Article Page

DOI: 10.31038/JMG.2020324

Abstract

HidradenitisSuppurativa(HS), also named acne inversa, which is a common chronic inflammatory skin disorder characterized clinically by painful lumps, abscesses and scarring. Thirty-five unique mutations in patients with HS have been identified in three of the genes that encode members of the γ-secretase complex: nicastrin (NCSTN), presenilin 1 (PSEN1), and presenilin enhancer 2 (PSENEN) as well as in POGLUT1, an Endoplasmic Reticulum (ER) O-glucosyltransferase that is involved in Notch signaling. This review summarizes research updates on genetics of HS.

Keywords

Hidradenitis suppurativa, γ-secretase, nicastrin, presenilin

Introduction

Hidradenitis Suppurativa (HS), also named acne inversa, is a common chronic inflammatory skin disorder characterized clinically by painful lumps, abscesses and scarring (OMIM # 142690). The prevalence of HS in the population is 0.10%, or 98 per 100,000 persons in the United States (US) [1,2] and three times more common in female patients (73.8% women) than male patients (26.2% men), 3-fold greater in African Americans and 2-fold greater in biracial populations than in the overall population [1]. Antibiotics, anti-inflammation regiments, acne washes and medicines, and surgical procedure are the premirary current treatment options [3]. Major surgery demonstrated improvements in the HS patients’ overall work and daily activity impairment [4]. However, the disease progression often causes scars leading immobility, markedly affecting quality of life in severe patients who have poor responses to treatments [5].

The etiology of HS is associated with multi-factoralsincuding genetics and others. HS increased an independent risk of all-cause mortality [6]. Obesity, smoking, family history and environmental factors such as diet, are known to be associated with the HS disease pathogenesis. Obesity is linked to skin barrier function, sebaceous glands and sebum production, sweat gland, lymphatics, and collagen structure and function, wound healing, microcirculation and macrocirculation [7]. Obesity and smoking increase the HS incidence [8] [9]. HS patients classified as Hurley III HS were 28% more likely to be smokers and obese [10] and four times more likely to be obese compared to the general population by meta-analysis of case-control studies in Asia, Europe, and the US [11]. One-third (31%) of the HS patients who eliminated smoking or made dietary alterations including a reduction in gluten, dairy, refined sugars, tomatos, or alcohol showed improvement in HS clinical symptoms [12]. Patients with HS were at higher risk for long-term opioid use compared with controls [13].

HS lesion counts are increased with low serum zinc and vitamin D levels. Supplementation of zinc, vitamin D, vitamin B12, or exclusion of dairy or brewer’s yeast reduced lesion resolution. Bariatric surgery often causes weight loss which may lead to HS improvement but oftenresults in more severe malnutrition thatworsens or even leads to new HS onset post bariatric surgery [11]. The complement (C) system wasfound to be significantly down-regulated in the HS skin and blood transcriptomes and the HS blood proteome [81]. Porphyromonas species, which are able cleave inactive C5 into C5a, have been identified in the HS microbiome.  C5a levels in serum and tissue correlate with disease activity and degree of neutrophilic infiltrates in HS, suggesting that complement inhibition is a promising and potential therapeutic target for HS [82]. HS lesions showed 83% bacterial culture anaerobes compred to 53% of control samples, and milleri group streptococci and actinomycetes in 33% and 26% of cases, respectively [83]. Microarray analysis demonstrated that HS lesional skin samples had significantly decreased expression of enzymes involved in generating ceramide and sphingomyelin, increased expression of enzymes that catabolize ceramide to sphingosine, and increased expression of enzymes involved in converting ceramide to galactosylceramide and gangliosides, which suggests that sphingolipid metabolismi saltered in HS lesional skin comparedwith normal skin [86]. In HS patients, the serum and HS skin lension levels of chitinase-3-like protein 1 (YKL-40) were significantly elevated, suggesting that YKL-40 maybe one of the biomarkers of HS [87].

HS patients demonstrate a significantly higher heart rate in the HS groups than in the population [14]. HS often co-existed with psoriasis. Compared to patients with psoriasis alone, HS patients with psoriasis were significantly younger and had a higher prevalence of obesity and smoking [15].

Macrophages in HS infiltrates release a variety of pro-inflammatory cytokines such as interlukins and tumornecrosis factor α (TNFα), exacerbating the inflammation. Obesity and smoking contribute to macrophage dysfunction [9]. Elevated expression of TNFα has been identified in skin lesions, such as skin tunnels, of HS patients alongwith a clustering of interleukins (IL‐8, IL‐16, IL‐1α and IL‐1β) [68] [69]. Gene-sets related to Notchsignalling and Interferonpathwaysweredifferentiallyactivated in HS lesionalcompared to non-lesional skin [80].

Adalimumab is a TNFα inhibitor which has been used in both USA and Europe for treating HS patients. Adalimumab reduced flare, showed a higher efficacy on nodules-abscesses than on draining tunnels and increased the number of patients achieving a Hidradenitis Suppurativa Clinical Response [91]. By a Genome-Wide Association Study (GWAS) analysis one single Linkage Disequilibrium (LD) block in the BCL2 gene was significantly associated with adalimumab response (lead Single-Nucleotide Polymorphism [SNP] rs 59532114). Meanwhile, a correlation of the most strongly associated SNP minor allele with increased BCL2 gene and protein expression in hair follicle tissues was observed with bioinformatic analysis and functional genomics experiments [66]. HLA alleles may affect the treatment response in HS patients treated with adalimumab. Threre were three protective HLA alleles (HLA-DQB1*05, HLA-DRB1*01, and HLA-DRB1*07) less prevalent and two risk HLA alleles (HLA-DRB1*03 and HLA-DRB1*011) more abundant in HS patients developing anti-drug antibodies to adalimumab than these not [67].

Genes Linked to HS

Genetics is assciated with the pathogenesis of HS. One third of HS patients have a family history with an autosomal dominant inherentance trait [16] which pattern suggests a single gene disorder. Thirty-five unique mutations in patients with familial or sporadic HS have been found in genes encoding three of the four genes comprising the γ-secretase complex: nicastrin (NCSTN), presenilin 1 (PSEN1), presenilin enhancer 2 (PSENEN)  [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] and in POGLUT1, an Endoplasmic Reticulum (ER) O-glucosyltransferase involving in Notch signaling [33] with a diversity of mutation types in Caucasian, Chinese, Japanese, Indian or African ethnic origin (Table 1) [34][35]. NCSTN possesses majority (74%, 26/35) of the mutations (6 missenses, 8 nonsenses, 6 frameshifts, and 6 in splice sites resulted in frameshift or in-frame deletions). A single frameshift PSEN1-P242LfsX11 mutation was detected in PSEN1[21]. Six mutations were found in PSENEN (18%, 6/34) (3 frameshifts, 1 nonsense, 1 splicing, 1 missense). Two mutations were in POGLU1 (1 nonsense, 1 splicing).  NCSTN-R117X and Q568X were identified in more than one ethnic population and multiple families; the rest HS-linked mutations are private to each HS family or subject. NCSTN-c.1799delTG is a two-base deletion that leads to a nonsense change L600X, while 2 splicing site mutations in NCSTN, c.582+1delG p. F145fs_X54 and c.1551+1G>A p.A486_T517del result in frame-shifts while the other 4 splicing mutations cause in-frame deletions (Table 1) [34]. HS-associated mutation types in NCSTN, PSEN1, PSENEN andPOGLU1 are missense 20% (7/35), nonsense 29% (10/35), frameshift 29% (10/35) and splicing site changes 22% (8/35).

Table 1.Mutation spectrum of NCSTN, PSEN1, PSENEN and POGLUT1 in HS patients

ID

Mutation category

Nucleotide Change

Amino Acid Change

TM

Ethnic origin

Reference

NCSTN

1

Missense

c. 223G>A

p.V75I

Yes

Chinese

36

2

c.553G>A

p.D185N

Yes

Caucasian

17

3

c.632C>G

p.P211R

Yes

Chinese

18

4

c.647A>C

p.Q216P

Yes

Chinese

36

5

c.944C>T

p.A315V

Yes

Chinese

19

6

c.1229C>T

p.A410V

Yes

Chinese

20

7

Nonsense

c. 349C>T

p.R117X

No

Chinese,
Caucasian,
African

21
20
22

8

c.477C>A

p.C159X

No

Chinese

23

9

c.497C>A

p.S166X

No

Chinese

24

10

c.1258C>T

p.Q420X

No

Chinese

94

11

c. 1300C>T

p.R434X

No

Caucasian

25

12

c. 1695T>G

p.Y565X

No

Chinese

18

13

c.1702C>T

p.Q568X

No

Caucasian
Japanese

95

14

c.1799delTG

p.L600X

No

Indian

26

15

Frameshift

c.210_211delAG

p.T70fsX18

No

Chinese

27

16

c.487delC

p.Q163SfsX39

No

Chinese

21

17

c.687insCC

p.C230PfsX31

No

Indian

26

18

c.1752delG

p.E584DfsX44

No

Chinese

21

19

c.1768A>G

p.590AfsX3

No

Caucasian

25

20

c.1912_1915delCAGT

p.S638fsX1

No

Caucasian

35

21

Splice Site

c.582+1delG

p. F145fs_X54

No

Japanese

95

21

c.996+7 G>A

p.L282_G332del

Yes

Caucasian

17

23

c.1101+1 G>A

p.E333_Q367del

Yes

Caucasian

28

24

c.1101+10 A>G

p.E333_Q367del

Yes

African

17

25

c.1352+1 G>A

p.Q393fs_X9

No

Chinese

27

26

c.1551+1G>A

p.A486_T517del

No

Chinese

21

PSEN1

27

Frameshift

c.725delC

P242LfsX11

Chinese

21

PSENEN

28

Frameshift

c.66delG

p.F23LfsX46

Chinese

21 29

29

c.66_67insG

p.F23VfsX98

Caucasian

17

30

c.279delC

p.P94SfsX51

Chinese

21

31

Nonsense

c.168T>G

p.Y56-101Pdel

Caucasian

30

32

Splicing

c.167-2A>G

p.G55-101Pdel

Chinese

31

33

Missense

c.194T>G

p.L65R

Chinese

31

POGLUT1

34

Nonsense

c.814C>T

p.R272*

Caucasian

32

35

Splicing

c.430‐1G>A

p.K246_392Ldel

Caucasian

33

HS patients who carry a mutation in NCSTN, PSEN1, PSENEN or POGLUT1 display severe or typical symptoms of HS lesions [36][17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32][33]s. HS patients who carrying a PSENEN or POFUT1 mutation also have co-ocurrent Dowling–DegosDisease (DDD) syndrome), an abnormally dark skin coloring condition (hyperpigmentation) [32] [37] [38] [30] [33] while mutations in NCSTN and PSEN1 occur in patients with HS only. There were 2.8 -fold patients with complex HS demenstating increase in pathogenic variants of an innate immunity regulator pyrin (also known as marenostrin, MEFV) compared to the healthy controls in the general Turkish population [39].

Structures of HS-Linked Genetic Mutations

The putative functions of the HS-linked mutations were analyzed by in silico analysis of using a variety of programs. By SWISS-MODEL, most of the HS-linked nonsense, frameshift, and splice site mutations resulted in marked 3D structural changes, and a C-terminal end frameshift mutation NCSTN-E584DfsX44 led to a striking 3D structural change while another nearby downstream frameshift mutation NCSTN-p.590AfsX3 (6 amino acids apart) caused only a minor 3D change [34]. This finding suggests that this NCSTN-E584DfsX44 mutation is likely located at a critical site for NCSTN conformation [34]. By PolyPhen-2, SNP & Go and Proven prediction, among 6 NCSTN missense mutations, NCSTN-P211R and Q216P were most deleterious; PolyPhen-2 predicts that V75I is probably damaging and D185N, A315V and A410V are predicted to have benign or neutral effects. 62% (16/26) of NCSTN mutations are nonsense or frameshift mutations that causes a truncation of the protein product. Structurally, NCSTN contains a large extra cellular domain and a single TM [40], that is located at amino acid position 670-692. 39% (10/26)  of NCSTN mutations (6 missense mutations and 4 splicing site mutations) retain the TM region, while 61% (16/26) of other NCSTN nonsense, frameshift mutations and c.582+1delG [17] and c.1352+1 G>A (experimental confirmed) [27] lose the TM domain to become cytosolic proteins that cannot enter the cell to initial signaling (Table 1). Among 4 splicing site mutations that do not affect TM regions, 3 potentially affect two key NCSTN substrate recruitment sites Gry333 and Tyr337. The p. L282_G332del occurs next to a residue of the NCSTN substrate recruitment site G333; and E333_Q367del and E333_Q367del completely abolish the 2 NCSTN substrate recruitment sites Gry333 and Tyr337 [40], which suggest that these NCSTN mutations affect important substrate recruitment structures. 50% (3/6) of the NCSTN splicing site mutations affect substrate recruitment [34].

Post Translation of HS-Linked Genetic Mutations

NCSTN mutatioNS Y565X occurs on a tyrosine phosphorylation site and R434X occurs on a glycosylation site. NCSTN-R434X disrupts the protein immediately before Asn435, one of the two NCSTN prominent glycans Asn55 and Asn435 [40]. 21% (5 of 24) of the NCSTN mutations, NCTSN-P211R, L600X, C230PfsX31, P590AfsX3 and F145fs_X54 occur at cysteine residues participating in disulfide bonds [41] [42]. Six potential NCSTN ubiquitination sites are predicted:  K78, T127, K386, K403, K591 and K597. Six residues in NCSTN undergo sumolyation: G146, S341, K386, P423, T459, and D476. NCSTN-P590AfsX3 occurs immediately before the predicted ubiquitination site K591 and abolishes two ubiquitination sites – K591 and K597. F145fs_X54 abolishes sumolyation site G146. Both NCSTN- E333_Q367del and E333_Q367del abolish sumolyation site S341. NCSTN-T70fsX18 and R117X abolish all the ubiquitination and sumolyation sites and C159X and S166X abolish four of the six ubiquitination sites and five of the six sumolyation sites [34]. The C-terminal end frameshift mutation NCSTN-E584DfsX44 resulted in a striking 3D structural change suggesting that this mutation is likely located at a critical site for NCSTN conformation [34]. Ubiquitiation and sumoylation are involved in post-translational modification. A large number of NCSTN mutations affect predicted ubiquitiation and sumoylation sites, suggesting that post-translational modification might contribute to HS pathogenesis.

HS-Linkled Mutational Effect

HS associated mutations in NCSTN are predicted to cause a loss of function as a result of frameshift and premature translation termination and a loss of  the TM domain, to affect NCSTN substrate recruitment sites, to cause a loss or creation of new ligand binging sites, and to alter post-translational modifications and disulfide bonds [41] [42], all of which support the notion that the NCSTN mutations result in significantly reduced levels of NCT and reduced γ-secretase-mediated processing of Notch and signaling in the skin [43].  Silencing of the keratinocyte NCSTN by CRISPR-Cas9 in both the keratinocyte cell line HEK001 and an embryonic kidney cell line HEK293 showed a significantly increased expression of genes related to the type I interferon response pathway [44]. NCSTNWild Type (WT) were upregulated in myeloid cells including monocytes, macrophages and non-lymphoid dendritic cells [35]. NCSTN knockdown in HaCaT cells impaired γ-secretase activity and proliferation and differentiation of keratinocytes. Expression levels of several γ-secretase substrates involved in the Notch pathway were significantly attenuated in NCSTN-silencing HaCaT cells and the lesion from a HS patient. Phosphoinositide 3-kinase (PI3K) as well as AKT and its activated form pAKT were markedly elevated in NCSTN-silencing HaCaT cells [23]. NCSTN mutations led to decreased miR-30a-3p levels, which negatively regulated RAB31 expression. Moreover, enhanced RAB31 levels accelerated degradation of activated EGFR, leading to abnormal differentiation in keratinocytes. Familial HS patients and mouse knocked out for Ncstn showed impaired EGFR signaling and epidermal differentiation [45].

However, testing four NCSTN-missense mutations, V75I, D185N, P211R, and Q216P for their effects on mediating Notch processing and signaling demonstrated a vague role of HS-linked NCSTN mutations in HS pathogenesis. The NCSTN-V75I, D185N, and P211R mutants can function in Notch signaling in vivo; in contrast mutant Q216P failed to rescue Notch processing and nuclear signaling [46]. Mouse models where components of the ¡-secretase with resultant Notch dysregulation have been knocked out have resulted in the development of dermal cysts and histological features of follicular occlusion [21][47] although these models rapidly developed multiple squamous cell carcinomas,which is not consistent with the typical progression of HS [47]. These findings suggest that although NCSTN-V75I, D185N, and P211R and some other NCSTN mutations have a significant role in the pathogenesis of the disease, this role is through a mechanism(s) other than impaired Notch signaling.

A single frameshiftPSEN1-P242LfsX11 mutation is predicted to truncate the PS1 protein after the 5th TM domain at the cytosolic region of the N terminal, which would markedly alter the 3D structure of PS1. PSENEN contains three TMs, at amino acid positions 18-38, 60-80 and 85-101. The PSENEN N-terminus is cytoplasmic, followed by two short helices that dip into the membrane [40]. All the PSENEN mutations occur within TM regions: frameshift mutations F23LfsX46 and F23VfsX98 delete all 3 TM regions, while P94SfsX51 disrupts TM region 3. Nonsense Y56-101Pdel and c.167-2A>G splicing site mutations lead to similar disruptions of TM regions 2 & 3. The missense mutation PSENEN-L65R laysin the TM 2 region and is predicted to be deleterious. POGLUT1 is located in the lumen of the endoplasmic reticulum. Both POGLUT1-R272* and C.430-1G>A, K246* lead to an early termination of protein synthesis. POGLUT1-R272* is located in the C-terminal domain and results in a truncated form of POGLUT1 with partial loss of the C-terminal domain. The splicing site c.430‐1G>A mutation was identified in exon 4 of the POFUT1 gene in patients with HS and DDD syndrome, which potentially generates aberrant splicing with loss of functionality [33]. POGLUT1 is predicted to possess 17 ligand binding sites of interactions with chain A. Hydrogen bonds include A.Y117, A.S152, A.R158, A.R158, A.D196, A.V197, A.V197, A.L199, A.V214, A.A215, A.A215, A.S217, A.F218, A.R219, A.R219 and salt bridges: A.R158 and A.R219. Both POGLUT1- c.430‐1G>A (K246*) or R272* completely abolish ligand binding function and show significant alteration of global quality estimate by Qualitative Model Energy Analysis (QMEAN) values:  POGLUT1-WT:-71; POGLUT1- c.430‐1G>A (K246*): 0.90; and R272* 0.45, indicating a greater deviation in mutant forms from the POGLUT1-WT [34].

A higher and prolonged TNFα expression and differential gene expression of four cytokine or chemokines than that of PS1-WT in response to LPS stimulation was observed in overexpression of the HS-associated PSEN1 mutation PSEN1-P242LfsX11 in PMA-differentiated macrophages [34]. Of  the overexpressing PSEN1-WT and PSEN1-P242LfsX11 induced under-expressed genes [34],  LIF and CSF2 are essential for the proliferation and differentiation of hematopoietic progenitor cells into granulocytes and macrophages [48] [49], IL12 is critical for the activation and maintenance of immune responses [50], and BMP2 regulates stem cell activation in the process of hair follicle regeneration in the dermis [51]. Theincreased expression of proinflammatoryTNFα and the decreased expression of LIF, IL12B, CSF2, BMP2 and other genes associated with the overexpression of PSEN1-P242LfsX11 may promote inflammatory processes, impair the activation/maintenance of immune cells and reduce hair follicle regeneration [34]. HS patients with a PSEN1 mutation may benefit greatly from TNFα inhibiting agents such as infliximab, adalimumab, rituximab, and ustekinumab, in particular after anti-inflammatory regimens fail to control the disease process.

PSEN1 has pleiotropic nature [52]. PSEN1 is linked to early-onset familial Alzheimer’s Disease (AD) (OMIM # 104300), a neurodegenerative disorder and the most common form of dementia in the elderly [53]. A single frameshiftPSEN1-P242LfsX11 mutation was detected in familial HS patients [21]. More than 185 missense or inframe deletion mutations and promoter variants in PSEN1 have previously been found in patients with familial AD (http://www. alz.org/) and sporadic Dilated Cardiomyopathy (DCM) [54], and 685 genes have been associated with AD (www.alz.org). The familial HS patients with PSEN1-P242LfsX11 mutation did not show the symptoms of AD [21]. Significant differential expression of ErbB4, SCNB1, and Tie1 was observed in HS lesional skin, and of EphB2, EPHB4, KCNE1, LRP6, MUSK, SDC3, Sortilin1 were observed in blood specific to AD [55]. AD-associated PSEN1 mutations alters the -secretases cleavage of β-APP to increase Aβ 42/40 ratio resulting in Aβ plague formation and related AD pathology [21]. Overexpression or silencing of presenilin caused cardiac dysfunction in Drosophila [56]. Overexpression of PSEN1-P242LfsX11 in zebrafish embryos enhanced Notch signaling but did not affect γ-secretase cleavage of APP [57], which suggests that the involvement of the PSEN1 mutation in HS pathogenesis also has a mechanism that is independent of γ-secretase activity. Different from the effectiveness of administration of TNFα inhibitor Adalimumab in the treatment of HS patients, administration of the TNFα modulator etanercept in AD patients demonstrated no apparent effect on cognitive functioning, though TNFα has been implicated in the pathogenesis of AD [58] [59]. In AD patients, only one side of each TM helix in PS1 is affected, the hot spot of Leu219, Glu222, Leu226, Ser230, Met233, and Phe237 are placed on the same side of TM5 [40] while the HS-linked PSEN1-P242LfsX11 is on the other side of TM5 in PS1. This distribution or structure of AD-linked PSEN1 mutation is significantly different from HS-linked PSEN mutations which may indicate functional importance.

POGLUT1 is an Endoplasmic Reticulum (ER) O-glucosyltransferase that adds glucose moieties to serine residues in EGF-like repeats, such as NOTCH intracellular domain [60]. Mutations in POGLUT1, including W4X, R218X, R279PfsX3 and R279W, have been previously described in unrelated caucasian patients with Dowling-Degos disease (DDD) [32] [37] [38]. Mutations in POGLUT1 caused an approximately 50% weaker POGLU1 expression in patient lesional skin compared to controls, by immunohistologic staining for POGLUT1 [38]. In addition, a missense mutation in POGLUT1 was identified with patients with muscular dystrophy. Muscles from patients demonstrated decreased Notch signaling, dramatic reduction in satellite cell pool and a muscle-specific α-dystroglycanhypoglycosylation not present in patients’ fibroblasts, suggesting a Notch-dependent pathomechanism for this novel form of muscular dystrophy [60]. Mutations in PSENEN are also identified in DDD patients [30]. Evidence has suggested the association between decreased Notch activity and POFUT1 mutations [61]. The finding of POGLUT1 mutations in patients with HS-DDD syndrome indicates aberrant Notch signaling is involved in both HS and DDD pathogenesis. Notably, mutations in POGLUT1 and NCSTN are linked to dysregulation of Notch signaling which might also contribute to small vessel disease, as well as to vascular cognitive impairment [62].

Epigenetics of HS-Linked Genes

Significant epigenetic modifications were observed in HS skin lensions [63]. mRNA of all the studied genes were significantly under-expressed in lesional HS skin compared to healthy skin by RT-PCR analyses of The Expression of Translocation (TET) and Isocitrate Dehydrogenase (IDH) family genes in the lensional skins of HS patients, suggesting that epigenetic changes occur in HS tissue and that aberrant expression of the DNA hydroxymethylation regulators may play a role in the pathogenesis of HS [63]. HS was associated with a 1.69-fold increased odds of diabetes; however, the absolute risk difference was small and is probably not clinically relevant [64]. A significant overexpression of miRNA-155-5p, miRNA-223-5p, miRNA-31-5p, miRNA-21-5p, and miRNA-146a-5p was observed in lesional HS skin compared to healthy controls, suggesting that these miRNAs may be potential disease biomarkers and therapeutic targets for HS [65].

Acknowledgments

This work was supported by the National Institutes of Health [R01AG014713 and R01MH060009 to R.E.T; R03AR063271 and R15EB019704 to A.L.]; National Science Foundation [NSF1455613 to A.L] and the Cure Alzheimer’s Fund [to R.E.T].

References

  1. Garg A, Kirby JS, Lavian J, Lin G, Strunk A (2017) Sex- and Age-Adjusted Population Analysis of Prevalence Estimates for Hidradenitis Suppurativa in the United States. JAMA Dermatol 153: 760-764.[crossref]
  2. Wipperman J, Bragg DA, Litzner B (2019) Hidradenitis Suppurativa: Rapid Evidence Review. Am Fam Physician 100: 562-569.[crossref]
  3. Alikhan A, Lynch PJ, Eisen DB (2009) Hidradenitis suppurativa: a comprehensive review. J Am Acad Dermatol 60: 539-561.[crossref]
  4. Prens LM, Huizinga J, Janse IC, Horvath B (2019) Surgical outcomes and the impact of major surgery on quality of life, activity impairment and sexual health in hidradenitis suppurativa patients: a prospective single centre study. J Eur Acad Dermatol Venereol 33: 1941-1946.[crossref]
  5. Lasko LA, Post C, Kathju S (2008) Hidradenitis suppurativa: a disease of apocrine gland physiology. JAAPA 21: 23-25.[crossref]
  6. Reddy S, Strunk A, Garg A (2019) All-cause mortality among patients with hidradenitis suppurativa: A population-based cohort study in the United States. J Am Acad Dermatol 81: 937-942.[crossref]
  7. Yosipovitch G, DeVore A, Dawn A (2007) Obesity and the skin: skin physiology and skin manifestations of obesity. J Am Acad Dermatol 56: 901-916.[crossref]
  8. Acharya P, Mathur M (2020) Hidradenitis suppurativa and smoking: a systematic review and meta-analysis. J Am Acad Dermatol82:1006-1011[crossref]
  9. Shah A, Alhusayen R, Amini-Nik S (2017) The critical role of macrophages in the pathogenesis of hidradenitis suppurativa. Inflamm Res 66: 931-945.[crossref]
  10. Vankeviciute RA, Polozovaite B, Trapikas J, Raudonis T, Grigaitiene J et al. (2019) A 12-Year Experience of Hidradenitis Suppurativa Management. Adv Skin Wound Care 32: 1-7.[crossref]
  11. Choi F, Lehmer L, Ekelem C, Mesinkovska NA (2020) Dietary and metabolic factors in the pathogenesis of hidradenitis suppurativa: a systematic review. Int J Dermatol 59: 143-153[crossref]
  12. Dempsey A, Butt M, Kirby JS (2019) Prevalence and Impact of Dietary Avoidance among Individuals with Hidradenitis Suppurativa. Dermatology 1: 1-7.[crossref]
  13. Reddy S, Orenstein LAV, Strunk A, Garg A (2019) Incidence of Long-term Opioid Use Among Opioid-Naive Patients With Hidradenitis Suppurativa in the United States. JAMA Dermatol.[crossref]
  14. Miller IM, Ahlehoff O, Vinding G, Rytgaard H, Mogensen UB et al. (2018) Hidradenitis Suppurativa is Associated with Higher Heart Rate but Not Atrial Fibrillation: A Comparative Cross-sectional Study of 462 Individuals with Hidradenitis Suppurativa in Denmark. Acta Dermatovenerol Croat 26: 289-296.[crossref]
  15. Kridin K, Shani M, Schonmann Y, Fisher S, Shalom G et al.(2018) Psoriasis and Hidradenitis Suppurativa: A Large-scale Population-based Study. J Am Acad Dermatol.[crossref]
  16. Ingram JR (2016) The Genetics of Hidradenitis Suppurativa. Dermatol Clin 34: 23-28.[crossref]
  17. Pink AE, Simpson MA, Desai N, Dafou D, Hills A,et al. (2012) Mutations in the gamma-secretase genes NCSTN, PSENEN, and PSEN1 underlie rare forms of hidradenitis suppurativa (acne inversa). J Invest Dermatol 132: 2459-2461.[crossref]
  18. Li CR, Jiang MJ, Shen DB, Xu HX, Wang HS et al. (2011) Two novel mutations of the nicastrin gene in Chinese patients with acne inversa. Br J Dermatol 165: 415-418.[crossref]
  19. Zhang S, Meng J, Jiang M, Zhao J (2016) Characterization of a Novel Mutation in the NCSTN Gene in a Large Chinese Family with Acne Inversa. Acta Derm Venereol 96: 408-409.[crossref]
  20. Liu M, Davis JW, Idler KB, Mostafa NM, Okun MM et al. (2016) Genetic analysis of NCSTN for potential association with hidradenitis suppurativa in familial and nonfamilial patients. Br J Dermatol175: 414-416.[crossref]
  21. Wang B, Yang W, Wen W, Sun J, Su B et al. (2010) Gamma-secretase gene mutations in familial acne inversa. Science 330: 1065.[crossref]
  22. Chen S, Mattei P, You J, Sobreira NL, Hinds GA (2015) gamma-Secretase Mutation in an African American Family With Hidradenitis Suppurativa. JAMA Dermatol151: 668-670.[crossref]
  23. Xiao X, He Y, Li C, Zhang X, Xu H et al. (2016) Nicastrin mutations in familial acne inversa impact keratinocyte proliferation and differentiation through the Notch and phosphoinositide 3-kinase/AKT signalling pathways. Br J Dermatol 174: 522-532.[crossref]
  24. Ma S, Yu Y, Yu G, Zhang F (2014) Identification of one novel mutation of the NCSTN gene in one Chinese acne inversa family. Dermatologica Sinica 32: 126-128.
  25. Miskinyte S, Nassif A, Merabtene F, Ungeheuer MN, Join-Lambert O et al. (2012) Nicastrin mutations in French families with hidradenitis suppurativa. J Invest Dermatol 132: 1728-1730.[crossref]
  26. Ratnamala U, Jhala D, Jain NK, Saiyed NM, Raveendrababu M (2016) Expanding the spectrum of gamma-secretase gene mutation-associated phenotypes: two novel mutations segregating with familial hidradenitis suppurativa (acne inversa) and acne conglobata. Exp Dermatol 25: 314-316.[crossref]
  27. Liu Y, Gao M, Lv YM, Yang X, Ren YQ et al. (2011) Confirmation by exome sequencing of the pathogenic role of NCSTN mutations in acne inversa (hidradenitis suppurativa). J Invest Dermatol 131: 1570-1572.[crossref]
  28. Pink AE, Simpson MA, Brice GW, Smith CH, Desai N et al. (2011) PSENEN and NCSTN mutations in familial hidradenitis suppurativa (Acne Inversa). J Invest Dermatol 131: 1568-1570.[crossref]
  29. Liu Y, Miao T, Ma J, Shao L, Luo S et al. (2016) PSENEN c.66delG in sporadic acne inversa. Eur J Dermatol 26: 298-299.[crossref]
  30. Pavlovsky M, Sarig O, Eskin-Schwartz M, Malchin N, Bochner R et al. (2018)A phenotype combining hidradenitis suppurativa with Dowling-Degos disease caused by a founder mutation in PSENEN. Br J Dermatol 178: 502-508.[crossref]
  31. Zhou C, Wen GD, Soe LM, Xu HJ, Du J et al. (2016) Novel Mutations in PSENEN Gene in Two Chinese Acne Inversa Families Manifested as Familial Multiple Comedones and Dowling-Degos Disease. Chin Med J (Engl) 129: 2834-2839.[crossref]
  32. Duchatelet S, Clerc H, Machet L, Gaboriaud P, Miskinyte S et al. (2018) A new nonsense mutation in the POGLUT1 gene in two sisters with Dowling-Degos disease. J Eur Acad Dermatol Venereol.[crossref]
  33. Gonzalez-Villanueva I, Gutierrez M, Hispan P, Betlloch I, Pascual JC (2018) Novel POFUT1 mutation associated with hidradenitis suppurativa-Dowling-Degos disease firm up a role for Notch signalling in the pathogenesis of this disorder. Br J Dermatol178: 984-986.[crossref]
  34. Li A, Peng Y, Taiclet LM, Tanzi RE (2019) Analysis of hidradenitis suppurativa-linked mutations in four genes and the effects of PSEN1-P242LfsX11 on cytokine and chemokine expression in macrophages. Hum Mol Genet 28: 1173-1182.[crossref]
  35. Allard RJV, Vossen KRS, Sigrid MA, Swagemakersn JEMM de Klein, Andrew et al. (2019) A novel nicastrin mutation in a three generation Dutch family with hidradenitis suppurativa: a search for functional significance. Journal of the European Academy of Dermatology and Venereology.
  36. Zhang C, Wang L, Chen L, Ren W, Mei A et al. (2013) Two novel mutations of the NCSTN gene in Chinese familial acne inverse. J Eur Acad Dermatol Venereol 27: 1571-1574.[crossref]
  37. Mauerer A, Betz RC, Pasternack SM, Landthaler M, Hafner C (2010) Generalized solar lentigines in a patient with a history of radon exposure. Dermatology 221: 206-210.[crossref]
  38. Basmanav FB, Oprisoreanu AM, Pasternack SM, Thiele H, Fritz G et al. (2014) Mutations in POGLUT1, encoding protein O-glucosyltransferase 1, cause autosomal-dominant Dowling-Degos disease. Am J Hum Genet 94: 135-143.[crossref]
  39. Vural S, Gundogdu M, Gokpinar Ili E, Durmaz CD, Vural A et al. (2019) Association of pyrin mutations and autoinflammation with complex phenotype hidradenitis suppurativa: a case-control study. Br J Dermatol 180: 1459-1467.[crossref]
  40. Bai Y, Dai X, Harrison AP, Chen M (2015) RNA regulatory networks in animals and plants: a long noncoding RNA perspective. Brief Funct Genomics 14: 91-101.[crossref]
  41. Sun L, Zhao L, Yang G, Yan C, Zhou R et al. (2015)Structural basis of human gamma-secretase assembly. Proc Natl Acad Sci U S A 112: 6003-6008.[crossref]
  42. Bai XC, Rajendra E, Yang G, Shi Y, Scheres SH (2015) Sampling the conformational space of the catalytic subunit of human gamma-secretase. Elife4.[crossref]
  43. Pink AE, Simpson MA, Desai N, Trembath RC, Barker JNW (2013) gamma-Secretase mutations in hidradenitis suppurativa: new insights into disease pathogenesis. J Invest Dermatol 133: 601-607.[crossref]
  44. Cao L, Morales-Heil DJ, Roberson EDO (2019) Nicastrin haploinsufficiency alters expression of type I interferon-stimulated genes: the relationship to familial hidradenitis suppurativa. Clin Exp Dermatol, 44: e118-e125.[crossref]
  45. He Y, Xu H, Li C, Zhang X, Zhou P et al. (2019) Nicastrin/miR-30a-3p/RAB31 Axis Regulates Keratinocyte Differentiation by Impairing EGFR Signaling in Familial Acne Inversa. J Invest Dermatol, 139: 124-134.[crossref]
  46. Zhang X, Sisodia SS (2015) Acne inversa caused by missense mutations in NCSTN is not fully compatible with impairments in Notch signaling. J Invest Dermatol 135: 618-20.[crossref]
  47. Frew JW, Navrazhina K (2020) No Evidence that Impaired Notch Signaling Differentiates Hidradenitis Suppurativa From Other Inflammatory Skin Diseases. Br J Dermatol182:1042-1043[crossref]
  48. Nicola NA, Babon JJ (2015) Leukemia inhibitory factor (LIF). Cytokine Growth Factor Rev26(5):533-44.[crossref]
  49. Cantrell MA, Anderson D, Cerretti DP, Price V, McKereghan K et al. (1985) Cloning, sequence, and expression of a human granulocyte/macrophage colony-stimulating factor. Proc Natl Acad Sci U S A 82: 6250-6254.[crossref]
  50. Wolf SF, Sieburth D, Sypek J (1994) Interleukin 12: a key modulator of immune function. Stem Cells 12: 154-168. [crossref]
  51. Plikus MV, Mayer JA, de la Cruz D, Baker RE, Maini PK (2008) Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature 451: 340-344.[crossref]
  52. Li A, Tanzi RE (2012) Pleiotropy of presenilins. Hereditary Genetics 1: e105.
  53. Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom J et al. (2001) Practice parameter: diagnosis of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology56: 1143-1153.[crossref]
  54. Gianni D, Li A, Tesco G, McKay KM, Moore J et al. (2010) Protein aggregates and novel presenilin gene variants in idiopathic dilated cardiomyopathy. Circulation121: 1216-1226.[crossref]
  55. Frew JW, Navrazhina K (2019) In silico Analysis of Gamma-Secretase-Complex Mutations in Hidradenitis Suppurativa Demonstrates Disease-Specific Substrate Recognition and Cleavage Alterations. Front Med (Lausanne)6: 206.[crossref]
  56. Li A, Zhou C, Moore J, Zhang P, Tsai TH et al. (2011) Changes in the expression of the Alzheimer’s disease-associated presenilin gene in drosophila heart leads to cardiac dysfunction. Curr Alzheimer Res 8: 313-322.[crossref]
  57. Newman M, Wilson L, Verdile G, Lim A, Khan I et al. (2014) Differential, dominant activation and inhibition of Notch signalling and APP cleavage by truncations of PSEN1 in human disease. Hum Mol Genet 23: 602-617.[crossref]
  58. Tobinick E, Gross H, Weinberger A, Cohen H (2006) TNF-alpha modulation for treatment of Alzheimer’s disease: a 6-month pilot study. MedGenMed 8: 25.[crossref]
  59. Butchart J, Brook L, Hopkins V, Teeling J, Puntener U et al. (2015) Etanercept in Alzheimer disease: A randomized, placebo-controlled, double-blind, phase 2 trial. Neurology 84: 2161-2168.[crossref]
  60. Servian-Morilla E, Takeuchi H, Lee TV, Clarimon J, Mavillard F et al. (2016) A POGLUT1 mutation causes a muscular dystrophy with reduced Notch signaling and satellite cell loss. EMBO Mol Med 8: 1289-1309.[crossref]
  61. Li M, Cheng R, Liang J, Yan H, Zhang H et al. (2013) Mutations in POFUT1, encoding protein O-fucosyltransferase 1, cause generalized Dowling-Degos disease. Am J Hum Genet92: 895-903.[crossref]
  62. Montagne A, Zhao Z, Zlokovic BV (2017) Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J Exp Med 214: 3151-3169.[crossref]
  63. Hessam S, Gambichler T, Skrygan M, Sand M, Ruddel I et al. (2018) Reduced ten-eleven translocation and isocitrate dehydrogenase expression in inflammatory hidradenitis suppurativa lesions. Eur J Dermatol  28: 449-456. [crossref]
  64. Phan K, Charlton O, Smith SD (2019) Hidradenitis suppurativa and diabetes mellitus: updated systematic review and adjusted meta-analysis. Clin Exp Dermatol 44: e126-e132.[crossref]
  65. Hessam S, Sand M, Skrygan M, Gambichler T, Bechara FG (2017) Expression of miRNA-155, miRNA-223, miRNA-31, miRNA-21, miRNA-125b, and miRNA-146a in the Inflammatory Pathway of Hidradenitis Suppurativa. Inflammation 40: 464-472.
  66. Liu M, Degner J, Georgantas RW, Nader A, Mostafa NM et al. (2020) A Genetic Variant in the Bcl2 Gene Associates with Adalimumab Response in Hidradenitis Suppurativa Clinical Trials and Regulates Expression of Bcl2. J Invest Dermatol140: 574-582.[crossref]
  67. Liu M, Degner J, Davis JW, Idle KB, Nader A et al. (2018) Identification of HLA-DRB1 association to adalimumab immunogenicity. PLoS One 13: e0195325. [crossref]
  68. Jorgensen AR, Thomsen SF, Karmisholt KE, RingH C (2020) Clinical, microbiological, immunological and imaging characteristics of tunnels and fistulas in hidradenitis suppurativa and Crohn’s disease. Exp Dermatol29:118-123.[crossref]
  69. Mozeika E, Pilmane M, Nurnberg BM, Jemec GB (2013) Tumour necrosis factor-alpha and matrix metalloproteinase-2 are expressed strongly in hidradenitis suppurativa. Acta Derm Venereol 93: 301-314.[crossref]
  70. Byrd AS, Carmona-Rivera C, O’Neil LJ, Carlucci PM, Cisar C et al. (2019) Neutrophil extracellular traps, B cells, and type I interferons contribute to immune dysregulation in hidradenitis suppurativa. Sci Transl Med 11.[crossref]
  71. Xie L, Huang Z, Li H, Liu X, Zheng S et al. (2019) IL-38: A New Player in Inflammatory Autoimmune Disorders. Biomolecules 9. [crossref]
  72. Bernardini N, Skroza N, Tolino E, Mambrin A, Anzalone A et al. (2020) IL-17 and its role in inflammatory, autoimmune, and oncological skin diseases: state of art. Int J Dermatol59:406-411.[crossref]
  73. Thomi R, Cazzaniga S, Seyed Jafari SM, Schlapbach C, Hunger RE (2018) Association of Hidradenitis Suppurativa With T Helper 1/T Helper 17 Phenotypes: A Semantic Map Analysis. JAMA Dermatol154: 592-595.[crossref]
  74. Scala E, Di Caprio R, Cacciapuoti S, Caiazzo G, Fusco A et al. (2019) A new T helper 17 cytokine in hidradenitis suppurativa: antimicrobial and proinflammatory role of interleukin-26. Br J Dermatol181: 1038-1045.[crossref]
  75. Thomi R, Schlapbach C, Yawalkar N, Simon D, Yerly D et al. (2018) Elevated levels of the antimicrobial peptide LL-37 in hidradenitis suppurativa are associated with a Th1/Th17 immune response. Exp Dermatol27: 172-177.[crossref]
  76. Di Caprio R, Balato A, Caiazzo G, Lembo S, Raimondo A et al. (2017) IL-36 cytokines are increased in acne and hidradenitis suppurativa. Arch Dermatol Res 309: 673-678.[crossref]
  77. Hotz C, Boniotto M, Guguin A, Surenaud M, Jean-Louis F et al. (2016) Intrinsic Defect in Keratinocyte Function Leads to Inflammation in Hidradenitis Suppurativa. J Invest Dermatol 136: 1768-1780.[crossref]
  78. Manfredini M, Giuliani AL, Ruina G, Gafa R, Bosi C et al. (2019) The P2X7 Receptor Is Overexpressed in the Lesional Skin of Subjects Affected by Hidradenitis Suppurativa: A Preliminary Study. Dermatology27: 1-8.[crossref]
  79. Coates M, Mariottoni P, Corcoran DL, Kirshner HF, Jaleel T et al. (2019) The skin transcriptome in hidradenitis suppurativa uncovers an antimicrobial and sweat gland gene signature which has distinct overlap with wounded skin. PLoS One 14: e0216249.[crossref]
  80. Shanmugam VK, Jones D, McNish S, Bendall ML, Crandall KA (2019) Transcriptome patterns in hidradenitis suppurativa: support for the role of antimicrobial peptides and interferon pathways in disease pathogenesis. Clin Exp Dermatol44:882-892.[crossref]
  81. Hoffman LK, Tomalin LE, Schultz G, Howell MD, Anandasabapathy N et al. (2018) Suarez-Farinas, M.; Lowes, M. A., Integrating the skin and blood transcriptomes and serum proteome in hidradenitis suppurativa reveals complement dysregulation and a plasma cell signature. PLoS One 13: e0203672.[crossref]
  82. Grand D, Navrazhina K, Frew JW (2019) Integrating Complement into the Molecular Pathogenesis of Hidradenitis Suppurativa. Exp Dermatol.
  83. Guet-Revillet H, Jais JP, Ungeheuer MN, Coignard-Biehler H, Duchatelet S et al. (2017) Join-Lambert, O., The Microbiological Landscape of Anaerobic Infections in Hidradenitis Suppurativa: A Prospective Metagenomic Study. Clin Infect Dis65: 282-291.[crossref]
  84. Hispan P, Murcian O, Gonzalez-Villanueva I, Frances R, Gimenez P et al. (2019) Identification of bacterial DNA in the peripheral blood of patients with active hidradenitis suppurativa. Arch Dermatol Res312:159-163.[crossref]
  85. Jones D, Banerjee A, Berger PZ, Gross A, McNish S et al. (2018) Inherent differences in keratinocyte function in hidradenitis suppurativa: Evidence for the role of IL-22 in disease pathogenesis. Immunol Invest 47: 57-70.[crossref]
  86. Dany M, Elston D (2017) Gene expression of sphingolipid metabolism pathways is altered in hidradenitis suppurativa. J Am Acad Dermatol 77: 268-273.[crossref]
  87. Salomon J, Piotrowska A, Matusiak L, Dziegiel P, Szepietowski JC (2019) Chitinase-3-like Protein 1 (YKL-40) Is Expressed in Lesional Skin in Hidradenitis Suppurativa. In Vivo 33: 141-143.
  88. Miller I, Lynggaard CD, Lophaven S, Zachariae C, Dufour DN et al. (2011) A double-blind placebo-controlled randomized trial of adalimumab in the treatment of hidradenitis suppurativa. Br J Dermatol 165: 391-398.[crossref]
  89. Kimball AB, Okun MM, Williams DA, Gottlieb AB, Papp KA et al. (2016) Two Phase 3 Trials of Adalimumab for Hidradenitis Suppurativa. N Engl J Med 375: 422-34.[crossref]
  90. van der Zee HH, Longcore M, Geng Z, Garg A (2019) Weekly adalimumab treatment decreased disease flare in hidradenitis suppurativa over 36 weeks: integrated results from the phase 3 PIONEER trials. J Eur Acad Dermatol Venereol.[crossref]
  91. Caposiena Caro RD, Cannizzaro MV, Tartaglia C, Bianchi L (2019) Clinical response rate and flares of hidradenitis suppurativa in the treatment with adalimumab. Clin Exp Dermatol.[crossref]
  92. Zouboulis CC, Hansen H, Caposiena Caro RD, Damiani G, Delorme I et al. (2020) Adalimumab Dose Intensification in Recalcitrant Hidradenitis Suppurativa/Acne Inversa. Dermatology236:25-30[crossref]
  93. Grant A, Gonzalez T, Montgomery MO, Cardenas V, Kerdel FA (2010) Infliximab therapy for patients with moderate to severe hidradenitis suppurativa: a randomized, double-blind, placebo-controlled crossover trial. J Am Acad Dermatol 62: 205-217.[crossref]
  94. Yang JQ, Wu XJ, Dou TT, Jiao T, Chen XB et al. (2015) Haploinsufficiency caused by a nonsense mutation in NCSTN underlying hidradenitis suppurativa in a Chinese family. Clin Exp Dermatol 40: 916-9.[crossref]
  95. Nomura Y, Nomura T, Suzuki S, Takeda M, Mizuno O et al. (2014) A novel NCSTN mutation alone may be insufficient for the development of familial hidradenitis suppurativa.J Dermatol Sci 74: 180-182.[crossref]

Article Type

Review Article

Publication history

Received: March 29, 2020;
Accepted: April 06, 2020;
Published: April 22, 2020

Citation

Airong Li and Rudolph E. Tanzi (2020) Genetics of Hidradenitis Suppurativa. J Mol Genet Volume 3(2): 1-7. DOI: 10.31038/JMG.2020324

Corresponding authors

Airong Li MD PhD,
Genetics and Aging Research Unit Massachusetts General Hospital,
Harvard Medical School,
114, 16th Street MA 02129,
US
Tel: 6177249397

Rudolph E. Tanzi PhD,
Genetics and Aging Research Unit Massachusetts General Hospital,
Harvard Medical School,
114, 16th Street MA 02129,
US
Tel: 6177266845
Fax: 617-724-1823