience of fathers throughout their children’s development. This study examined the role of the quality of mother–child and father–child relationships in buffering the influence of ineffective parenting practices on subsequent adolescent aggression. Measures of parental psychological control, the quality of the parent–child relationship, and youth aggressive behavior were completed by 163 (49% female) mostly White and Asian adolescents and their parents during the eighth and ninth grades. Paternal psychological control predicted aggression when adolescents perceived low-quality relationships with their mothers. Similarly, maternal psychological control predicted aggression when adolescents perceived low-quality relationships with their fathers. Maternal psychological control was also associated with lower levels of aggression among adolescent males who reported a high-quality relationship with their father. These findings indicate that, when one parent exerts psychological control, the low-quality relationship the adolescent shares with the opposite gender parent increases risk for adolescent aggression. The findings also suggest that, as mothers exert psychological control, the high-quality parent–child relationship a son shares with his father decreases risk for adolescent aggression [21].

The Revelence of the Study

The study sought to identify the role of cognitive distortion and parental bonding in depressive symptoms among Fe male adolescents in rural India. The study also aims to ascertain the extent to which parent-child relationship, specifically father care and mother care; and, father overprotection and mother overprotection differ in the way they contribute to depressive symptoms of adolescents [22].

Materials and Methods

A total of 150Fe male adolescents aged 18-19 were drawn through random sampling. The educational institution was randomly selected from a list of higher educational institutions in India. The subject chosen for the study were also randomly selected from a class of 40-50 students.

All tests were administered in the group of 20-30 students. Stepwise multiple regression analysis was carried out to ascertain the contribution of cognitive distortion (self-criticism, self-blame, helplessness, hopelessness and preoccupation with danger); parent-child relationship (mother care, mother overprotection, father care, father overprotection) towards depressive symptoms.

Survey Instrument

Reynolds Adolescent Depression Scale (RADS-2) was developed by [23] to measure the severity of depressive symptoms in adolescents in clinical settings. The RADS-2 is a brief, 30-item self-report measure that includes subscales which evaluate the current level of an adolescent’s depressive symptoms along four basic dimensions of depression: (1) dysphoric mood; (2) anhedonia; (3) negative self-evaluation; and, (4) somatic complaints. In addition to the four subscale scores, the RADS-2 yields a depression total score that represents the overall severity of depressive symptoms. The reliability and validity of the test is well-established with internal consistency of 0.86, test-retest of 0.80, and validity criterion of 0.83.

Cognitive Distortion Scales (CDS) was developed by [23]. It measures distorted or negative cognitions and consists of 40 items. Each symptom item is rated according to its frequency of occurrence over the preceding month; using a five-point scale range from never to very often. The five subscales are self-criticism, self-blame, helplessness, hopelessness, and preoccupation with danger. The score on each dimension can be added to 9, which is the total score. The reliability and validity of the test is well-established, with reliability of 0.89 and validity of 0.94.

Parental Bonding Instrument (PBI) was developed by [24]. PBI is a 25-item instrument designed to assess the children’s perception to parent-child relationship in terms of parental behaviours and attitudes. The authors identified two variables that are important in developing parent-child bonding: (1) care and, (2) overprotection. Out of 25 items, 12 items measure children’s perception of their parents as caring with the opposite end of the spectrum being indifference or rejection, the remaining 13 items assess children’s overprotectiveness with the extreme opposite  being encouragement and independence. The care subscale allows maximum of 36 and overprotection a score of 39. The scale yields information on four dimensions, namely: mother care, father care, mother overprotection, and father overprotection. The participants’ responses are scored on a four-point scale ranging from “very likely” to “very unlikely”. Some of the items are reverse scored. The PBI demonstrated high internal consistency with split-half reliability coefficients of 0.88 for care and 0.74 for overprotection. The instrument shows good concurrent validity and correlated significantly well with independently rated judgement of parental care and overprotection.

Analysis

Stepwise multiple regression analysis was carried out to identify the level of variance in dependent variable that could be accounted by the different variables (cognitive distortion dimensions and parent-child relationship dimensions) and the impact of each dependent variable. Total depression scores generated from RADS-2 were taken as the criterion.

Interpretation and Discussion

As can be gleaned from Table 1, the highest positively contributing dimension is self-criticism (β=0.60) which was followed by helplessness (β=0.34), preoccupation with danger (β=0.22), self-blame (0.14), and father overprotection (β=0.10). Whereas, father care dimension of parent-child relationship was contributing negatively towards adolescent depression (β=0.10).

Table 1: Stepwise Multiple Regression Analysis for Adolescent Depression

 

R

R2

R2 Δ

P

β

P

Self-criticism

0.60

0.36

0.36

0

0.60

<0.01

Helplessness

0.67

0.44

0.08

0

0.34

<0.01

Preoccupation with danger

0.68

0.47

0.03

0

0.22

<0.01

Father overprotection

0.69

0.48

0.01

0

0.10

<0.01

Father care

0.70

0.49

0.01

0

0.10

<0.01

Self-blame

0.71

0.50

0.01

0

0.14

<0.01

Table 1 further suggests that various cognitive distortion dimensions are also contributing towards depression in adolescents. It has been reported that self-reported exposure to stressful life events was associated longitudinally with increased engagement in rumination. In addition, rumination mediated the longitudinal relationship between self-reported stressors and symptoms of anxiety in both samples and the relationship between self-reported life events and symptoms of depression in the adult sample. Identifying the psychological and neurobiological mechanisms that explain a greater propensity for rumination following stressors remains an important goal for future research. This study provides novel evidence for the role of stressful life events in shaping characteristic responses to distress, specifically engagement in rumination, highlighting potentially useful targets for interventions aimed at preventing the onset of depression and anxiety [25].

According to Blatt and others (e.g., A. T. Beck), self-definition, or one’s sense of self and one’s sense of relatedness to others represent core lifespan developmental tasks. This study examined the role of events pertaining to self-definition or relatedness in the development of personality traits from each domain (self-criticism and dependency), and their relationship to the development of depressive and anxiety symptoms. Two hundred seventy-six early adolescents completed a measure of self-criticism and dependency at baseline and again 24 months later, along with measures of depressive and anxiety symptoms. Every three months, participants completed a measure of life events, which were coded as self-definitional or relatedness oriented (80% rater agreement, kappa = .70). Structural equation models showed that self-definitional events predicted increases in self-criticism, which in turn predicted increases in depressive symptoms, whereas relatedness events predicted increases in dependency, although dependency was unrelated to change in symptoms [26].

One study [27] has suggested that early intervention with mother–child dyads during this developmental period may promote more adaptive attachment behaviours that could subsequently change the developmental trajectory of these “at-risk” children. Moreover, a special focus might be on improving mother–child interactions, as we know now that disorganised children are likely to have interactions that are characterised by role reversal. In addition, these findings point to the need to work with mothers to help them with their roles as parents to prevent caregiving helplessness, which we know now, along with role reversal, plays an important role in explaining the association between disorganised/controlling attachments and externalising problems in adolescence. These findings will be important for future prevention and intervention efforts.

Conclusion

Preoccupation with danger (β=0.22) as dimension of cognitive distortion is contributing positively towards adolescent depression. Self-blame (β=0.71) as dimension of cognitive distortion is contributing positively towards adolescent depression. It seems that adolescents give up against the problem and they have no way of dealing with the depression. Depressed adolescents appear to have predominantly cognitive symptoms with negative thought processes such as feelings of self-blame, self-hate, punishment, dissatisfaction and failure. Moreover, because of the digital age, adolescent are facing more stressors. For instance, smartphone ownership was related to more electronic media use in bed before sleep, particularly calling/sending messages and spending time online compared to adolescents with a conventional mobile phone. Smartphone ownership was also related to later bedtimes while it was unrelated to sleep disturbance and symptoms of depression. Sleep disturbance partially mediated the relationship between electronic media use in bed before sleep and symptoms of depression. Electronic media use was negatively related with sleep duration and positively with sleep difficulties, which in turn were related to depressive symptoms. Sleep difficulties were the more important mediator than sleep duration. The results of this study suggest that adolescents might benefit from education regarding sleep hygiene and the risks of electronic media use at night [28].

Previous works show that eliminating these distortions and negative thoughts is said to improve mood and discourage maladies such as depression and chronic anxiety. The process of learning to refute these distortions is knows and “cognitive restructuring”.

Father overprotection (β=0.10) is positively contributing to depression among male adolescents. According to theoretical views, parental overprotection may lead to anxiety by increasing beliefs in dangerousness of the situation and the lack of ability to avoid the danger [29].

Descriptions of parental authority and of the formation of a secure parent-child bond have remained unconnected in conceptualisations about parenting and child development. The parental anchoring function is here presented as an integrative metaphor for the two fields. Parents who fulfil an anchoring function offer a secure relational frame for the child, while also manifesting a stabilising and legitimate kind of authority. The anchoring function enriches the two fields by: (1) adding a dimension of authority to the acknowledged functions of the safe haven and the secure base that are seen as core to a secure parent-child bond, and (2) adding considerations about the parent-child bond to Baumrind’s classical description of authoritative parenting [30-33].

Finding

The main finding suggests that out of cognitive distortion and parent-child relationship dimensions, self-criticism, hopelessness, preoccupation with danger, father overprotection and self-blame are contributing positively towards adolescent depression. Father care is negatively contributing to adolescent depression scores.

Father care plays an important role in the depression among male adolescents. It is clear that parent-child relationship and inaccurate thoughts and ideas are important determinants of depressive symptoms among adolescents. Adolescence is a challenging phase of life. However, healthy parent-child relationship can cushion the effects of ruthless biopsychosocial changes of this period.  Adolescents need to be educated as to how to make healthy appraisals of events and occurrences within and around them and a healthy parent-child relationship can ensure better psychological health in adolescents.

Limitations and Future Directions

Although the current investigation provides useful insight for understanding depression which has important implications for dealing positively with the issue of adolescent depression but the study is not free from limitation. The study was only limited to with Fe male adolescents ageing 18-19. Hence, the findings should not be generalised. Moreover, the sample was selected from all the major city of India City which limits the scope for the generalisation of the findings. The focus of the investigation was on studying the relative contribution of subscales of cognitive distortion (self-criticism, self-blame, helplessness, and preoccurance and preoccupation with danger) and the dimensions of parent-child relationship (mother care, mother overprotection, father care, and father overprotection). However, there are many other variables that might contribute towards adolescent depression which might be studies in the future. This finding calls for the improvement of access to adolescent mental health services in rural India.

References

  1. Erikson EH (1951) Childhood and society (Rev. ed.).
  2. Brooks FM, Magnusson J, Spencer N, Morgan A (2012) Adolescent multiple risk behaviour: an asset approach to the role of family, school and community. Journal of Public Health 34: i48-i56. [crossref]
  3. Rushton JL, Forcier M, Schectman RM (2002) Epidemiology of depressive symptoms in the National Longitudinal Study of Adolescent Health. Journal of the American Academy of Child & Adolescent Psychiatry 41: 199-205. [crossref]
  4. Done DJ, Crow TJ, Johnstone EC, Sacker A (1994) Childhood antecedents of schizophrenia and affective illness: social adjustment at ages 7 and 11. British Medical Journal 309: 699-703. [crossref]
  5. Pilao SJ, Relojo D, Tubon G, Subida M (2016) Positive Ageing and Perception of Loneliness among Elderly Population. International Journal of Scientific Research and Innovative Technology 3: 105-114.
  6. Ollendick TH, Seligman LD, Goza AB, Byrd DA, Singh K (2003) Anxiety and depression in children and adolescents: A factor-analytic examination of the tripartite model. Journal of Child and Family Studies 12: 157-170.
  7. Nolen-Hoeksema S, Girgus JS (1994) The emergence of gender differences in depression during adolescence. Psychological Bulletin 115: 424-443. [crossref]
  8. Rapee RM, Wignall A, Hudson JL, Schniering CA (2000) Treating anxious children and adolescents: An evidence-based approach. New Harbinger Publications.
  9. Relojo D, dela Rosa R, Pilao SJ (2016) Reading abilities among adolescent second language readers: Theoretical, cultural and educational implications. International Research Journal of Humanities & Social Science 1.
  10. Beck CT, Gable RK (2001) Comparative analysis of the performance of the Postpartum Depression Screening Scale with two other depression instruments. Nursing Research 50: 242-250. [crossref]
  11. Bowlby J (1969) Attachment and loss v. 3 (Vol. 1). Random House. Furman W, Buhrmester D (2009) Methods and measures: The network of relationships inventory: Behavioral systems version. International Journal of Behavioral Development 33: 470-478.
  12. Bowlby J, Ainsworth MD (1967) Maternal Care and Mental Health. Deprivation of Maternal Care: A Reassessment of Its Effects. Schocken
  13. Gee A, Troop NA (2003) Shame, depressive symptoms and eating, weight and shape concerns in a non-clinical sample. Eating and Weight Disorders-Studies on Anorexia, Bulimia and Obesity 8: 72-75. [crossref]
  14. Jenkins SR, Goodness K, Buhrmester D (2002) Gender differences in early adolescents’ relationship qualities, self-efficacy, and depression symptoms. The Journal of Early Adolescence 22: 277-309.
  15. Al-Yagon M, Kopelman-Rubin D, Brunstein Klomek A, Mikulincer M (2016) Four-model approach to adolescent-parent attachment relationships and adolescents’ loneliness, schoool belonging, and teacher appraisal. Personality Relationships 23: 141-158.
  16. Relojo D (2016) Moderating Effects of Dietary Restraint and Self-Compassion in an Expressive Writing Intervention in Southeast Asian Population. Journal of Behavioural Sciences 26: 1-20.
  17. Kim KJ, Conger RD, Elder Jr GH, Lorenz FO (2003) Reciprocal influences between stressful life events and adolescent internalizing and externalizing problems. Child Development 74: 127-143. [crossref]
  18. Eamon MK (2002) Influences and mediators of the effect of poverty on young adolescent depressive symptoms. Journal of Youth and Adolescence 31: 231-242.
  19. Levendosky AA, Huth-Bocks AC, Shapiro DL, Semel MA (2003) The impact of domestic violence on the maternal-child relationship and preschool-age children’s functioning. Journal of Family Psychology 17: 275-287. [crossref]
  20. Johnson WL, Giordano PC, Longmore MA, Manning WD (2014) Intimate partner violence and depressive symptoms during adolescence and young adulthood. Journal of Health and Social Behavior 55: 39-55. [crossref]
  21. Murray KW, Dwyer KM, Rubin KH, Knighton-Wisor S, Booth-LaForce C (2014) Parent–child relationships, parental psychological control, and aggression: Maternal and paternal relationships. Journal of Youth and Adolescence 43: 1361-1373. [crossref]
  22. Reynolds WM (2010) Reynolds adolescent depression scale. John Wiley & Sons, Inc.
  23. Briere, J. (2000). Cognitive distortion scales: Professional manual. Psychological Assessment Resources.
  24. Parker G, Tupling H, Brown LB (1979) A parental bonding instrument. British journal of medical psychology 52: 1-10.
  25. Michl LC, McLaughlin KA, Shepherd K, Nolen-Hoeksema S (2013) Rumination as a mechanism linking stressful life events to symptoms of depression and anxiety: Longitudinal evidence in early adolescents and adults. Journal of Abnormal Psychology 122: 339-352. [crossref]
  26. Kopala-Sibley DC, Zuroff DC, Hankin BL, Abela JR (2015) The Development of Self-Criticism and Dependency in Early Adolescence and Their Role in the Development of Depressive and Anxiety Symptoms. Personality and Social Psychology Bulletin 41: 1094-1109. [crossref]
  27. Lecompte V, Moss E (2014) Disorganized and controlling patterns of attachment, role reversal, and caregiving helplessness: links to adolescents’ externalizing problems. American Journal of Orthopsychiatry 84: 581-589. [crossref]
  28. Lemola S, Perkinson-Gloor N, Brand S, Dewald-Kaufmann JF, Grob A (2015) Adolescents’ electronic media use at night, sleep disturbance, and depressive symptoms in the smartphone age. Journal of Youth and Adolescence 44: 405-418. [crossref]
  29. Rapee RM (1997) Potential role of childrearing practices in the development of anxiety and depression. Clinical Psychology Review 17: 47-67. [crossref]
  30. Omer H, Steinmetz SG, Carthy T, Schlippe A (2013) The anchoring function: Parental authority and the parent‐child bond. Family process 52: 193-206. [crossref]
  31. Blatt SJ, Zuroff DC (1992) Interpersonal relatedness and self-definition: Two prototypes for depression. Clinical Psychology Review 12: 527-562.
  32. Carlson MJ, Berger LM (2013) What kids get from parents: Packages of parental involvement across complex family forms. Social Service Review 87: 213-249. [crossref]
  33. Jeynes WH (2015) A meta-analysis: The relationship between father involvement and student academic achievement. Urban Education 50: 387-423.