Monthly Archives: February 2020

Internal Loop Recording of Prolonged (39 Second) Sinus Pause Causing Syncope

DOI: 10.31038/JCCP.2020312

Abstract

Presentation: A 53 year old woman presented with a syncopal episode which occurred whilst sitting in bed reading, witnessed by her husband. She was unrousable for maybe half a minute. She had no other medical history, clinical examination was unremarkable, as were multiple investigations.

Diagnosis: A loop recorder was inserted without difficulty. A few weeks later she again blacked out. Her loop recorder showed asystole for 39 seconds, followed by a return to sinus bradycardia and then sinus rhythm, accompanied by recovery of consciousness.

Treatment: She went on to have a dual chamber pacemaker inserted. At review after six weeks, six months and one year she had had no further syncopal episodes.

Conclusion: This case reports one of the longest documented episodes of asystole with spontaneous recovery and serves as a reminder of the utility of internal loop recorders in the investigation of syncope.

Introduction

This case reports a patient with syncope in whom an internal loop recorder revealed one of the longest documented episodes of asystole with spontaneous recovery.

Case Report

A 53 year old woman presented with a syncopal episode which occurred whilst sitting in bed reading, witnessed by her husband. She was unrousable for maybe half a minute. She was a non-smoker, on no medications. Her father had a pacemaker inserted in his 70s, for slow atrial fibrillation; there was no family history of sudden adult death. She had no other clinical history; specifically, nothing to suggest sarcoidosis, haemochromatosis or other deposition disorders. Clinical examination was unremarkable. Multiple investigations, including echocardiography, electroencephalography and Holter monitoring, were essentially normal, as was brain magnetic resonance imaging. A Medtronic Reveal LINQ™ loop recorder was inserted without difficulty. A few weeks later she awoke as usual, went to the toilet, micturated and returned to bed feeling fine. Whilst lying in bed, she again blacked out. She recovered consciousness a while later, and felt nauseated with a slight headache.  That morning she was contacted by the hospital and advised to come in directly. Her loop recorder showed prolonged asystole!  Figure 1. She had gradually developed sinus bradycardia and then sinus arrest. After 39.4 seconds she returned to sinus bradycardia and then sinus rhythm without any compensatory tachycardia. There was no ventricular escape rhythm, simply a sinus pause with no rescue. The next day she was brought to the operating room for pacemaker insertion under local anaesthetic. Just after left subclavian vein cannulation, she had another 20 second pause, though she recovered before external pacing could be initiated. She regained consciousness and went on to have a dual chamber pacemaker inserted, with good thresholds in the right ventricular apex and right atrial appendage. At review after six weeks, six months and one year she had had no further syncopal episodes.

JCCP-2020-303_G. Pate_F1

Figure 1. Internal loop recorder tracing of 39.4 second asystolic pause.

Discussion

Loop recorders have proved very useful in documenting previously unrecognized arrhythmias, particularly intermittent pauses [1]. Current guidelines recommend loop recorder insertion for any unexplained syncope [2]. In the event of documentation of any symptomatic pauses greater than 3 seconds, or nocturnal pauses greater than 6 seconds, pacemaker insertion is recommended [3]. Anything more than 10 seconds is described as a very long pause. Multiple papers have documented pauses of 10 seconds or more [4–7], including one of 44 seconds [8]. During a tilt table test one paper described asystole for 72 seconds [9], but this was a provoked pause and cardiopulmonary resuscitation was commenced so this cannot truly be described as spontaneous recovery of rhythm. Another consideration here was of artefact, a recognized issue with loop recorders [10]. However, the patient’s clinical picture was consistent with the observed pause, she had not been undertaking any activity that might have produced an artefactual pause, and the fact she had another witnessed pause of 20 seconds in the hospital confirm that the observed pause was real.

No cause was identified for her pauses. However, she did not have tilt-table testing, electrophysiological studies or cardiac MRI scanning prior to pacemaker insertion; regardless, management would still have involved pacemaker insertion. This case reports one of the longest documented episodes of asystole with spontaneous recovery and provides a very graphic reminder of the utility of internal loop recorders in the investigation of syncope.

References

  1. Maggi R, Rafanelli M, Ceccofiglio A, Solari D, Brignole M, et al. (2014) Additional diagnostic value of implantable loop recorder in patients with initial diagnosis of real or apparent transient loss of consciousness of uncertain origin. Europace 16: 1226–1230. [Crossref]
  2. Varosy PD, Chen LY, Miller AL, Noseworthy PA, Slotwiner DJ, et al. (2017) Pacing as a Treatment for Reflex-Mediated (Vasovagal, Situational, or Carotid Sinus Hypersensitivity) Syncope: A Systematic Review for the 2017 ACC/AHA/HRS Guideline for the Evaluation and Management of Patients With Syncope: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol 70: 664–679. [Crossref]
  3. European Society of C, European Heart Rhythm A, Brignole M, Auricchio A, Baron-Esquivias G, et al. (2013) 2013 ESC guidelines on cardiac pacing and cardiac resynchronization therapy: the task force on cardiac pacing and resynchronization therapy of the European Society of Cardiology (ESC). Developed in collaboration with the European Heart Rhythm Association (EHRA). Europace 15: 1070–1118. [Crossref]
  4. Mairesse GH, Marchand B (2003) Prolonged asymptomatic sinus pause indicated by implantable loop recording. Heart 89: 244. [Crossref]
  5. Zaidi A, Clough P, Mawer G, Fitzpatrick A (1999) Accurate diagnosis of convulsive syncope: role of an implantable subcutaneous ECG monitor. Seizure 8: 184–186. [Crossref]
  6. Deharo JC, Jego C, Lanteaume A, Djiane P (2006) An implantable loop recorder study of highly symptomatic vasovagal patients: the heart rhythm observed during a spontaneous syncope is identical to the recurrent syncope but not correlated with the head-up tilt test or adenosine triphosphate test. J Am Coll Cardiol 47: 587–593. [Crossref]
  7. Menozzi C, Brignole M, Garcia-Civera R, Moya A, Botto G, et al. (2002) Mechanism of syncope in patients with heart disease and negative electrophysiologic test. Circulation 105: 2741–2745.
  8. Kanjwal K, Karabin B, Kanjwal Y, Grubb BP (2010) A case of mistaken identity: asystole causing convulsions identified using implantable loop recorder. Int J Med Sci 7: 209–212.
  9. Leftheriotis DI, Theodorakis GN, Kremastinos DT (2003) Prolonged asystole during head-up tilt testing with clomipramine infusion. Europace 5: 313–315. [Crossref]
  10. Ali H, Sorgente A, Daleffe E, Cappato R (2014) Asystole detected by implantable loop recorders: true or false? Ann Noninvasive Electrocardiol 19: 595–597. [Crossref]

Bladder Necrosis Due to Septic Shock

DOI: 10.31038/SRR.2020311

Abstract

Background: Chronic constipation leading to fecal impaction and stercoral perforation is an important cause of morbidity and mortality in the aging population. Gangrenous cystitis, an even more rare entity, previously reported primarily in obstetric cases, has not been reported in association with stercoral perforation until this report.

Summary: This case report details the clinical presentation and treatment of a 61-year-old female with history of end-stage renal disease and history of kidney transplantation who presented in septic shock with a positive urinalysis. She underwent an emergent computed topography scan of the abdomen and pelvis which demonstrated a perforation originating from the sigmoid colon. Damage control surgery was performed, the sigmoid colon was resected, and the intestine was left in discontinuity. The abdomen was left open with a temporary abdominal dressing and a catheter for direct peritoneal resuscitation. She returned for a “second look” laparotomy where ischemic bladder tissue was noted. A partial cystectomy was performed, and the bladder was reconstructed over a Foley catheter and her abdomen remained open for continued direct peritoneal resuscitation. Ultimately, the
family determined that this level of care was not consistent with the patient’s wishes and she was made comfort measures.

Conclusion: This is the first report of stercoral perforation of the sigmoid colon in association with gangrenous cystitis in the literature. This case report highlights the importance of a thorough evaluation of the pelvis during surgical management of stercoral perforation so as not to miss concurrent gangrenous cystitis.

Keywords

sepsis, sterocoral perforation, gangrenous cystitis

Introduction

Gangrenous cystitis is a rare condition that has only been reported in the literature a total of 240 times worldwide since 1934. It was once seen in obstetric cases, associated with labor and delivery, but is now most commonly secondary to pelvic radiation/surgery, chemotherapeutic agents, urinary retention, urosepsis, pelvic thrombophlebitis, and colovesical fistulae. The pathogenesis is unknown but is thought to be initiated by bladder ischemia that is then subject to microbial superinfection [1] . Fecal impaction secondary to chronic constipation can lead to Stercoral Perforation (SP) through pressure necrosis of the large bowel [2]. SP is a well-defined entity in the literature that is associated with significant morbidity and mortality and has also been associated with a prior history of renal transplantation [3]. This case report is the first to identify SP of sigmoid colon as a cause of gangrenous cystitis.

Case History/Examination

The patient is a 61-year-old female with a history of End Stage Renal Disease (ESRD) and remote kidney transplantation, with recent admission to the hospital for intertrochanteric fracture after fall. She was subsequently discharged to a rehab facility after a surgical repair of the fracture. The patient presented to our hospital two weeks later in septic shock. On physical examination she had an altered mental status, she was in respiratory distress, and profoundly hypotensive. The abdominal examination was remarkable for a soft but distended abdomen and a palpable transplant kidney in the right lower quadrant with no peritoneal signs.

Investigations and Treatment

White blood cell count was 11,000 with 94% neutrophils and serum lactate was found to be 6.1 cells per cubic millimeter of blood. A Urinalysis (UA) was grossly positive Urinary Tract Infection (UTI) so the patient was transferred to the Medical Intensive Care Unit (ICU) for suspected urosepsis. The patient was intubated and fluid resuscitation along with empiric broad spectrum antibiotics were initiated. An emergent Computed Tomography (CT) scan of the abdomen and pelvis was obtained and showed radiological evidence of perforated viscous, most likely from the sigmoid colon (Figure 1).

SRR 20 - 101_Joshua A Bloom_F1

Figure 1. CT scan of the abdomen and pelvis showing significant stranding and multiple locules of air outside the bowel lumen in the left lower quadrant adjacent to the sigmoid colon and along the left paracolic gutter.

An emergent surgical consult was obtained and the patient was taken immediately to the operating room for exploratory laparotomy. Upon entry to the abdomen, there was foul smelling, turbid fluid and an ischemic, dilated proximal sigmoid colon was encountered. A stercoral perforation of the sigmoid colon on the mesenteric border into the retroperitoneum was noted. The left side of the retroperitoneum had a large amount of necrotic tissue extending into the pelvis.

A damage control surgery was performed. The sigmoid colon was resected, and the intestine was left in discontinuity. The left side of the retroperitoneum and pelvis were opened, and debridement of the necrotic retroperitoneal tissue was performed. A 19 Fr blake drain was placed intra peritoneally for direct peritoneal resuscitation [4, 5]. At the end of the case, the patient was transferred to the Surgical ICU for further resuscitation and correction of metabolic abnormalities.

Postoperatively, the patient’s hemodynamics improved transiently, and vasopressor requirement did decrease somewhat but her serum lactate level continued to rise. With a new elevation of serum lactate the patient was taken to the operating room for a “second look” laparotomy approximately 24 hours later. No further bowel ischemia or necrosis was noted. There was still a significant amount of retroperitoneal necrotic tissue, especially in the pelvis. Further dissection into the pelvis did reveal a 7 cm cystostomy with ischemic bladder tissue mostly on the left side with clear demarcation of ischemic changes. There was some ischemia of the right side of the bladder as well. A Urology and Transplant Surgery consultation were obtained due her history of kidney transplant.

Outcome and Follow-Up

A multidisciplinary discussion resulted in the decision to perform a partial cystectomy and not explant the donor kidney. All of the obvious necrotic tissue was then resected, and the bladder was reconstructed over a Foley catheter. Afterwards, since the patient still was in a profound state of shock, the patient’s bowel was left in discontinuity, the abdomen was temporarily closed, and the patient was transferred back to the Surgical ICU for further management.

The next day the patient underwent a planned relaparotomy with abdominal wash out and temporary abdominal closure. No further areas of bowel or bladder necrosis were noted. Due to the patient’s extensive comorbidities and lack of improvement over the following 48 hours as well as the extent of the care she would continue to require, the family felt this was not consistent with her wishes and elected to make her comfort measures. The patient passed away shortly thereafter.

Discussion

This is the first case report of bladder necrosis due to stercoral perforation of the sigmoid colon. With no literature to guide us, it is difficult to make absolute conclusions, but we believe that the stercoral perforation into the retroperitoneum led to an inflammatory reaction and local infection around the bladder leading to ischemia and gangrenous cystitis.

In the literature, there are numerous causes of bladder necrosis published, mostly in the pre-antibiotic era. These causes include prolonged labor, pelvic radiation, chemotherapy, urinary retention, urosepsis, and pelvic thrombophlebitis to name a few. In these published cases, the mainstay of treatment has been early antibiotic therapy and surgical treatment with extensive debridement of necrotic bladder and wide drainage [6, 7].

In this case report, we differed from the traditional management in that we performed a damage control laparotomy with temporary abdominal closure and direct peritoneal resuscitation. This strategy allowed for source control in the OR and rapid transfer to the ICU for further resuscitation and correction of metabolic abnormalities. This provided our patient with the best chance of survival. Ultimately, it was her wishes that she would not want to live if she were not guaranteed to return to her former quality of life, and she was made comfort measures. However, we believe that she most likely would have survived this hospitalization had this been in line with her wishes.

Conclusion

Whether this patient’s bladder necrosis was due to her stercoral ulcer or her septic shock remains unclear as the literature is equally vague regarding both topics. However, surgeons should be aware of this phenomenon and while operating on the bowel emergently for perforation, should take the time to assess the pelvis, especially in patients with a grossly positive urinalysis.

Lessons Learned

Stercoral perforation of the sigmoid colon leading to septic shock can be associated with gangrenous cystitis and necessitates a thorough evaluation of the pelvis while operating on this entity.

References

  1. De Rosa A, Amer T, Waraich B, Bello A, Parkinson R (2011) Gangrenous cystitis in a 42-year-old male. BMJ Case Reports  2011: 1–4. [Crossref]
  2. Chakravartty S, Chang A, Nunoo-Mensah V (2013) A systematic review of stercoral perforation. Colorectal Disease 15: 930–935. [Crossref]
  3. Dubinsky I (1996) Stercoral Perforation of the Colon: Case Report and Review of the Literature. Journal of Emergency Medicine 14: 323–325. [Crossref]
  4. Rai R, Sikka P, Aggarwal N, Shankaregowda SA (2015) Gangrenous Cystitis in a Woman Following Vaginal Delievery: An Uncommon Occurrence -A Case Report. Journal of Clinical and Diagnostic Reasearch 9: 13–14.
  5. Smith JW, Neal Garrison R, Matheson PJ, Harbrecht BG, Benns MV, et al. (2014) Adjunctive treatment of abdominal catastrophes and sepsis with direct peritoneal resuscitation: indications for use in acute care surgery. J Trauma Acute Care Surg 77: 393–398 [Crossref]
  6. Hinev A, Anakievski D, Krasnaliev I (2010) Gangrenous Cystitis: Report of a Case and Review of the Literature. Urologia Internationalis 85: 479–481. [Crossref]
  7. Piraprez M, Ben Chehida M, Fillet M (2017) Case Report: Emphysematous cystitis. Red Med Liege 384–387.

Bronchoscopy: A not-so-innocent invasive examination

DOI: 10.31038/NAMS.2020311

Abstract

Bronchoscopy has for many years been the only invasive examination used to screen for bronchus in real time, remove objects that have entered the airway, healing properties, and finally a sample including missing tissue for biopsy for the right follow up medical evaluation and treatment. The usual examination is performed by a flexible bronchoscope which does not cause much discomfort to the patient and causes few complications, complications such as bleeding, cardiac arrhythmias, fever, pneumonia, death, introduction and exacerbation of infection, etc.

This short research is about to overturn this nice and enjoyable environment that has been created regarding the safety of this invasive examination that will eventually and happily very soon be replaced. The present short research is here to overthrow the benefits of bronchoscopy and present the reality as this has not been presented to patients before undergoing an invasive examination that may not eventually need to be done.

Dedication: This short research is dedicated to my Father Panagiotis, who recently died.

Keywords

bronchoscopy, hemoptysis, death, complications, pneumonia

Introduction

Very easily, mainly after a CT scan, a PET/CT has appeared in our lives to improve the quality of imaging data, also including the case but not always the lack of knowledge, recommend that patients undergo an invasive bronchoscopy. that the diagnostic process in short time will end, and the patient is transferred to the next stage of treatment, but if the patient never reaches the treatment stage and is instead hospitalized with complications? according to the literature, it is reported that bronchoscopy is performed by an experienced physician, how can the patient know if the physician is experienced or not?

How can the patient know if during the examination they will not contribute to the examination and trainees which is prohibitive in certain coexisting lung diseases where particular experience is required, such as a patient with bronchectasis, COPD, tec, who they need bronchoscopy from a very experienced physician. Is the procedure is followed of informing the patient prior to bronchoscopy about the pros and cons of the examination as well as their complications? Finally, is it worth doing a bronchoscopy which, depending on the clinical picture of the patient, may eventually have the opposite effect? bronchoscopy is not for everyone.

Analysis

Bronchoscopy is an invasive examination performed in public and private hospitals as well as in private diagnostic centers and clinics. Before conducting the examination blood tests are performed, including blood and cardiology tests, so, how many tests for a diagnostic examination that is considered safe? The complications that it presents contradict the advantages, advantages that in cases depending on the severity of the patient are disadvantages.

Advantages that become disadvantages when one of the following reasons are met.

  1. Inexperienced Invasive physician
  2. Bronchoscopy by trainees (depends on case).
  3. Inexperienced staff.
  4. Hygiene conditions.
  5. Disinfection, cleaning, and sterilization of a bronchoscope.
  6. The clinical picture of the patient.
  7. Consideration of medical examinations to avoid bronchoscopy.
  8. Inform the patient about the complications of bronchoscopy.

When one or more of the above is not observed, then the patient is at high risk, as the complications of the bronchoscopy examination can cause the patient even more serious damage than the disease itself led the patient to bronchoscopy. What safe examination are they talking about?

Is there a mortality rate of about 0.5% these patients were aware of the complications? Should they have done bronchoscopy? have the hygiene rules been adhered to or fallen into a category of the above 8 listed? Although international literature indicates guidelines for safe bronchoscopy examinations, does the patient know whether the guidelines are being followed?

One of the major complications of bronchoscopy is bleeding, ranges for example from low 10ml to 150ml which is very high. It is a serious complication that needs to be addressed immediately since its origin is complex, it can come from many areas such as pulmonary capillaries, pulmonary arteries, pulmonary veins, and finally large thoracic vessels. Bleeding is directly related to the biopsy, in most cases bronchoscopy is performed to obtain a biopsy, an experienced physician in a burdensome patient with a history who knows before, noted here the bronchoscopy requires the patient to hold already a CT, which will require a CT scan. is the one who will judge it must be done, if it is to be done, to pay attention, what requires observation, coexisted, possible complications, time of examination in relation to the severity of the patient, the participation of trainees or not, and how to get a sample.

There are two types of sample deficiencies: endobronchial and transobronchial biopsy, transobronchial biopsy causes the patient and most bleeding, here is a big question, in patients with bronchiectasis, or similar medical cases that usually occur in these medical cases colonies and bacteria such as pseudomonas aeruginosa that are usually resistant to antibiotics, a transobronchial biopsy is responsible for burdening the patient’s clinical picture? Thus, correlated the bleeding with the number of biopsies and how they were obtained. Considering the fact that patients with congenital diseases, medicines that are taken for various important reasons, a bleeding can reach or exceed 40% depending on the case of patients undergoing bronchoscopy. So why should they do a bronchoscopy? The treating physician should be able to judge the overall picture of the patient in order to be able to make a correct diagnosis, taking into account patients with a poor medical history considering the negative aspects of a bronchoscopy, however, bronchoscopy is an invasive risk examination depending on the patient’s physical condition and medical history. Bronchoscopy examination is not for everyone.

It is concluded that the bleeding of the patient after bronchoscopy implies with further clinical progress, although the incidence of sudden death is small as has been mentioned above. Small bleeding can be easily treated large bleeding to stop may require laser and even surgery, the patient must consider all options and exhaust all parameters. It is worth considering whether it is worth the effort, pain and risk to perform a bronchoscopy examination that no one can guarantee his or her physical situation after the examination is completed.

Most complications that have occurred during the examination or thereafter have to do with how to obtain and receive biopsy material. Such cases are brushes that have broken down or have not worked properly. Continuous aspiration during the examination may lead to hypoxemia and atelectasis. Pseudomonas aeruginosa is the most frequently transmitted organism from an inappropriate bronchoscope (see above 8 reasons) which has not been properly disinfected and cannot be used. Using such a bronchoscope is a criminal act against the patient. During bronchoscopy, patients have been observed to have increased blood pressure and increased heart rate, this is due to increased oxygen demand from the myocardium which may then lead the patient to arrhythmias and ischemia. Cardiac arrhythmias that occur after bronchoscopy confess hypoxemia. Particularly in COPD patients with hypercapnia hypoxia is contraindicated for flexible bronchoscopy. It is worth noting at this point that complications of bronchoscopy often occur when there are concomitant diseases such as pneumonia or tumor, as well as the type of forceps to be used, that is toothed or non-serrated forceps. In patients with chronic bronchiectasis, the physician should consider all indications for the overall picture of the patient and whether or not to perform the bronchoscopy. It is the physician that will analyze the patient’s respiratory function and the degree of criticality. It should also be borne in mind that some patients with infections will need antibiotics after bronchoscopy, these patients likely to need antibiotics that will be able to deal with germs and bacteria that are resistant. These patients are more likely to develop febrile episodes as well as critical respiratory function.

The physician is solely responsible for analyzing the pros and cons, it is the one who weighs the benefit of the risk. Patients with bronchiectasis should not be bronchoscopied by trainees or by physicians who rarely perform bronchoscopy. When these patients show haemoptysis, the detection of bleeding is crucial. Finally, bronchoscopy in COPD patients is particularly burdensome compared to non-COPD patients, COPD patients should be evaluated on the findings of their clinical trials together with medical evacuation before performing the bronchoscopy. Assessment of potential advantages over advantages must be evaluated.

Conclusion

Bronchoscopy is a conditional safe examination when all safety rules are met, medical evaluation is performed separately for each patient, each patient is different. Bronchoscopy is not for all patients, generally described as a safe invasive examination but with so many complications that there are people who have had a bronchoscopy and have not returned home, is this safe medical examination they say? it is not a safe medical examination, it is a safe conditional examination, evaluating the clinical picture of the patient in combination with the degree of experience of the physician and the clinical picture of the patient judging its effectiveness.

Patients with co-existing health problems should be assessed for the advantages of screening in conjunction with screening complications. Physicians should not be dragged into the altar of the rapid effect of detecting the problem by undergoing bronchoscopy in patients who are not allowed to do a bronchoscopy based on physical or medical pre-existing health conditions. It is a medical examination with uncertain complications sometimes simple, sometimes heavy and sometimes deadly.

Classical bronchoscopy has begun to come to a second myrrh as soon as virtual bronchoscopy is implemented and thus the complications of classical bronchoscopy will be eliminated. Bronchoscopy should be chosen as a medical examination after all other medical examinations and methods have been utilized and under conditions based on the clinical picture of the patient, in strict compliance with the hygiene guidelines.

References

  1. Kovaleva J, Peters FT, van der Mei HC, Degener JE (2013) Transmission of infection by flexible gastrointestinal endoscopy and bronchoscopy. Clin Microbiol Rev 26: 231–254. [Crossref]
  2. Pereira W Jr, Kovnat DM, Snider GL (1978) A prospective cooperative study of complications following flexible fiberoptic bronchoscopy. Chest 73: 813–816. [Crossref]
  3. Kreider ME, Lipson DA (2003) Bronchoscopy for atelectasis in the ICU: a case report and review of the literature. Chest 124: 344–350. [Crossref]
  4. Trouillet JL, Guiguet M, Gibert C, Fagon JY, Dreyfuss D, et al. (1990) Fiberoptic bronchoscopy in ventilated patients. Evaluation of cardiopulmonary risk under midazolam sedation. Chest 97: 927–933. [Crossref]
  5. Hanson RR, Zavala DC, Rhodes ML, Keim LW, Smith JD (1976) Transbronchial biopsy via flexible fiberoptic bronchoscope; results in 164 patients. Am Rev Respir Dis 114: 67–72. [Crossref]
  6. Da Conceiçao M, Genco G, Favier JC, Bidallier I, Pitti R (2000) Fiberoptic bronchoscopy during noninvasive positive-pressure ventilation in patients with chronic obstructive lung disease with hypoxemia and hypercapnia. Ann Fr Anesth Reanim 19: 231–236. [Crossref]

Relationship between polycystic ovarian syndrome (PCOS) and fatty liver disease

DOI: 10.31038/EDMJ.2020413

Abstract

Background – Polycystic ovarian syndrome (PCOS) and fatty liver disease are considered to be off-shoots of insulin resistance. However, the link between the two disorders have recently been found to be variable.

Objective – To explore the association of fatty liver disease with PCOS and to model the association of hyperandrogenism, obesity and insulin resistance indices among subjects with or without PCOS and fatty liver disease.

Methodology – This study was carried out between Jan-2018 to Aug-2019 at PNS HAFEEZ hospital. Odds ratio (OR) for having PCOS with fatty liver disease were calculated. We utilized general linear model (GLM) by keeping hyperandrogenism, abdominal volume index (AVI) and insulin resistance as dependent variables against PCOS and fatty liver disease as independent factors.

Results – OR for having PCOS in subjects with fatty liver disease was 1.34(95%CI: 1.34(0.868-2.065). Using Free Androgen Index(FAI) as dependent variable and fatty liver and PCOS as fixed factors in a GLM analysis, the group wise differences are depicted as: Group-1: No Fatty liver+No PCOS=2.37(+1.30),n=97), Group-2: No PCOS+Fatty Liver=3.35(+2.23),n=66), Group-3: PCOS+No Fatty Liver=6.08(+5.05),n=88), and Group-4: PCOS+Fatty Liver=6.83(+4.70),n=80).(p<0.001). Abdominal obesity was not associated with PCOS (p=0.980), but was significantly associated with fatty liver disease (p<0.001).

Conclusion – Presence of fatty liver disease in females can lead to slightly higher frequency of PCOS. Biochemical and clinical hyperandrogenism were associated with presence of PCOS, while abdominal obesity and insulin resistance were linked with fatty liver disease.

Keywords

Polycystic ovarian syndrome (PCOS), free androgen index (FAI), modified Ferriman-Gallwey score (mFG score), fatty liver and Rotterdam PCOS criteria.

Background

Lipids are considered as the mainstream culprits in pathogenesis of development of polycystic ovarian syndrome (PCOS) and fatty liver disease. [1, 2] Therefore the probability based upon the famous “Common Soil Hypothesis” dealing in directly with metabolic syndrome suggest a common linkage between the two pathogenic disorders. [3] Both PCOS and Fatty liver disease amounts to multiple complications. PCOS has been shown to result in hirsutism and reproductive issues. [4], while fatty liver disease is associated with acceleration liver damage leading to cirrhosis and hepatocellular carcinoma (HCC). [5]

While the mechanism seems more understandable in terms of extra fat accumulation within liver but some evidence indicates PCOS phenotype variability does allow differential association with fatty liver disease. Macut et al have shown that subjects having higher serum testosterone, a hallmark of PCOS are associated with a two-fold increased risk of fatty liver. [6] Rocha et al in their systematic review from 2007 to 2017 highlighted that prevalence of PCOS with NAFLD remains variable, but the combined presence of PCOS and NAFLD results in higher degree of clinical and biochemical hyperandrogenism than the female subjects only demonstrating PCOS with former category more associated with metabolic derangements. [7] Contrary to that there is evidence where both fatty liver disease and PCOS are associated and attributed to common pathogenesis. The research work by Vassilatou et al have demonstrated that lipid based abnormalities are central to the development of PCOS and NAFLD; however, he categorized data from PCOS patients as either being obese or otherwise. [8] Similarly, Kauffman et al have demonstrated that PCOS and NAFLD can occur due to common pathogenic mechanisms including androgen excess and insulin resistance. [9, 2]

So, the obvious question arises as to how fat accumulation or associated metabolic defects differentially affect the two organs? While PubMed data search indicates both fatty liver disease /NASH/NAFLD and PCOS both being the off-shoots of insulin resistance, [10] still there are both genetic and epigenetic  mechanism at work which can probably change the overall phenotype of metabolic syndrome and related diseases. In this regard a study from Poland had identified GG genotype variation to result in higher frequency of NAFLD in PCOS than PCOS subjects without Cannabinoid Receptor-1 gene polymorphism thus highlighting genetic predilections towards PCOS-fatty liver occurrence. [11] Similarly, Jones et al have identified higher risk of fatty liver disease with hyperandrogenism associated PCOS in comparison to female subjects having reduced hyperandrogenism independent of insulin resistance. [12] Bruce et al by highlighting the concept of “Metabolic Circadian Clock System” have attempted to highlight epigenetic factor from environmental triggers which change the phenotypic presentation of metabolic syndrome, and thus can be interpreted indirectly as the effect of regional effects or ethnic triggers. [13] Another aspect of this is the adipocyte size which has been shown to result in variability in hepatic steatosis [14], which have been smaller but with increase adipocyte area in Asian population than Caucasians and so the affect could be due to specific racial reasons [15]. So racial differences due to variability adipocyte area, genetic factors and epigenetic influences need to be considered and may probably be different in whatever has been observed elsewhere. Based upon these newly emerging evidence a compelling need was therefore felt to learn the association between the PCOS, fatty liver disease and androgenicity which were supposedly to be associated due to common trigger i.e., insulin resistance.

A study is therefore planned to explore the association of fatty liver disease, PCOS and hyperandrogenism by estimating the hirsutism, anthropometric, biochemical and endocrine differences (Free androgen index) between subjects with fatty liver associated PCOS and non-fatty liver PCOS.

Methods

We planned a cross-sectional analysis among reproductive-age female subjects after formal approval of hospital’s ethical review committee of PNS HAFEEZ Hospital. The study involved department of radiology, pathology, obstetrics and gynecology. The duration of study was 18 months from Jan-2018 to Aug-2019. Females in reproductive age post 2 years of menarche were initially included as target population. Patient selection was based non-probability convenience mode of sampling. Females who some acute or chronic conditions including pelvic inflammatory disease, metabolic diseases like hypertension, diabetes and ischemic heart disease, autoimmune disorder, on any medication, fertility treatment, or other unknown drugs were excluded from the study. Subjects who could not complete all testing formalities for any reason were excluded from the study. Finally selected females were formally requested to visit pathology department on 2nd day of menstrual cycle Subjects who finally followed up for study in medical fasting were formally

explained about study design, requirements and were asked to sign a consent form. Further clinical evaluation included history, evaluation of hirsutism as per modified Ferriman-Gallwey scores [16], anthropometric measurements, vital signs measurements and a generalized clinical examination for any signs of chronic disease. They were questioned about duration of menstrual cycle for presence or absence of oligo/anovulation as per criteria utilized by Kollmann et al. [17]

Radiological and laboratory analysis: 10 ml (approx.) of blood was collected for fasting plasma glucose, ALT, HbA1c, total testosterone, Sex Hormone Binding Globulin (SHBG) and serum insulin and lipid profile. Following sample collection, females were requested to go for radiological examination for evaluation of reproductive tract examination for presence or absence of polycystic ovarian syndrome as per Rotterdam criteria [18]

Lab analysis and measurements: Fasting plasma glucose, total cholesterol and triglycerides were measured on Selctra-ProM (Clinical chemistry analyzer) by GPO-PAP, CHOD-PAP and GPO-PAP method. LDL-cholesterol and HDL-cholesterol were measured by direct enzymatic methods using accelerator selective detergent on Selectra-proM. ALT was measured by IFCC method at 370C. Total testosterone, SHBG, glycated hemoglobin were analyzed by using chemiluminescent microparticle Immunoassay (CMIA) on ARCHITECT (iSystem) developed by Abbot Diagnostics. Serum insulin was measured on Chemiluminescence method on Immulite® 1000 analyzer. Internal and National External Quality Assurance Program are in place to monitor inter and intra lab imprecision along with internal QC management for precision and accuracy by documented trouble shooting on instrument generated charts as per Westgards’s standard protocols.

Calculated parameters – Free Androgen Index (FAI) was measured as: FAI = (Total testosterone/SHBG) x 100. FAI > 5%was greater than 5% was termed as biochemical hyperandrogenism as per Al Kindi et al. [19] Homeostasis Model Assessment of Insulin resistance (HOMAIR) was measured using Mathew’s et al mathematical model. [20]

Data analysis – We used SPSS-version 24 for statistical analysis. Age was calculated as per mean and SD by descriptive statistics. Descriptive statistics for frequency of PCOS, hirsutism and fatty liver disease % were calculated for subjects with or without fatty liver disease. Inferential statistics involved calculation of Odds Ratio for presence of PCOS as per Rotterdam criteria in subjects with or without fatty liver disease. We utilized General linear models (GLM) where we evaluated sequentially FAI, mFG scores for hirsutism, abdominal volume index (AVI) and insulin resistance (HOMAIR) as dependent variables against presence or absence of PCOS and fatty liver disease as fixed factors.

Results

Mean age among subjects with Rotterdam defined PCOS was (26.71+ 6.99, n=168) and without PCOS was (29.07+ 8.11, n=163). Similarly subjects who had fatty liver had a mean age of (30.95+7.10, n=146) in comparison to females without fatty liver [25.45+7.17, n=185). 236 females in our data set were married, while 95 were non-married. Hirsutism was diagnosed in 157 females while 174 were having a modified FG score of less than 8. 88/185 (47.57%) of the cases without fatty liver disease were diagnosed with PCOS, while 80/146 (54.79%) of cases diagnosed to have fatty liver disease had PCOS indicating no statistically significant association between presence of PCOS and fatty liver disease in our regional data set. Odds ratio (OR) for having PCOS in subjects with fatty liver disease is 1.34 but not statistically significant. (Table-1)

Table 1. Odds ratio for presence of PCOS as per Rotterdam criteria in subjects with fatty liver disease (p=NS).

PCOS

Odds Ratio

(Rotterdam criteria)

(95 % CI)

NO

YES

Fatty liver diagnosed

Fatty liver not

97

88

185

on ultrasonography

diagnosed

1.34

Fatty liver

66

80

146

(0.868-2.065)

diagnosed

Total

163

168

331

Keeping free androgen index (FAI) as dependent variable with PCOS and fatty liver disease as fixed variables indicates a definite increase in FAI due to presence of PCOS which get slightly worsened with the presence of fatty liver depicted as: Group-1: No Fatty liver + No PCOS= 2.37 (+ 1.30), n=97), Group-2: No PCOS + Fatty Liver = 3.35(+

2.23), n=66), Group-3: PCOS + No Fatty Liver = 6.08 (+ 5.05), n=88), and Group-4: PCOS + Fatty Liver = 6.83 (+ 4.70), n=80). Figure-1 shows a General Linear Model (GLM) with FAI being a dependent variable is evaluated against independent variables i.e., presence or absence of PCOS (p<0.001) and fatty liver disease (p=0.035) demonstrating that PCOS presence is more associated with increase in biochemical hyperandrogenism than fatty liver disease. (Overall Model significance<0.001). Similarly, hirsutism as measured by mFG scores was not significantly related to fatty liver disease but was associated with presence of PCOS. [Figure-2] Abdominal Volume Index (AVI) as a measure of abdominal obesity was not associated with PCOS, but was significantly associated with fatty liver disease. [Figure-3] Furthermore, insulin resistance as a dependent variable was seen to rise more with the presence of fatty liver in comparison to PCOS. [Figure-4]

Sikandar EDMJ_f2

Figure 1. General Linear Model (GLM) where FAI being a dependent variable is evaluated against independent variables i.e., presence or absence of PCOS (p<0.001) and fatty liver disease (p=0.035). (Overall Model significance<0.001).

Sikandar EDMJ_f3

Figure 2. General Linear Model (GLM) where mFG score as surrogate markers for hirsutism being a dependent variable is evaluated against independent variables i.e., presence or absence of PCOS(p <0.001) and fatty liver disease(p=0.820). (Overall Model significance<0.001).

Sikandar EDMJ_f4

Figure 3. General Linear Model (GLM) where Abdominal Volume Index (AVI) as a marker for abdominal obesity being a dependent variable is evaluated against independent variables i.e., presence or absence of PCOS (p=0.980) and fatty liver disease (p<0.001). (Overall Model significance<0.001).

Sikandar EDMJ_f5

Figure 4. General Linear Model (GLM) with insulin resistance as dependent variable and presence or absence of PCOS (p=0.157) and fatty liver disease (p<0.001) as independent variables (Overall Model significance<0.001).

Discussion

Polycystic ovarian syndrome (PCOS) and fatty co-occurrence in study subjects was slightly higher than subjects with having one of the pathology, implying a very weak association between the two entities. Though the OR for presence of PCOS in subjects with fatty liver disease was above i.e., 1.34 but it was not statistically significant. Our data identified free androgen index and hirsutism to be more associated than fat deposition within liver as factors in contributing polycystic ovarian pathology. Furthermore we identified increase in AVI i.e., as a surrogate for abdominal fat deposition and insulin resistance to be more associated with presence of fatty liver disease than with PCOS. The trend, hence identified a strong link for insulin resistance in causation of fatty liver disease and hyperandrogenism for PCOS. Contrary to our findings the data from some studies have highlighted a stronger link between obesity an insulin resistance markers with as common culprits in associating fatty liver and PCOS. [7, 8, 10, 11].

Macut et al have also identified a much higher combination diagnosis of PCOS and fatty liver disease than presence of a single pathology in Serbian population. [21] Coming to our data in terms of finding a weaker link it is important to understand that lean PCOS types are well-recognized in sub-continental population. [22] More so as Pande et al evaluated obesity and insulin resistance indices in these lean category of PCOS and observe them to be dissimilar than obese PCOS subjects, where the latter group were found to have more metabolic derangements.

[23] Similar to that Chinese have results quite similar to ours, where BMI and NAFLD were minimally associated with PCOS females. [24] Even some European studies have found a higher percentage of up to 40% lean-PCOS in their studies. [25] However, if we study the western population a contrast appear where insulin resistance and higher trunk fat mass clearly supersedes the Asian PCOS phenotype. [26] Therefore regional differences and possible epigenetic triggers can be attributed to the differential or minimal relationship between fatty liver disease and insulin resistance in subjects with PCOS.

Possible understanding of authors on this minimally existent relationship between PCOS and fatty liver disease in our data subjects is being explained below: Firstly, we described in the introduction that adipocyte size variation between Asian and Caucasian population, so possibly smaller adipocyte among Asians with capacity to accumulate more fat led to lesser abdominal volume indices and fatty liver disease in our data of female subjects. [15, 24] The argument therefore can be made that these most often obese-PCOS phenotypes females had higher insulin resistance due to increase fat mass and least contributed by hyperandrogenism. [26] However, we as author feel more interventional trials can clarify the real causation of PCOS and in specific its relation with abdominal and hepatic steatosis. Secondly, what impact the weather, environment and lifestyle could lead to aforementioned variation of PCOS patterns? We highlighted earlier the findings of

Bruce et al who described the role of environment in causing variable PCOS phenotypes. [13] A Spanish study by Concha et al evaluated the epigenetic marks by analyzing miRNAs and histone methylation in various body fluids and tissues to conclude a very strong impact of environmental triggers in association

with development of PCOS. [27] Pursuant and related directly to epigenetic mechanisms Monniaux et al was able to demonstrate in ovine fetuses that over exposure of follicular and uterine tissues may lay the foundation stone for a possible PCOS phenotype. [28] Finally, dietary patterns may be a significant contributor to PCOS as studies on gut microbiome had a different phylogenetic type along with lower density than control female subjects. [29] Thus shifting diet patterns from traditional and more natural food consumption to refined and high caloric diets could be an add on, if not causative factor in leading to PCOS development. We, therefore believe with globalization and mixing of people and in specific adopting western style diets and life styles the disease pattern may change overtime tilting more in favor of obese-phenotype. [30]

Certain limitations regarding our cross-sectional analysis needs to be taken into account: Firstly, we did this study in hospital-setting and our findings must be extrapolated to an epidemiological level to understand PCOS and its phenotype prevalence within our community. Secondly, we feel certain cultural barriers persist in our community due to lack of basic medical education and history. Provided our questionnaire format was structured and no language barrier was there still we interpret and this aspect to cause some degree of bias.

This study has important clinical implication especially in backdrop of wide regional and racial variation PCOS phenotypes. Taken the study setting and cross-sectional design the study can be further replicated to validate phenotypes of PCOS prevailing at broader level. Thus more data should follow this study project. However, this article does provide a broader guideline in terms of interpreting and segregating PCOS phenotypes within our population, which can help treating physicians to specify their treatment options in a more personalized manner. On a national scale the data also highlights the need for local guidelines for our community using this data and evidence from surrounding regions.

Conclusion

Presence of fatty liver disease in females can lead to slightly higher frequency of PCOS. Biochemical and clinical hyperandrogenism were more associated with presence of PCOS than with fatty liver disease, while abdominal obesity and insulin resistance were more often than not associated with fatty liver disease in comparison to PCOS.

Declarations

  • The data sets and SPSS outputs used and/or analyzed during the current study are available from the corresponding author on formal request.
  • Ethical approval – The study “Relationship between polycystic ovarian syndrome (PCOS) and fatty liver disease” was approved by ethical committee of the hospital. All participants were volunteer and provided “written consent” for the study.
  • Author’s contributions – SHK: (Author for all Correspondence) Study plan, Involved in study plan, involved in sampling, methodology, lab analysis, data analysis, manuscript writing. SA: History collection, data analysis, and contributed towards discussion. RS: Radiological diagnosis of PCOS, Defining PCOS as per criteria, manuscript writing. RM: Initial patient history collection and examination, defining oligo/anovulation, data analysis, manuscript writing. RA: Patient examination, history writing, anthropometric measurements, manuscript writing. TC: Study plan, methodology, manuscript writing. Final manuscript was approved by all authors.
  • Consent for publication: Signed consent was sorted from all study participants.
  • Competing interests – There are no competing interests to declare.
  • Data funding – The study had no funding source to disclose.
  • Acknowledgements – The authors acknowledge the work of Miss Huma, and Lab technician Ibrahim and Iftikhar for the support.

Abbreviations

Fatty liver disease, Polycystic ovarian syndrome (PCOS), Free Androgen Index (FAI), Homeostasis Model Assessment for Insulin Resistance (HOMAIR), Modified Ferrimen Gellwey (mFG) score, Abdominal Volume Index (AVI).

References

  1. Tsouma I, Kouskouni E, Demeridou S, Boutsikou M, Hassiakos D and Chasiakou A. Lipid lipoprotein profile alterations in Greek infertile women with polycystic ovaries: influence of adipocytokines levels. In Vivo. 2014 Sep-Oct; 28(5): 935-9.
  2. Zhang X, Wang Y and Liu P. Omic studies reveal the pathogenic lipid droplet proteins in non-alcoholic fatty liver disease. Protein Cell. 2017 Jan; 8(1): 4-13. DOI: 10.1007/s13238-016-0327-9.
  3. Donati MB. The “common soil hypothesis”: evidence from population studies? Thromb Res. 2010 Apr; 125 Suppl 2: S92-5. DOI: 10.1016/S0049-3848(10)70023-2.
  4. Spritzer PM, Barone CR and Oliveira FB. Hirsutism in Polycystic Ovary Syndrome: Pathophysiology and Management. Curr Pharm Des. 2016; 22(36): 5603-5613.
  5. Margini C and Dufour JF. The story of HCC in NAFLD: from epidemiology, across pathogenesis, to prevention and treatment. Liver Int. 2016 Mar; 36(3): 317-24. DOI: 10.1111/liv.13031.
  6. Macut D, Bjekić-Macut J, Livadas S, Stanojlović O, Hrnčić D and Rašić-Marković A, Nonalcoholic Fatty Liver Disease in Patients with Polycystic Ovary Syndrome. Curr Pharm Des. 2018; 24(38): 4593-4597. DOI: 10.2174/1381612825666190117100751.
  7. Rocha ALL, Faria LC, Guimarães TCM, Moreira GV, Cândido AL, Couto CA and Reis FM. Non-alcoholic fatty liver disease in women with polycystic ovary syndrome: systematic review and meta-analysis. J Endocrinol Invest. 2017 Dec; 40(12): 1279-1288. DOI: 10.1007/s40618-017-0708-9.
  8. Vassilatou E. Nonalcoholic fatty liver disease and polycystic ovary syndrome. World J Gastroenterol. 2014 Jul 14; 20(26): 8351-63. DOI: 10.3748/wjg.v20.i26.8351.
  9. Kauffman RP, Baker TE, Baker V, Kauffman MM and Castracane VD. Endocrine factors associated with non-alcoholic fatty liver disease in women with polycystic ovary syndrome: do androgens play a role? Gynecol Endocrinol. 2010 Jan; 26(1): 39-46. DOI: 10.3109/09513590903184084.
  10. Baranova A, Tran TP, Birerdinc A and Younossi ZM. Systematic review: association of polycystic ovary syndrome with metabolic syndrome and non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2011 Apr; 33(7): 801-14. DOI: 10.1111/j.1365-2036.2011.04579.x.
  11. Kuliczkowska Plaksej J, Laczmanski L, Milewicz A, Lenarcik-Kabza A, Trzmiel-Bira A and Zaleska-Dorobisz U. Cannabinoid receptor 1 gene polymorphisms and nonalcoholic Fatty liver disease in women with polycystic ovary syndrome and in healthy controls. Int J Endocrinol. 2014; 2014: 232975. DOI: 10.1155/2014/232975.
  12. Jones H, Sprung VS, Pugh CJ, Daousi C, Irwin A and Aziz N. Polycystic ovary syndrome with hyperandrogenism is characterized by an increased risk of hepatic steatosis compared to nonhyperandrogenic PCOS phenotypes and healthy controls, independent of obesity and insulin resistance. J Clin Endocrinol Metab. 2012 Oct; 97(10): 3709-16. DOI: 10.1210/jc.2012-1382.
  13. Bruce KD and Cagampang FR. Epigenetic priming of the metabolic syndrome. Toxicol Mech Methods. 2011 May; 21(4): 353-61. DOI: 10.3109/15376516.2011.559370.
  14. Petäjä EM, Sevastianova K, Hakkarainen A, Orho-Melander M, Lundbom N and Yki-Järvinen H. Adipocyte size is associated with NAFLD independent of obesity, fat distribution, and PNPLA3 genotype. Obesity (Silver Spring). 2013 Jun; 21(6): 1174-9. DOI: 10.1002/oby.20114.
  15. Anand SS, Tarnopolsky MA, Rashid S, Schulze KM, Desai D and Mente A. Adipocyte hypertrophy, fatty liver and metabolic risk factors in South Asians: the Molecular Study of Health and Risk in Ethnic Groups (mol-SHARE). PLoS One. 2011; 6(7): e22112. DOI: 10.1371/journal.pone.0022112.
  16. Goodman N, Bledsoe M, Cobin R, Futterweit W, Goldzieher J, Petak S, Smith K and Steinberger E: “American Association of Clinical Endocrinologists Hyperandrogenism Guidelines”. Endocrine Practice 2001; 7(2): 120–13.
  17. Kollmann M, Klaritsch P, Martins WP, Guenther F, Schneider V and Herzog SA. Maternal and neonatal outcomes in pregnant women with PCOS: comparison of different diagnostic definitions. Hum Reprod. 2015 Oct; 30(10): 2396-403. DOI: 10.1093/humrep/dev187.
  18. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group 2004 Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil Steril 81: 19–25.
  19. Al Kindi MK, Al Essry FS, Al Essry FS and Mula-Abed WA. Validity of serum testosterone, free androgen index, and calculated free testosterone in women with suspected hyperandrogenism. Oman Med J. 2012 Nov; 27(6): 471-4. DOI: 10.5001/omj.2012.112.
  20. Matthews DR, Hosker  JP,  Rudenski  AS,  Naylor  BA,  Treacher  DF and Turner  RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985 Jul; 28(7): 412-9.
  21. Macut D, Tziomalos K, Božić-Antić I, Bjekić-Macut J, Katsikis I and Papadakis E. Non-alcoholic fatty liver disease is associated with insulin resistance and lipid accumulation product in women with polycystic ovary syndrome. Hum Reprod. 2016 Jun; 31(6): 1347-53. DOI: 10.1093/humrep/dew076.
  22. Goyal M and Dawood AS. Debates Regarding Lean Patients with Polycystic Ovary Syndrome: A Narrative Review. J Hum Reprod Sci. 2017 Jul-Sep; 10(3): 154-161. DOI: 10.4103/jhrs.JHRS_77_17.
  23. Pande AR, Guleria AK, Singh SD, Shukla M and Dabadghao P. β cell function and insulin resistance in lean cases with polycystic ovary syndrome. Gynecol Endocrinol. 2017 Nov; 33(11): 877-881. DOI: 10.1080/09513590.2017.1342165.
  24. Qu Z, Zhu Y, Jiang J, Shi Y and Chen Z. The clinical characteristics and etiological study of nonalcoholic fatty liver disease in Chinese women with PCOS. Iran J Reprod Med. 2013 Sep; 11(9): 725-32.
  25. Ciotta L, Pagano I, Stracquadanio M and Formuso C. Polycystic ovarian syndrome incidence in young women with non-alcoholic fatty liver disease. Minerva Ginecol. 2011 Oct; 63(5): 429-37.
  26. Glintborg D, Petersen MH, Ravn P, Hermann AP and Andersen M. Comparison of regional fat mass measurement by whole body DXA scans and anthropometric measures to predict insulin resistance in women with polycystic ovary syndrome and controls. Acta Obstet Gynecol Scand. 2016 Nov; 95(11): 1235-1243. DOI: 10.1111/aogs.12964.
  27. Concha C F, Sir P T, Recabarren SE and Pérez B F. Epigenetics of polycystic ovary syndrome. Rev Med Chil. 2017 Jul; 145(7): 907-915. DOI: 10.4067/s0034-98872017000700907.
  28. Monniaux D, Genêt C, Maillard V, Jarrier P, Adriaensen H and Hennequet-Antier C. Prenatal programming by testosterone of follicular theca cell functions in ovary. Cell Mol Life Sci. 2019 Jul 20. DOI: 10.1007/s00018-019-03230-1.
  29. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V and Leber B. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS One. 2017 Jan 3; 12(1): e0168390. DOI: 10.1371/journal.pone.0168390. ECollection 2017.
  30. Rodgers RJ, Suturina L, Lizneva D, Davies MJ, Hummitzsch K, Irving-Rodgers HF and Robertson SA. Is polycystic ovary syndrome a 20th Century phenomenon? Med Hypotheses. 2019 Mar; 124: 31-34. DOI: 10.1016/j.mehy.2019.01.019.

Effect of Glucose Oxidase on the Cytokine Profile and Cholesterol of Monocytes in Liver Cancer

DOI: 10.31038/MIP.2020112

Abstract

Introduction: Cancer has an extremely important human and socio-economic impact. It is due to several cellular disturbances causing uncontrollable proliferation, which stimulate immune system elements including monocytes that can play a dual role either in the elimination or progression of cancer cells [1], these disturbances affect a variety of functions such as glucose metabolism [2].

Keywords

Liver cancer, Monocyte, GOx, Cytokines. Cholesterol

Objectives

This work hopes to investigate the effect of glucose oxidase at the level of the monocyte on the tumor growth thus determining its impact on the glycolysis activity and on the mitochondrial metabolism.

Aim: The aim of this study is to show the role of GOx in the polarization of monocytes in contact with tumor cells.

Materials and methods

Monocytes isolated from the blood of the cancer patient were co- cultured with the tumor epithelial cells isolated from a biopsy of the liver cancer, in a culture medium supplemented or not with GOx.

Results

GOx induced an increase in the INF-γ and decrease of IL-10 as well as in NO and Arginase in the presence of Glucose oxidase compared to thus without GOx, the Cholesterol in microenvironment was decreased in presence of GOx.

Conclusion

In conclusion, our results showed an pro-inflammatory effect was reported in monocytes in contact with liver tumor epithelial cells.

References

  1. Elliott LA, Doherty GA, Sheahan K, Ryan EJ (2017) Human Tumor-Infiltrating Myeloid Cells: Phenotypic and Functional Diversity. Front Immunol 8. [Crossref]
  2. Annibaldi A, Widmann C (2010) Glucose metabolism in cancer cells. Curr Opin Clin Nutr Metab Care 13: 466-470. [Crossref]

Biomarkers of Oxidative with Colorectal Cancer

DOI: 10.31038/MIP.2020111

Abstract

The number of   colorectal cancer increases regularly every year in Algeria and precisely in Tlemcen. It is a disease involving several genetic, hormonal, professional, environmental, but also behavioral factors, namely nutrition, the objective is to evaluate the nutritional and lipid profile on the one hand and some oxidative parameters of And to determine the relationship that may exist between nutritional factors and oxidative stress in patients with this type of cancer.

Keywords

Oxidative Stress, Colorectal Cancer

Materials and Methods

Thirty-three patients with newly diagnosed colorectal cancer were recruited from the Department of Gastroenterology, C.H.U. (CL, HDL-LC, LDL-CL, TG) and the oxidative status (ORAC, Catalase, Vitamin C, MDA) of Tlemcen and thirty-five healthy controls.

Results

A very significant difference was observed for patients with liver cancer compared to ORAC controls (1, 143 ± 0.121) (0, 4197 ± 0.0456) p <0.000; MDA (0, 2400 ± 0.0492) (0, 083 ± 0.0275) P <0.008; Catalase (1.385 ± 0.162) (0, 588 ± 0.219) p <0.008; LDL-CHOL (0.4300 ± 0.0239) (1. 0692 ± 0.0627 p <0.000, on the other hand no difference was observed for Vitamin C (0.221 ± 0.0465) (0, 1947 ± 0.0889) p <0.615 HDL-CHOL (0.4300 ± 0.0322) (0.466 ± 0.031) p <0.355.

The balance of the oxidizing / antioxidant status is a primary factor in oncology. The free radicals can lead to the appearance of mutations and, conversely.

References

  1. Golbidi S, Laher I (2010) Antioxidant therapy in human endocrine disorders. Med Sci Monit 16: 9–24.
  2. Jaeschke H (2011) Reactive oxygen and mechanisms of inflammatory liver injury: Present concepts. J Gastroenterol Hepatol 1: 173–179. [Crossref]

Concurrent Collapses of Demersal Fish and Sea Trout (Salmo trutta) on Scotland’s West Coast Following the Removal of the “Three-Mile Fishing Limit”

DOI: 10.31038/AFS.2019112

Abstract

The collapse of the sea trout (Salmo trutta) fishery in Loch Maree on Scotland’s north west coast occurred during the late 1980s.  Stocks of west coast demersal marine fish, especially around the Clyde Estuary have also declined over a similar time span. The decline of these marine fish stocks can be attributed to the removal of the “three-mile fishing limit” in 1984 by UK Government legislation. Sea trout inhabit the same inshore waters as targeted demersal fish and can be caught as unrecorded by-catch. Comparisons of the decline of demersal species and the sea trout the west coast show a high degree of correlation.

Keywords

fisheries, demersal fish, sea trout, stock collapse, three-mile fishing limit

Introduction

Catches of sea trout (Salmo trutta) using rod and line from rivers along Scotland’s north west coast have been in decline since the UK Government’s Scottish Office first began to collect records in 1952. Many different factors have been implicated in the decline including forestry, land use changes, acid rain, global and local climate change and overexploitation. However, attribution to any one factor is mostly anecdotal Picken [1].

Butler & Walker [2] detailed the collapse of the world-renowned Loch Maree sea trout fishery during the late 1980s. They found that prior to 1987, annual catches of sea trout ranged between 546 and 1575 whilst from 1988 to 2001, catches fluctuated between 35 and 342, the lowest number being recorded in 2001. Butler & Walker blame the collapse on the establishment of a salmon farm in the adjacent sea loch, Loch Ewe in 1987, but do not provide any evidence to support their claim. They state that there is no other explanation for the collapse.

In 1984, the Inshore Fishing (Scotland) Act, implemented by the Scottish Office, repealed a ban on fishing within three nautical miles of the low water mark. This “three-mile limit” had been introduced in 1889 in the interest of protecting fish stocks against overexploitation Thurstan & Roberts [3]. The opening of inshore waters in 1984 meant that fishing vessels were able to trawl for fish along the north-west coast and within the confines of many sea lochs. This paper considers whether the removal of the “three-mile limit” may have been a significant contributory factor in the collapse of sea trout stocks in the Loch Maree fishery and elsewhere along Scotland’s north-west coast.

Materials & Methods

Examination of pre-existing data- Sea trout and the Ewe catchment including Loch Maree

Loch Maree is a 20km long freshwater loch located in north-west Scotland. It is connected to Loch Ewe, a 16km long sea loch by the 4km long River Ewe. Loch Maree had a reputation of world renown for its sea trout angling. Records of fish catches, both numbers and weights, are collected by the holders of fishing rights and submitted to Scottish government scientists for analysis. The catch records for sea trout from 1980 onwards for the whole Ewe System are shown in Figure 1.

AFS_2020-Martin Jaffa_F1

Figure 1. Annual weight of sea trout caught from the Loch Ewe catchment including Loch Maree. The bars represent the annual catch in kg. The line represents the moving five-year average.

The bars represent the annual catch in kg. The line represents the moving five-year average.

The decline in the number of sea trout is apparent from both the annual weight of sea trout caught and the moving five-year average.

Examination of Pre-Existing Data – Demersal Fish Catches and the “Three-Mile Limit”

Thurstan & Roberts [3]. examined the decline of catches of marine fish such as cod (Gadus morhua), whiting (Merlangius merlangus) and saithe (Pollachius virens) from the Inner Clyde fishing grounds around Scotland’s west coast. Figure 2.

AFS_2020-Martin Jaffa_F2

Figure 2. Comparison of cod catches from the Inner Clyde fishing grounds. The bars represent the annual catch in tonnes. The line represents the moving five-year average. The decline of cod landings from the Inner Clyde is apparent from the annual record and the five-year average.

Collection of Data

The Scottish Government publish catch data for sea trout and landings of demersal fish annually on the statistics section of their website. Catch data for sea trout was collated from all fishery districts from Ullapool to the Clyde and landings for cod, whiting and saithe for all west coast fishing ports except Kinlochbervie, which is promoted as a drop off port for distant water fishing vessels. The various data sets were analysed to ascertain the likelihood that the decline of sea trout stocks during the late 1980s including the collapse of the Loch Maree fishery might be one consequence of changes to marine fishing legislation, specifically the removal of the “three-mile limit”.

Statistical Analysis

Regression Analyses

One complication in analysing such data is the different ways in which information for the different species are expressed in different locations. Some data was recorded as numbers of fish, whilst other data was based around weight of fish. Given the varying numbers for different species/locations, the approach taken in the analysis was to examine the relative change in fish numbers/weight over time using simple linear regression.

AFS_2020-Martin Jaffa_eq1

Where β0 is the intercept, β1 represents the beta (β) coefficient (parameter estimate), X1 is the values of the variable in the regression model and  is the error term (residuals).

Comparison of regression slopes

For comparison of the slopes, a methodology using Z-statistic has been applied.

AFS_2020-Martin Jaffa_eq2

Where b1 is the slope of line 1, b2 is the slope of line 2, SEb1 is the standard error of slope 1 and  SEb2 is the standard error of the slope of line 2. The resulting Z-value was compared to the normal distribution to identify the statistical significance.

Graphical Presentation

All the sets of data were converted to a centred moving average and subsequently standardised to the same scale. The use of standardisation focuses on the pattern of catches rather than the actual volume.

The transformation of each variable was achieved using the formula

AFS_2020-Martin Jaffa_eq3

Where x is the volume of each fish species in a single year, AFS_2020-Martin Jaffa_eq4 is the mean average volume of each fish species over all years and σ is the standard deviation of volume of each fish species over all years. This will result in a variable with a mean of zero and a standard deviation of 1.

Results

Demersal Fish and the Lifting of the “Three-Mile Limit”

In 1984, the government removed the “three-mile limit” allowing fishing boats to trawl in inshore waters around Scotland Thurstan & Roberts [3] demonstrated that this resulted in a reduction of catches of marine demersal fish in the Inner Clyde. This is the most intensively studied area of inshore waters in Scotland, but the impacts are likely to be similar elsewhere around the Scottish coast.

West Coast

The Inner Clyde fishing grounds are located at the most southerly point of the north-west Highland region. Other fishing grounds are located along the length of the west coast with fish landed at several small fishing ports including Oban, Mallaig and Ullapool. The landings for cod, whiting and saithe are recorded by the Scottish Government and are subsequently published on the Scottish Government website.

The decline in demersal fish landed at west coast ports follows a similar pattern to the decline identified by Thurstan & Roberts [3] from within the Inner Clyde. Opening the “three-mile limit” to inshore fishing appear to have negatively impacted the tonnage of fish caught along the whole of the Scottish west coast.

Figure 3. combines the data for the four species using a five-point centred moving average on a standardised scale of the weight of fish landed. This clearly shows the similarities between the decline of marine species and sea trout.

AFS_2020-Martin Jaffa_F3

Figure 3. Five-point centred moving weight average on a standardised scale for west coast landings of cod (dashed line), whiting (dashed and dotted line), saithe (dotted line) and sea trout (solid line).

The data was analysed to determine the correlation between species. Sea trout is strongly correlated with cod (r = 0.83, p < 0.0001), whiting (r = 0.88, p < 0.0001) and saithe (r = 0.83, p < 0.0001).

Comparison of the rate of decline between species using regression slopes found that sea trout declined at a similar rate to cod (Z = -1.30, p = 0.1933) and whiting (Z = -0.34, p = 0.7322) but faster than saithe (Z = -6.90, p < 0.0001). This difference may be due to variability of different stocks in the west coast fishing grounds.

The west coast data covers the whole of the west coast excluding the furthest north. Landings of demersal can be identified from specific ports. Mallaig is an important fishing port half way up the west coast.  Figure 4 shows the landings at Mallaig compared to sea trout catches as a five-point centred moving average on a standardised scale for the weight of fish landed.

AFS_2020-Martin Jaffa_F4

Figure 4. Five-point centred moving weight average on a standardised scale for Mallaig catches of cod (dashed line), whiting (dashed and dotted line), saithe (dotted line) and sea trout (solid line).

The correlation between sea trout and the demersal species is less strong than for the west coast. This is not unexpected as marine fish stocks will vary along the coast due to the nature of the coastline. Sea trout is most correlated with cod (r = 0.68, p < 0.0001) but less correlated with whiting (r = 0.56, p = 0.0005) and saithe (r = 0.59, p = 0.0002). The comparison of the rate of decline using regression lines suggests that sea trout declined at a faster rate than demersal fish around Mallaig. However, the regression line for sea trout is likely to have bene influenced by the outlying points of high numbers in 1980 and 1981. Had the data started from 1982, the rate of decline would have been less steep. The data suggests that sea trout have declined faster than cod (Z = -4.01, p < 0.0001), whiting (Z = -2.17, p = 0.0300) and saithe (Z = -7.15, p < 0.0001).

Nephrops Fishery

Thurstan & Roberts [3] chart changes to in-shore fishing catches and noted that following the collapse of demersal fish stocks in in-shore waters, fishing boats have targeted Nephrops prawns. This crustacean has almost replaced demersal fish has the main catch in the Inner Clyde. Prawns are also targeted from ports around the whole Scottish coast. In 2016, Nephrops represented the second largest value of seafood landed at Scottish ports at £79 million with 21,000 tonnes which is greater than both cod and haddock together at £65 million. McIntyre et al. [4] highlight that the North East Atlantic Nephrops trawling fishery has been ranked as having the fifth highest discard rate in the world.  In the Inner Clyde, this is estimated to be between 66% and 80%. These are species that are not specifically targeted by the fishing boat and are subsequently discarded. It is not recorded whether these discards include sea trout.

Discussion

Collapse of Stocks of Demersal Fish

Thurstan & Roberts [3] report that prior to the removal of the “three-mile limit” in 1984, catches had increased during the 1960s and 1970s because key fishing grounds around the Clyde had been opened to more efficient pair-trawling. However, these higher catches were not maintained which led to fishermen demanding help from the Government. As a result, the UK Government introduced the Inshore Fishing (Scotland) Act in 1984 which repealed a ban on trawling in inshore waters. The new legislation removed the restriction on fishing within the “three-mile limit” expanding fishing opportunities as fishermen struggled to catch enough fish to sustain their livelihoods. The Firth of Clyde, as well as other west coast inshore fishing grounds, were opened to trawling allowing exploitation of species such as cod, whiting and saithe. Yet, the reopening of the “three-mile limit” did not bring an increase in landings as between 1984 and 2009, landings decreased by 99% Hislop [5]. In addition to cod, saithe and whiting, stocks of haddock, hake, herring, flounder and plaice all showed a similar downward trajectory (Thurstan & Roberts; McIntyre et al.) [3,4].

Sea trout stocks

There are 109 fishery districts across all of Scotland. Each is very different in nature and the number of fish caught annually. Most sea trout stocks from west coast fishery districts have shown a decline over many years. It is unclear why these stocks have declined, and several different factors have been proposed, but none are likely to be the sole reason. Instead, it is likely that there are a combination of factors bringing about these declines or else there is no obvious explaination. Sea trout catches in the Clayburn fishery district in the Outer Hebrides collapsed in 1976 and have never recovered. There has been no scientific investigation into this collapse, and the cause remains unknown

Relationship Between Sea Trout and Demersal Stock Collapses

The decline of sea trout catches from Scotland’s west coast strongly correlates with declines of landings of marine demersal fish at west coast fish ports with similar declines of saithe, cod and whiting. Whilst correlation does not equate to causation, the loss of marine fish can be attributed to the removal of the “three-mile limit” in 1984 which allowed fishing boats to trawl in inshore waters including sea lochs. Although sea trout stocks were already in decline, the removal of the “three-mile fishing limit” provides a plausible explanation as to cause of the collapse of the Loch Maree sea trout fishery Walker AF [6].

Significance Statement

Sea trout catches by rod and line have been in decline along the Scottish west coast since records began in 1952. No specific reason has been identified for the decline.  During the 1980s, the world-renowned sea trout fishery in Loch Maree collapsed. The cause of this collapse also remains unexplained.  This research has identified similarities between the collapse of demersal fish stocks and sea trout following the removal of the three-mile limit in 1984.

References

  1. Picken MJ (1990) The Sea Trout in Scotland. Symposium Proceedings. Scottish Marine Biological Association: 53–59.
  2. Butler JRA, Walker AF (2006) Characteristics of the Sea Trout (Salmo trutta) Stock  Collapse in the River Ewe (Wester Ross Scotland), in 1988–2001. In Sea Trout: Biology, Conservation and Management. Edited by Harris, G. & Milner, N. Blackwell Publishing Page: 45–59.
  3. Thurstan RH, Roberts CM (2010) Ecological meltdown in the Firth of Clyde, Scotland, Two centuries of change in a coastal marine ecosystem. PLos ONE 5.
  4. McIntyre F, Fernandes PG, Turrell WR (2012) Clyde Ecosystem Review. Scottish Marine and Freshwater Science: 3
  5. Hislop JRG (1986) The demersal fishery in the Clyde sea area. Proc. Roy. Soc.  90: 423–437.
  6. Walker AF (2006) The rapid establishment of a resident brown trout population from sea trout progeny in a fishless stream. In: Sea Trout Biology, Conservation & Management (Harris, G.S. & Milner N.J. eds). Proceedings of the First International Sea Trout Symposium. July 2004. Cardiff, Wales. Blackwell Publishing Oxford: 389–400.

Augmentation of Extraction Sockets – A Requisite for Future Rehabilitation

DOI: 10.31038/JDMR.2020313

Abstract

Purpose: In this prospective study, bone formation in human extraction sockets is augmented with bovine- cancellous bone (Bio-Oss) and compared to bone formation in unaugmented extraction sockets.

Methods: Twenty patients with bilateral extraction sockets were included in this study. After all 40 extractions, one socket was augmented using cancellous bovine bone and another socket left to heal naturally. Bone density was measured by means of histogram, after 1 week, 1 month & 3 months following extractions.

Results: Extraction sites with graft by the means of histogram showed significant difference in the bone density. They were significantly augmented as compared to the sockets without graft.

Conclusion: The results of the present study suggests that the use of bone xenograft as graft material for alveolar ridge preservation after extraction. This aids in future rehabilitation with implant placement or fixed prosthesis.

Keywords

alveolar bone augmentation, extraction socket, socket preservation, bone density

Introduction

Tooth extraction is one of widely the performed procedures in dentistry and it has been historically well documented that this procedure may induce significant dimensional changes of alveolar ridge. The dilemma that clinician face today is how to manage tooth extraction to provide a good ridge for the future placement of a dental implant or to maximize ridge dimension for the fabrication of fixed or removal prosthesis. If performed inadequately the resulting deformity can be considerable obstacle to the aesthetic, phonetic, and functional results; that both our patients and we clinician expect at this current time [1].

Bone resorption occurs in the first three months following extraction. The condition appears to be progressive and irreversible, resulting in a host of prosthodontics, aesthetic, and functional problems. Post extraction bone loss is accelerated in the first 6 month, followed by a gradual modelling and remodelling of the remaining bone, with as much as 40% of the alveolar height and 60% of alveolar width lost in the first 6 months [2].

Jaw deformities from tooth removal can be prevented and repaired by a procedure called socket preservation. The procedure begins with atraumatic tooth extraction. Every attempt is made to preserve the surrounding bone and soft tissue, with an example on being careful not to fracture delicate buccal plate. After the extraction, a bone graft material is placed into the socket and covered with a resorbable or non-resorbable membrane and sutured. Most importantly, socket preservation helps to maintain the alveolar architecture and significantly reduces the loss of ridge width and height following tooth removal[3].

Various materials are used in modern dental and maxillofacial surgery for bone tissue substitution and reconstruction. All osteoplastic materials can be divided into four groups by origin: autogenic, allogeneic, xenogeneic, and synthetic.

Increasing the height and width of bone helps ensure the success and longevity of dental implants. Surgeons can utilize a variety of surgical procedure in ridge augmentation. The surgical procedure will depend on amount of available bone, the amount of augmentation necessary and patient related factors.

In this prospective clinical study, after multiple extractions, one socket is filled with xenograft graft and other socket left to heal naturally and then evaluated by means of bone density test, post-operatively.

Materials and Methods

After procuring the ethical approval from the ethical committee, 20 patients who reported to the Department of Oral and Maxillofacial Surgery, who required extractions of bilateral teeth either from maxilla or mandible. Out of these 40 extractions; all left sided sockets were preserved with bovine-cancellous bone (Bio-Oss) and were grouped as Group A; and all right sided extractions were left to heal naturally and were grouped as Group B.

To keep the study unbiased in regards of surgical expertise, only one surgeon performed all the cases in the present study.

The study protocol was explained to the patient in detail and their consent was obtained.

Inclusion Criteria: –

  • Systemically healthy patients.
  • Patients requiring bilateral extractions.
  • Tooth which were un-restorable by endodontic treatment and free from acute infections.
  • Patients who required intra-alveolar extractions.
  • Patients within 18–55 years of age group.
  • Patients who are willing to sign the informed consent.

Exclusion Criteria:-

  • Pregnancy or lactation
  • Systemically ill patients.
  • Chronic generalised/localized aggressive periodontitis patients or patients having severe bone loss more than 80%.
  • Bone disease or the use of medications that interfered with bone metabolism

Materials

  1. Deproteinized cancellous bovine bone supplied by Geistlich biomaterial, Inc. It has a granules size of 0.25mm-1mm. It is marketed in a sterile airtight pack, sterilized by gamma irradiation (Geistlich Bio-Oss).
  2. Vicryl suture material.

Methodology

The surgical procedure was carried out in an operating room under strict aseptic conditions. The surgeries were performed under local anaesthesia (lignocaine 2% with epinephrine 1:80,000). Furthermore, patients were given an antibiotic prophylaxis a day before surgery. The surgical procedure was performed with standard instruments set used for minor oral surgical procedures.

Multiple tooth extraction was done carefully & atraumatically, while preserving the alveolar bone plates around the teeth. The socket was then gently flushed with normal saline.

In all left sockets, bone graft was placed. Sutures were used to approximate the grafted sockets and in all right sided ungrafted sockets were compressed digitally and allowed to heal naturally.

All patients were given postoperative instructions following extractions. Antibiotics and Analgesics were prescribed postoperatively.

Assessment of patients was done at the end of 1st week, 1st and 3rd months postoperatively, based on the following parameters:

  • Pain Visual Analogue Scale (VAS) (Figure 1)
  • Infection – present/absent
  • Inflammation – present/absent
  • Swelling – present/absent
  • Graft – accepted/rejected
  • Bone density by means of histogram. Changes in alveolar bone level were measured by means of histogram (adobe photoshop) pixels ranging between 15000–16000.

JDMR 2020-301_Savina Gupta_f1

Figure 1. Visual Analogue Scale (VAS).

Results

20 patients (10 males and 10 females) with mean age of 25 participated in this prospective study. Regarding the parameters of pain, swelling, infection & inflammation in both the groups there was no significant difference. Table I represents those cases who reported with such complications. Thus, we did not procure a graphical representation of it.

Table 1. Prospective observations of parameters in volunteer participants Count (%).

Graft

 Parameter

 Indication 

Time of Observation

Preop

Week 1

Month 1

Month 3

Group A
(n=20)

Swelling

Absent

17(85%)

20(100%)

20(100%)

20(100%)

Present

3(15%)

0(0%)

0(0%)

0(0%)

Pain VAS Scale

Absent

11(55%)

20(100%)

20(100%)

20(100%)

Present

9(45%)

0(0%)

0(0%)

0(0%)

Infection

Absent

17(85%)

20(100%)

20(100%)

20(100%)

Present

3(15%)

0(0%)

0(0%)

0(0%)

Inflammation

Absent

17(85%)

20(100%)

20(100%)

20(100%)

Present

3(15%)

0(0%)

0(0%)

0(0%)

Graft

With Graft All were accepted

Group B
(n=20)

Swelling

Absent

15(75%)

20(100%)

20(100%)

20(100%)

Present

5(25%)

0(0%)

0(0%)

0(0%)

Pain VAS Scale

Absent

15(75%)

20(100%)

20(100%)

20(100%)

Present

5(25%)

0(0%)

0(0%)

0(0%)

Infection

Absent

15(75%)

20(100%)

20(100%)

20(100%)

Present

5(25%)

0(0%)

0(0%)

0(0%)

Inflammation

Absent

15(75%)

20(100%)

20(100%)

20(100%)

Present

5(25%)

0(0%)

0(0%)

0(0%)

For the grafts assessment in group A, clinical observation shows that all 20 patients indicated quicker healing with graft acceptance without any Necrosis (Figure 2).

JDMR 2020-301_Savina Gupta_f2

Figure 2. Placement of Bio Oss® Bone Graft & Post-operative healing of the socket.

On every follow up, bone density in both the groups were calculated with the help of histogram (Figure 3).

JDMR 2020-301_Savina Gupta_f3

Figure 3. Mean Bone Density Measurement by Abode Photoshop® Histogram.
Range: 15000 To 16000 Pixels.

The histogram displayed on overlay plot of different stages with increase or decrease of bone density at different times, for both the groups (Figure IV). The mean range of bone densities in group A on completion of 1st week it was 80.41±16.13 after 1 month it was 109.52±22.25 & after 3 months it was 124.84±23.17. In group B the mean range on 1st week it was 71.67±15.17 after 1 month 81.24±16.26 & after 3 months it was 89.21±15.34 (Table 2). The bone density presented in table 2 shows higher density in group A at the end of 3 months followed by 1month.

JDMR 2020-301_Savina Gupta_f4

Figure 4. Overlay Plot showing increase or decrease of bone density at different interval of times for both the groups.

Table 2. Prospective Bone Density (LS Mean ± SD).

Effect

Pre Op

Week 1

Month 1

Month 3

Group A

93.26±16.80 BC

80.41±16.13 CD

109.52±22.25 AB

124.84±23.17A

Group B

93.26±16.79 BC

71.67±15.17 D

81.24±16.26 CD

89.21±15.34 C

Superscript indicated connecting letters by Tukey HSD. Letters not similar are significantly different, (P<.001).

The ANOVA revealed significant interaction of grafting and time (P<.001) (Table 3 & Figure 5).

Table 3. Pearson Correlation of Bone Density between jaws within participants.

Group A

Group B

Bone Density Correlation

Pre Op

Pre Op

1.000**

Week 1

Week 1

0.932**

Month 1

Month 1

0.627**

Month 3

Month 3

0.740**

**Significant at P (<.001).

JDMR 2020-301_Savina Gupta_f5

Figure 5. Bone density correlation.

Discussion

In order to preserve the gift of nature, man is presently trying to be positively constructive and conservative, and thus from forest to fuel, all form of energy is being judiciously utilized [4]. The survival rate of human life has itself increased and hence maxillofacial structures, including natural teeth, are being preserved in an attempt toward this goal [5]. It is crucial to preserve the dimensions of the alveolar ridge after tooth extraction to achieve a predictable aesthetic and functional prosthetic restoration.

Alveolar preservation or Reconstruction is necessary for support, esthetics and function of any prosthodontic rehabilitation. M. M. Devan suggested that preservation of the alveolar ridge was an easier task than to reconstruct it, once it was lost [6].

The idea of directing host bone into a particular defect is not a new one [7]. The principle of sealing off an anatomical site for improved healing of a certain tissue type and directing regeneration by some type of mechanical barrier or by various resorbable bone grafts has been used in experimental osseous facial reconstruction since the mid-1950s [7].

Alveolar ridge resorption following tooth removal is physiologically undesirable and possibly unavoidable phenomenon. Significant knowledge exists of the healing process of extraction wounds, including contour changes caused by bone resorption and the cascade of histologic events in both animals and humans. It is common to see approximately 50% of the alveolar width and height resorbed within the first three months [8–10]. This can lead to ridge deformation and many other complications.

The use of bovine cancellous xenograft for bone regeneration has enhanced the surgeon’s ability to reconstruct deficient alveolus. Graft used in our study, is deproteinized cancellous (spongiosa) bovine bone supplied by Geistlich biomaterial, Inc. The graft particles are incorporated over time within living bone which provides long-term volume preservation [11–14]. The bio functionality of this graft is characterized by its topographic structure, hydrophilic properties and the biologic interaction that supports reliable bone formation [15].

In our study, the dimensions of the grafted alveolar ridge were largely preserved both horizontally and vertically after 3 months of extraction. Moreover, the bone density in the extraction socket was significantly greater 1month after extraction than in the naturally healed sockets. The slow resorption and long-term stability of xenograft appears to be an advantage for preserving alveolar ridge structures. Similar finding were demonstrated by Nevins et al showed that there is a good amount of ridge preservation after extraction of teeth in anterior region [16]. While Artzi et al observed minimal higher rates of bone in human extraction sockets filled with bovine bone mineral investigated after a healing period of 3 months than after a 6-week healing period [17].

These data demonstrates that bone resorption cannot be completely prevented even with effective socket preservation, but the dimensions of the alveolar ridge and keratinized soft tissue can be preserved to a major extent by socket preservation with Bio-Oss so that an optimal future implant site can be created predictably.

Conclusion

Alveolar ridge resorption has long been considered an unavoidable consequence of tooth extraction. With today’s increasing appearance consciousness, the days of just extracting a tooth and replacing it later is unacceptable to many patients. It is vital to preserve and maintain the edentulous ridge and gingival architecture.

In our study we discovered that bone regeneration using bovine cancellous seems promising. Clinical as well as stastical analysis of our study demonstrate an advantage of grafting after extraction for future rehabilitation with implant placement or fixed prosthesis, over the ungrafted socket. More long term studies are needed to confirm the success rate of implants placed in regenerated bone.

Compliance with ethical standards

  • Funding: no funding received
  • Conflict of interest:
  • Author 1 declares that she has no conflict of interest
  • Author 2 declares that she has no conflict of interest
  • Author 3 declares that he has no conflict of interest
  • Author 4 declares that she has no conflict of interest
  • Author 5 declares that he has no conflict of interest
  • Ethical approval: All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards
  • Informed consent: “Informed consent was obtained from all individual participants included in this study”.

References

  1. Robert Horowitz, Danny Holtzclaw, Paul S Rosen (2012) A review on alveolar ridge preservation following tooth extraction. J Evid Dent base pract S1: 149–160. [Crossref]
  2. Barry K Bartee (2001) Extraction site reconstruction for alveolar ridge preservation.Part1: Rationale and materials selection. Journal of oral Implantology 17: 187–193. [Crossref]
  3. Cena Dimova (2014) Socket preservation procedure after tooth extraction. Key engineering materials Trans Tech Publication 587: 325–330.
  4. Rohit Madan, Vivek Gupta, Vivek K Bains, Sudhir S Patil, G P Singh, et al. (2011) Socket preservation vis-à-vis natural healing: Literature review. Asian Journal of Oral Health & Allied Sciences 1:195–201.
  5. Chandra R, Bains R, Loomba K, Pal US, Ram H, et al. (2010) Endosseous dental implant vis-à-vis conservative management: is it dilemma? Natl j maxillofac Surg 1: 26–29.
  6. M. Devan (2005) Basic principles in impression making. J Prosthet Dent 95: 503–508. [Crossref]
  7. Tasso Irinakis (2006) Rationale for socket preservation after extraction of a single -rooted tooth when planning for future Implant placement. J Can Dent Assoc 72: 917.
  8. Dietmar Weng, Vera Stock, Henning Schliephake (2011) Are socket and ridge preservation techniques at the day of tooth extraction efficient in maintaining the tissues of the alveolar ridge? Systemic review. Eur J Implantol 4: 59–66.
  9.  Jung RE, Philipp A, Annen BM, Signorelli L, Thoma DS, et al. (2013) Radiographic evaluation of different techniques for ridge preservation after tooth extraction: a randomized controlled clinical trial. J Clinical Periodontol 40: 90–98. [Crossref]
  10.  Lars Schropp, Ann Wenzel, Lambros Kostopoulos, Thorkild Karring (2003) Bone healing and soft tissue contour changes following single-tooth extraction: A clinical and radiographic 12-month prospective study. Int J Periodontics Restorative Dent 23: 313–323. [Crossref]
  11. Carlo Maiorana, Mario Beretta, Giovanni Battista Grossi, Franco Santoro, Alna Scott Herford, et al. (2011) Histomorphometric evaluation of anorganic bovine bone coverage to reduce autogenous grafts resorption: Preliminary result. Int J Periodontics Restorative Dent 5: 71–78. [Crossref]
  12. Traini Traini, Pascal Valentini, Giovanna Lezzi, Adirano Piattelli (2007) A histologic and histomorphometric evaluation of anorganic bovine bone retrieved 9 years after a sinus augmentation procedure. J Periodontol 78: 955–961. [Crossref]
  13. Arne Mordenfeld, Tomas Albrektsson Mats Hallman (2014) A 10 year Clinical and radiographic study of implants placed after maxillary sinus floor augmentation with an 80:20 mixture of deprotinized bovine bone and autogenous bone. Clinical Implant Dentistry and Related Research 16: 435–436. [Crossref]
  14. Galindo-Moreno P, Pedro Hernandez-cortes, Francisco Mesa, Nelson Carranza, Gintaras Juodzbalys, et al. (2012) Slow resorption of anorganic bovine bone by osteoclasts in maxillary sinus augmentation. Clinical Implant Dentistry and Related Research 15: 858–866. [Crossref]
  15. Mueedul Islam, Mohammed Imran, Harikeerthy Panthala, Azhar Khan, Shoaib N Parkar, et al. (2017) Processed Bovine Dentine and Bone Xenograft (BIO OSS) For Bone Regeneration and Repair-A Comparative Animal Study. Saudi J. Oral. Dent. Res 2: 303.
  16. Myron Nevins, Marcelo Camelo, Sergio De Paoli, Bernard Friedland, Robert K Schenk, et al. (2006) A study of the fate of the buccal wall of extraction sockets of teeth with prominent roots. Int J Periodontics & Restorative Dentistry 26: 19–29. [Crossref]
  17. Zvi Artzi and Carlos E. Nemcovsky (1998) The application of deproteinized bovine bone mineral for ridge preservation prior to implantation. Clinical and histological observations in a case report. J Periodontol 69: 1062–1067. [Crossref]

Measurement and Clinical Significance of Novel Inflammatory Biomarkers in Patients with Psoriasis and Cardiovascular Risk Factors for the Primary Care Practitioner

DOI: 10.31038/JCRM.2020312

Summary

Large part of the current research in the field of psoriasis is directed towards identifying and characterising new biomarkers, for both diagnostic and prognostic purposes at clinical settings. This will help in developing prompt and effective therapeutic approaches, particularly to manage the cardiovascular risk factors often associated with psoriasis, to reduce cardiovascular morbidity.

This review highlights the biomarkers that have been identified to date, that are accessible and very useful to primary care physicians (PCPs), because, in most cases, these are the first indicators of psoriasis in the patients. We have focussed on the inflammatory and oxidative stress pathways to identify inflammatory biomarkers that link cardiovascular disease (CVD) risk with psoriasis, in both sexes. In addition, we have separately collated the information specific to women, unlike previous reviews.

Our review also emphasises the importance of staying abreast of the advances in the field, to understand the comorbidities of psoriasis. With new therapeutic approaches that offer novel treatment options, chances of detecting the comorbidities, even in their subclinical phase, improve. This can lead to accelerated clearance of skin lesions, and improved quality of life for the patients.

Key words

Psoriasis comorbidities, cardiovascular risk factors, inflammatory biomarkers, new biological treatments, early clinical diagnosis of psoriasis, primary health care

Introduction

Psoriasis vulgaris is a chronic, immune-mediated, inflammatory, polygenic skin disorder. Its prevalence is approximately 2% in the general population. It has a universal occurrence with a very similar proportion in men and women [1]. It can appear at any age, but two peaks in age of onset have been reported: the first between 20 and 30 years, and the second between 50 and 60 years [2].

Plaque-type psoriasis is the most frequent presentation representing 90% of cases and clinically manifests as well-demarcated erythematous plaques covered by silvery-white scales. They are located mainly on the extensor surfaces of the extremities, scalp, sacral area, and umbilicus. Psoriasis has a notable effect on quality of life; patients often experience depression, anxiety, and stigma as well as suicidal behaviour [3].

As Surcel et al. and Benson et al. [4,5] the knowledge of the pathogenesis of psoriasis has evolved over the years. Currently, the roles played by interleukin (IL) -23, IL-17, IL-22, T helper (Th) -17 cells, Th-22 cells, regulatory T cells, transforming growth factor (TGF) -β1 and IL-10 in the pathogenesis of the disease have been highlighted [4, 5].

It has been reported by Fernández-Armenteros et al. [6] that psoriasis is an independent risk factor for the development of coronary artery disease and acute myocardial infarction. Thus, patients with moderate and/or severe psoriasis have a higher risk of developing ischemic heart disease or cerebrovascular disease than the general population.

Kimball et al. [7] conducted a study with 1,591 patients with moderate or severe psoriasis, and, based on the Framingham table, they estimated the 10-year risk of coronary heart disease and stroke in this population. They verified that for patients with a Psoriasis Area and Severity Index (PASI) score (>20), the risk was estimated at 12.2% and 8.7%, respectively.

Hu et al. [8] analysed the risk of stroke in patients with psoriasis. They found that psoriasis, in mild and severe forms, is an independent factor directly related to the development of stroke. They found no relationship between psoriasis and suffering or depression.

In a thorough study, different comorbidities are listed that may accompany psoriasis, noting that several treatments target interleukin (IL) 17. However, it is still unclear whether they should be used in patients who are at a higher risk of developing CVD. There are some studies that describe its efficacy in psoriasis patients who are affected by myocardial infarction, hypertension, and chronic renal failure [9].

Anti-IL17A/F treatment has been tested in a randomized, double-blind, placebo-controlled Phase 1 clinical trial. Changes were observed in liver enzyme levels, and clinical readouts, such as vital signs, electrocardiograms, or body weight. In this trial, no deaths were reported [10].

“Author last name” briefly describes biomarkers for systemic inflammation, noting that some are prognostic biomarkers, and that some are diagnostic biomarkers. A complete review of biomarkers is described here in our manuscript.

Thus, psoriasis can be considered as the amplifier of the inflammatory march if the metabolic disorder appears before the diagnosis of psoriasis [11].

Patients with psoriasis have been found to have increased arterial stiffness. Increased risk of hypercholesterolemia, atherosclerosis, metabolic syndrome, and insulin resistance may also develop after chronic vascular inflammation; improvement of psoriasis can lead to a reduction in aortic vascular inflammation [12]. Additionally, patients with psoriasis are more likely to have subclinical atherosclerosis, greater carotid artery intimal thickening, and substantial endothelial dysfunction than the general population. The inflammatory nature of atherosclerosis and psoriasis has been highlighted recently; both are associated with T lymphocyte-mediated adaptive immune events and mechanisms involving innate immunity [13].

Alexandroff et al. [14] reported that obesity and psoriasis produce subclinical inflammation characterized by an increase in the levels of pro-inflammatory cytokines such as IL-6, TNF-α, and C-reactive protein (CRP).

Schäkel et al. [15] focused on the innate and adaptive immune system and found interactions among neutrophils, macrophages, TH1 lymphocytes, monocytes, platelets, and IL-17a, which have been implicated in the pathophysiology of psoriasis and atherosclerosis.

Chronic inflammatory systemic diseases (CISDs) are part of a spectrum of chronic diseases in which inflammation plays an important role in pathogenesis. CISDs and cardiovascular diseases, including atherosclerosis, share common pathogenic features in which inflammatory cytokines play an important role. Common pathogenic features include immunological processes (diseases mediated by Th-1 cells), inflammatory cytokine profiles, and the presence of inflammatory markers both locally and systemically. Activation of these inflammatory cells, together with the release of inflammatory cytokines and proteases, contributes to the development of psoriatic lesions and plays a major role in the breakdown of atherosclerotic plaques [14].

Boehncke et al. [16] found that reduced levels of adiponectin are associated with insulin resistance and several adverse vascular events and are inversely correlated with serum CRP in obese patients with diabetes and those with coronary artery disease. Risk factors for cardiovascular disease (obesity, insulin resistance, and dyslipidaemia) are associated with decreased levels of adiponectin and high levels of leptin. Elevated levels of leptin in patients with psoriasis are associated with overweight and obesity, but not with the severity of psoriasis [17].

The assessment of cardiovascular risk through scales helps in therapeutic decision making and in cardiovascular prevention. The following necessary variables were collected for the calculation of coronary risk using both the Registre Gironí del cor (REGICOR) and Systematic Coronary Risk Evaluation (SCORE) scales: age, sex, total cholesterol, high density lipoprotein (HDL) cholesterol, systolic blood pressure, diastolic blood pressure, smoking history, and antecedent of diabetes mellitus (DM).The calculation of the risk of cardiovascular death by SCORE is based on the figures of total cholesterol and for countries of low risk, it was based on the following: age, sex, total cholesterol, systolic blood pressure, and smoking history [18] or the JUPITER study where HDL was an inverse predictor of incident events and biomarker of residual risk [19].

However, there is a discrepancy in the estimation of these scales for cardiovascular risk since they have limitations. Comparative studies were performed among the SCORE, REGICOR, and Framingham scales in our setting, and discrepancies were found in the detection of high risk and therapeutic indications (SCORE favours intervention in women with hypertension and Framingham in men with dyslipidaemia). The SCORE has the aggravating circumstance that it only estimates cardiovascular mortality and not morbidity [18–20].

Other scales used to determine cardiovascular risk were the American Framingham and POOLED cohort [21]. In patients with moderate-severe psoriasis, it is probably convenient to multiply the score obtained according to Framingham or SCORE by 1.5 [22].

In Spain, the only valid scale is REGICOR, but it only measures coronary risk. European countries support the SCORE scale but it only measures mortality and does not include people over 65 years of age. Currently, some authors consider the English scale promoted by the National Institute for Health and Care Excellence (NICE) as one of the most complete. The scale is called QRISK. It is available online at https: //qrisk.org/three [23] is the 3rd version [24].

In any case it is convenient follow the guidelines proposed by Dauden et al. [2]. Following the Spanish recommendations, screening for cardiovascular risk factors should be performed every 6 months in patients with systemic treatment and yearly in patients with topical treatment.

In the study conducted by Cea-Calvo et al. [26], the presence of undiagnosed cardiovascular risk factors (CVRF) in patients with psoriasis was investigated. For this, 368 patients were recruited, with an average age of 48 years, 96% with psoriatic plaque, 23% with psoriatic arthritis, and 4% with other forms of psoriasis.

Through the measurement of blood pressure and fasting blood test, new cases of cardiovascular risk factors (CRFV) were detected in more than 27% of patients. In addition, the 10-year cardiovascular risk was similar to that of patients with known CVRF [26] macrophages.

The PCPs are important in the evaluation of cardiovascular risks in patients with psoriasis, because often they are the only physicians to treat and follow up with these patients. Their role can be more effective, if they possess sound knowledge of these pathologies and training to handle and treat them [27]. Research on this disease is primarily directed towards identification and characterization of new biomarkers. In this review, information about the main diagnostic and prognostic biomarkers, and their responses to different treatments, were collected from the patients of psoriasis with cardiovascular risk. These data can be useful in clinical practice.

Correlation between Psoriasis and Cardiovascular Functional Indices

Moore et al. [28] reported that the development of an atherosclerotic plaque is one of the most important risk factors. It is a multistep process and inflammation contributes to both the development of atheroma itself and the eventual rupture of the plaque; the first step is altered endothelial function with a change in the normal homeostatic responses; the next step is the recruitment of macrophages into the developing lesion and the accumulation of lipid-laden macrophages (foam cells) to form a fatty streak and the formation of a more advanced fibrous lesion, which is covered by a fibrous cap. Then, it gradually develops into an advanced and complex lesion, continuing in thinning of the fibrous cap to create a potentially unstable plaque that can eventually rupture, which leads to thrombosis and clinical evidence of vascular occlusion [28].

Family history of atherosclerosis, metabolic syndrome, diabetes, obesity, increased levels of inflammatory molecules (Figure 1) [29], discontinuity in the follow-up of patients [30] with newly diagnosed hypertension, diabetes, and hypercholesterolemia were associated with higher cardiovascular events, and associated mortality, escalating the financial cost of the health care.

JCRM Carmen Rodriguez-Cerdeira - 2020-302_F1

Figure 1.

Histopathological changes observed in psoriasis are due to important and complicated interactions between keratinocytes, dendritic cells, T lymphocytes, neutrophils, and mast cells. IL-23, IL-17, IL-22, Th-17 cells, Th-22 cells, regulatory T cells, TGF-β1, and IL-10 are involved in the pathogenesis of the disease. The IL-23/IL-17 axis is the main immune pathway in the pathogenesis of psoriasis. Multiple factors induce mDC activation with consequent IL-23 production (IFN-α, TSLP), which, in turn, mainly stimulates T-cell subsets and also ILC3, mast cells, and neutrophils, which secrete IL-17. Other cytokines derived from T cells, mast cells, and ILC3 (IL-22, IL-17F, and IL-21) and from mDCs (TNFα, NO, and IL-20) are responsible for the development of psoriasis. T17 and T22 cells represent all T-cell subsets producing mainly IL-17 and IL-22.

Chronically released proinflammatory cytokines in patients with poorly controlled psoriasis pass into the circulatory system and potentiate and perpetuate systemic inflammation. This systemic inflammation causes obesity, hypertension, dyslipidemia, and type 2 DM . Additionally, this leads to an increase in insulin resistance, endothelial dysfunction, and the onset of cardiovascular diseases.

 Environmental factors studied by Balak et al. [31] included infectious agents, a high-fat diet, smoking, alcohol consumption, low levels of antioxidants, and lack of exercise, as well as psychological stress, certain medications (antimalarial drugs, beta-blockers, lithium, and nonsteroidal anti-inflammatory drugs), and a history of skin disorders (e.g. psoriasis).

However, there is little data about subclinical alteration of myocardial function and adjacent vascular changes in patients with mild psoriasis. Milaniuk et al. [31] used two-dimensional stress echocardiography (2D-SE) for the quantification of alterations in all dimensions of the left ventricle. They reported that an increase in stiffness negatively affects cardiac function.

Dattilo et al. [33] evaluated cardiac performance and vascular rigidity in patients who had mild psoriasis and had no other risk factors or were undergoing treatment. The patients came from the department of Dermatology where they went periodically for follow up. Finally, 33 patients were included in the study sample. The control group consisted of healthy volunteers. The researchers found a significant correlation between global longitudinal strain and patient age. They also found that arterial stiffness values were significantly higher in psoriatic patients [33].

A study conducted by Sutton-Tyrrell et al. [34] showed arterial stiffness, a characteristic finding in patients with CISDs, is one of the most important determinants of increasing systolic blood pressure and pulse pressure; thus, it is a major cause of cardiovascular complications and events.

Ardic et al. [35] reported comparable left ventricle dimensions, wall thickness, and ejection fraction between patients with psoriasis and healthy controls.

Goga et al. [36] determined the presence of preclinical cardiac dysfunction in a cohort of 52 psoriatic patients without cardiovascular risk factors. They compared them with a control group. Both groups underwent echocardiographic conventional and tissue Doppler (TDI) studies. Left ventricular diastolic dysfunction was found in 36.5% patients in the psoriasis group versus 0% in control group. In addition, it was found that patients with psoriasis have a significant increase in mitral regurgitation [36].

Balbul-Sen et al. [37] conducted a cross-sectional and observational study with 65 patients with psoriasis and a control group. They verified that carotid intima-media thickness (CIMT) is a potential indicator of subclinical atherosclerosis in patients with psoriasis. Epicardial fat thickness (EFT) is proposed as a new cardiometabolic risk factor. They stated that EFT and CIMT are increased in patients with psoriasis, and that echocardiographic EFT will be correlated with CIMT in patients with psoriasis. This shows that echocardiographic study through EFT may be a marker of subclinical atherosclerosis and increased cardiovascular risk in patients with psoriasis.

Another comorbidity of psoriasis considered by Gisondi et al. [38] is the non-alcoholic fatty liver, which is currently considered to be the hepatic manifestation of metabolic syndrome. This condition is linked to an increased risk of future cardiovascular events independent of conventional risk factors [39].

Schiffrin et al. [40] found that a decreased flow velocity in the brachial artery indicated a risk of developing coronary artery disease.

El-Mongy et al. [41] reported that patients with psoriasis had more prevalent valvular regurgitation, abnormal diastolic relaxation, left ventricular hypertrophy, left ventricular diastolic dysfunction, left ventricular wall motion abnormalities. Mitral valve and tricuspid valve prolapse were found to be more prevalent in patients with psoriasis in a few studies, but in other studies, these results were not confirmed.

Echocardiographic findings from Biyik et al. [42] demonstrated that compared with the control group, patients with psoriasis had greater aortal diameter, greater septal and posterior wall thickness, more frequent tricuspid and mitral regurgitation, and increased systolic pressure in the right ventricle. Early atherosclerotic echocardiography predictors were found in the patients with psoriasis.

Goyal et al. [43] reported that psychological stress and depression have been increasingly in recent years and have been associated with multiple traditional cardiovascular risk factors. The amygdala regulates physiological and behavioural changes in response to stress and fear; using 18-fluorodeoxyglucose positron emission tomography computed tomographic (FDG PET/CT) imaging, the resting metabolic activity in the amygdala can be reproducibly assessed. Measures of stress-related neural activity are associated with anxious temperament and are upregulated in anxiety disorders, chronic stress, and depression. In 2017, Tawakol et al. [44] studied the importance of amygdalar activity (AmygA) with increased bone-marrow activity and arterial inflammation but they studied patients who did not have psoriasis. Goyal concluded that AmygA is strongly associated with aortic vascular inflammation (VI) and risk of major cardiovascular events. This association is partially mediated by upregulated hematopoietic system activity (HMPA). Psoriasis patients have elevated rates of anxiety and depression, and is also an independent factor directly related to development of CVD.

A retrospective study was conducted by Shiba et al. [45] to analyse the relationship between psoriasis and myocardial infarction, diabetes, and dyslipidaemia. They included more than 100,000 patients of all ages and performed multiple statistical analysis with the following results: patients with psoriasis have a higher prevalence of hypertension, DM, myocardial infarction, and hyperlipidaemia. There is an independent association between psoriasis and myocardial infarction.

Correlation between Cardiovascular Risk and Psoriasis Severity Indices

The importance of the severity of psoriasis in the development of cardiovascular disease is also supported by evidence of a correlation between PASI values and insulin secretion. Insulin resistance is a characteristic feature of metabolic syndrome. In patients with psoriasis, a statistically significant correlation has been observed between serum levels of resistin (a cytokine that is elevated in insulin resistance situations) and PASI [46]. These data indicate that the severity of psoriasis is related to an increased risk of cardiovascular complications and point to the pathogenic importance of a state of chronic inflammation in the development of vascular disease in these patients.

Prodanovich et al. [47] found a relationship between the severity of psoriasis with the risk for atrial fibrillation and ischemic cerebrovascular disease, with an atrial fibrillation incidence of 3.03 in patients without psoriasis compared to 5.96 in patients with severe psoriasis; similar figures were detected for ischemic cerebrovascular disease.

The risk score measured with the Framingham scale by Eder et al. [47] was significantly higher in patients with chronic plaque psoriasis than in the control group (grouped by age and sex). In patients older than 50 years, there was no correlation between the risk score and the severity of PASI [48].

To relate the severity of psoriasis to vascular inflammation, Naik et al. [49] recruited 60 patients with psoriasis and 20 controls, and measured the severity of the disease through PASI and vascular inflammation with the use of FDG PET/. They demonstrated that severe psoriasis is associated with atherosclerotic disease, and the presence of the disease alone increases cardiovascular risk, coupled with concomitant risk factors [49]. The contribution of PASI score in predicting vascular inflammation (aortic target-to-background ratio) beyond Framingham risk score and high-sensitivity C-reactive protein (hsCRP) was determined using likelihood ratio testing in nested models PASI score. The authors found that the severity of psoriatic skin disease increases vascular inflammation independently [49].

In a review by Prey et al. [50], they investigated the psoriasis cases (with different degrees of severity) and cardiovascular risk factors described in the literature between 1980 and 2009. Finally, 18 articles were selected from the main medical databases. There was an association between diabetes and psoriasis in 11 studies. The risk of diabetes was higher in patients with a moderate or severe degree of psoriasis [51, 52]. Three studies showed an association between hypertriglyceridemia and psoriasis, and only one study showed an association between psoriasis and total cholesterol levels. None of the studies showed an association between psoriasis and risk of dyslipidaemia or HDL cholesterol levels.

The risk of hypertension was described in 10 studies compiled by Herron et al. [53] but a relationship between the level of risk and the degree of severity of psoriasis was not established. Eight articles showed the risk of obesity in psoriasis and reported that the risk increases directly depending on the severity of psoriasis. All articles showed an increased risk of metabolic syndrome (MS) in patients with psoriasis [53].

Chronic stress-related neural activity as measured by AmygA was higher in patients with severe PASI. Increased AmygA was related to the severity of psoriatic skin disease and subclinical cardiovascular disease (CVD). The amelioration of psoriasis with therapy was accompanied by reductions in AmygA, HMPA, aortic vascular inflammation, and stabilization of non-calcified coronary plaque burden (NCB). Stress affects the endocrine system, leading to upregulation of circulating catecholamines, glucocorticoids, and inflammatory cytokines, which subsequently leads to CVD. Psychological stress should be considered in evaluating CVD risk, especially in inflammatory disease states. The effect of reducing inflammation, as proven by improvement in PASI severity, on AmygA, HMPA, aortic VI, and NCB, [43].

In a study conducted by Uyar et al. [54], the mean serum concentrations of fetuin-A, alanine aminotransferase (ALT), and rheumatoid factor levels were found to be higher in patients with psoriasis. Median CIMT levels were significantly higher in patients with mild-to-moderate psoriasis compared to controls, but no correlations were found among the serum fetuin-A levels, CIMT levels, and PASI scores.

Rosa et al. [55] showed that 75% of patients with moderate or high risk presented a with a PASI below 10. Thus, although psoriasis patients had an increased cardiovascular risk, there was no relationship between severity of the cutaneous manifestations and a higher risk.

In a study by Gisondi et al. [56], using the Framingham risk score, they found the severity of cutaneous manifestations, measured by PASI, was associated with an increased cardiovascular risk.

Sorokin et al. [57] demonstrated that patients with psoriasis have a relative and absolute increase in circulating monocyte aggregates as well as an increase in intermediate monocytes, which correlates with an increase in the severity of PASI.

Sphingolipids, specifically ceramides (CER) and sphingosine-1-phosphate (S1P), are signal molecules that are responsible for regulating cell growth, apoptosis, and immune reactions. Myśliwiec et al. [58] demonstrated that circulating levels of CER and S1P in plaque-type psoriasis were associated with disease activity through the PASI measure.

Women, psoriasis and CV risk

Interestingly, there were few instances of psoriatic women with increased risk of developing CVD. Some studies grouped patients based on sex, however, CVD risk was not always reported with patient sex. In this section, we collected information from the few studies that observed CVD risk in psoriatic women.

In Denmark, Blegvad et al. [59] studied 2,435 women with psoriasis (2.90%) and 81,388 women without psoriasis and concluded that psoriasis is significantly associated with hypercholesterolemia and hypertension. Women with psoriasis have an increased risk of developing cardiometabolic disorders in early adult life. Screening for classic risk factors such as hypertension, hypercholesterolaemia, and hyperglycaemia, is advised. Additionally, preventing type 2 diabetes may be important when treating patients with psoriasis, particularly in younger patients.

In a study by Pietrzak et al. [60], 432 patients with psoriasis and a control group of 546 were selected. Both psoriatic and control patients showed that CVD occurrence increased with age and concluded that: a) the incidence of psoriasis and CVD in women increases with age, b) it is unclear whether menopause increases the risk of psoriasis, c) it is unclear whether menopause and resulting hormonal disturbances are independent CVD risk factors, and d) investigating whether hormone replacement therapy reduces CVD risk in psoriatic patients may be important.

Li et al. [61] conducted several studies on psoriatic women and the risk of developing CVD. In particular, the study published in Nurses’ Health Studies (NHS) collected epidemiological information about the health status of women in different diseases, showing a strong relationship between psoriasis and CVD. In another study, there was a significantly higher risk of type 2 diabetes in psoriatic patients, and that this occurred particularly among younger women who had developed psoriasis at an early age [62]. Some of these patients eventually developed CVD. Li et al. [63] also showed that women with psoriasis had increased non-fatal CVD resistance, especially myocardial infarction. Therefore, women who developed psoriasis earlier, or those who have had it for a long time, presented an increased risk of myocardial infarction.

Prognostic Biomarker

The onset of psoriasis at an early age increases the risk of myocardial infarction. This appears to be due to an increase in the time of exposure to a chronic systemic inflammation. In a cross-sectional study with 301 participants, Kumdai et al. [64] found that certain HLA antigens, including A30, B50, Cw6, and DR7.20, were associated with early-onset psoriasis.

On the other hand, Wongpiyabovorn et al. [65] reported that IL-10 distal promoter polymorphisms are usually associated with late-onset disease.

Oxidized LDL (OxLDL) is very important in the exacerbation of atherosclerosis; thus, it is an important factor in the treatment of cardiovascular disease. In psoriasis, the efflux capacity of cholesterol is decreased, which changes after treatment. Activated oxidation lipid mediators (OMLs) abound in psoriatic skin, playing a critical role in the process of initial inflammation and resolution. A study by Sorokin et al. [57] with 252 patients with psoriasis and a control group measured LDL with modified oxidation, HDL, lipoprotein A, cholesterol efflux capacity, and size and number of lipoproteins. They observed an increase in oxidized Lp (a), Lp (a), and oxidized HDL. The activity of paraoxonase-1 was increased by possible compensatory oxidative effect. It was concluded that psoriasis is associated with increased levels of some OMLs as opposed to healthy individuals. OxLDLs have a direct association with the non-calcified plaque. The authors suggest that serum levels of OML could be useful as early markers in atherosclerosis by coronary computed angiography, which allows the quantification and characterisation of non-calcified plaque in the coronary arteries [57].

Dinić et al. [66] recruited 66 patients with chronic psoriasis plaque. The control group consisted of 20 patients with generalized eczema and 20 healthy controls, all of whom were between 18 and 60 years old, had no associated comorbidities, and underwent systemic treatment. Blood samples were taken and analysed. The values for the following were obtained: erythrocyte sedimentation rate (ESR), fibrinogen, PCR, Dimer-D, serum amyloid A, Apolipoprotein (Apo) A1, Apo B, Apo B / Apo A1 ratio, serum concentration of uric acid (SUAC), fasting insulin, Peptide-C, creatinine clearance obtained by Cockroft-Gault formula, microalbuminuria and proteinuria in 24 h. Colour duplex Doppler ultrasound was also performed to measure the following: arteriovenous resistance; the thickness of the medial intima of the common carotid artery distal to the posterior wall of the carotid bifurcation; the femoral artery and its resistance and pulsatility indices; and maximum flow velocity of the femoral and brachial arteries. B-mode ultrasound was used. Patients with psoriasis presented with the parameters associated with cardiovascular risk, in addition to metabolic syndrome, even in the absence of other risk factors [66].

Three distinct monocyte populations have been identified and genotyped within human blood: classical monocytes (CD14 ++ CD162), intermediate monocytes (CD14 ++ CD16 +), and nonclassical monocytes (CD14 + CD16 ++). Among the results obtained by Golden et al. [57], it was found that patients with psoriasis have a higher percentage of intermediate circulating CD14 + CD16 + monocytes compared to controls. This was obtained through peripheral blood samples, and/or biopsies in healthy controls and in patients with psoriasis. Systemic or topical therapy were not used for the preparation of mononuclear cell preparations in peripheral blood. It was also shown that the classical monocytes subset (CD14 ++ CD162) correlates negatively with PASI, and the nonclassical monocytes (CD14 + CD16 ++) do not show any correlation [67].

The vascular uptake of fluorodeoxyglucose (FDG) is associated with unstable atherosclerotic plaques. A relationship was found between arterial inflammation detected by FDG PET/CT and the presence of high-risk coronary plaque, which is mainly composed of cells of the psoriatic plaques. This suggests a potentially shared immune mechanism and demonstrates the role of neutrophils in the perpetuation of psoriasis and their comorbidities; the downregulation of CD16 and CD62L in this sample indicates a decreased ability to mediate inflammatory damage and a greater activation state. It was also reported that S100A8/A9 proteins may serve as mediators or by-products in one of the potential pathays linking psoriasis severity and arterial inflammation, confirming the role of neutrophils in cardiovascular disease associated with psoriasis [49].

Ryan at al. [68] found microparticles present in the atherosclerotic plaque breakdown, which causes cardiovascular events. They also showed an increased concentration in these microparticles in patients with psoriasis.

Homocysteine is an amino acid produced in the liver that is involved in the pathogenesis of several diseases, including psoriasis. It increases the levels of oxidative stress and inflammation, and induces apoptosis of endothelial cells, which favours atherosclerosis; atherosclerosis is the most common cause of cardiovascular pathology such as myocardial infarction or heart failure. Homocysteine levels can be used as predictors for cardiovascular risk, and they are directly related to the severity of psoriasis [69]. Homocysteine plays an important role in the DNA methylation cycle, which is a process related to epigenetic mechanisms. Wang et al. [70] reported that several studies have observed that hyperhomocysteinemia reduces levels of DNA methylation, which is related to the pathogenesis of psoriasis. Genes that encode proteins such as HLA-DRB1, protein tyrosine phosphatase 1 (SHP-1), or different high proliferative potential colony-forming cells (HPP-CFCs) are hypomethylated.

Qi at al. [71] reported that methylene tetrahydrofolate reductase (MTHFR) participates in the irreversible conversion of a cofactor that participates in the remethylation of homocysteine to methionine. Only one polymorphism of the MTHFR gene (677C> T) is related to psoriasis, and there are reports of the relationship between this polymorphism and cardiovascular diseases. This polymorphism is not a risk factor in the pathogenesis of psoriasis, but it can influence the severity of psoriasis.

Psoriasis in combination with obesity/MS produces greater systemic inflammation and greater oxidative stress, which promotes endothelial dysfunction and the formation of atherosclerotic plaques, which increase the risk of cardiovascular disease. Kaur et al. [72] found that obesity and psoriasis produce subclinical inflammation characterized by an increase in the levels of pro-inflammatory cytokines such as IL-6, TNF-α, and CRP. The levels of CRP and IL-6 are related to the severity of psoriasis.

Psoriatic skin disease severity showed a linear relationship with AmygA, and was associated positively with subclinical cardiovascular disease, a higher Framingham risk score, more insulin resistance, and higher CRP levels. When psoriatic skin disease severity improved, there was a significantly concurrent decrease in AmygA, bone marrow activity, and aortic elasticity, and found beneficial effects of skin disease clearance on the amygdala and aorta after one year of treatment [73, 74].

The CRP is an acute phase reactant protein produced from hepatocytes within hours after being stimulated from infection, inflammation, or tissue damage. Its levels decrease rapidly after the stimulating factor has been eliminated or has responded to treatment. Uaratanawong et al. [75] showed that the hs-CRP level was significantly higher in the psoriasis group with a significant correlation between hs-CRP and PASI score after controlling for other confounding factors. A hs-CRP level greater than 0.69 mg/L could be the marker of worsening psoriasis. The keratinocytes in psoriasis lesions secrete tumour necrosis factor-α and interleukin-1, which can stimulate hepatocytes to produce hs-CRP. Furthermore, hs-CRP level can predict cardiovascular risk, including cerebrovascular and peripheral arterial disease (hs-CRP less than 1 mg/L as a low risk, 1–3 mg/L as intermediate risk, and more than 3 mg/L as high risk for cardiovascular event). Although hs-CRP is not a specific marker and additional work will be imperative to validate this marker, it shows the inflammatory burden of psoriasis and could be a potential marker that can be used for grading psoriasis severity with great cost-effectiveness.

When assessing coronary artery disease and generalized atherosclerosis, CIMT is a good indicator of an increased risk of subclinical atherosclerosis. Fetuin-A is a serum glycoprotein that is synthesized mainly by hepatocytes, increases insulin resistance and dyslipidaemia and reduces ectopic calcification in vascular disease. There are reports on fetuin-A interacting with transforming growth factor (TGF)-β and epidermal growth factor (EGF), which play an important role in the pathogenesis of psoriasis [76].

Ix et al. [77] reported that lower fetuin-A levels initiate calcium deposition within the vasculature but not atherosclerosis directly, and that they are independently associated with greater coronary artery calcification severity but not peripheral arterial disease. They observed a correlation between serum fetuin-A levels and fasting glucose, as well as between fetuin-A and LDL cholesterol, so it is recommended that physicians should measure CIMT rather than the less appropriate serum fetuin-A values when assessing the cardiovascular disease risk of patients with psoriasis.

Serum adipokines have been considered in the pathogenesis of the psoriasis and have been demonstrated to be associated with insulin resistance. Adiponectin, resistin, and leptin have possible antiatherogenic and anti-inflammatory properties by regulating energy homeostasis and increasing insulin secretion and sensitivity. Hyperleptinemia has contributed to cardiovascular risk in patients with the metabolic syndrome; adiponectin and leptin are predictors of microvascular complications in patients with type 1DM [17].

A study by Yazıcı et al. [78] included 65 patients with type 1 DM. Leptin levels were reported to be decreased in adult patients with type 1 DM without association with CIMT. Resistin levels were decreased in adult patients with type 1 DM and increased in patients with type 2 DM with CAD and was correlated positively with CIMT; increased resistin levels in patients with type 1 DM seem to be suggestive of subclinical atherosclerosis. Leptin was associated with CIMT in patients with type 2 DM. Thus, after adjusting for other known predictors, only resistin was associated with subclinical atherosclerosis in this group of patients. Ataseven et al. [79] suggest that vaspin and VAP-1 may play a role in the pathogenesis of psoriasis and can be used as markers of the disease. Therefore, vaspin levels can be used as a marker of the severity of the disease.

Diagnostic Biomarkers

A strong expression of chemerin was observed by Nakajima et al. [79] in pre-psoriatic skin adjacent to active lesions and early lesions in the dermis. They also observed the presence of CD15+ neutrophils and CD123+/BDCA-2+/ChemR23+ pDC. In skin from chronic plaques, chemerin expression was lower. Additionally, the segregation of neutrophils in epidermal microabscesses and a few plasmatoid dendritic cells (pDCs) in the dermis were observed [80].

Chemerin stimulates the chemotaxis of pDCs and neutrophils, and high systemic chemerin level was found as an independent marker of the metabolic syndrome. Recently, Albane   si et al. [81] reported a strong expression of chemerin together with increased numbers of pDCs and neutrophils in the dermis of early psoriasis lesions.

Furue et al. [82] reported that condition which have been proposed include shared genetic factors, common inflammatory pathways, secretion of adipokines, insulin resistance, altered lipoprotein composition and function, angiogenesis, oxidative stress, microparticles, and hypercoagulability; CRP, Th1, and Th17 lymphocytes, resistin and leptin has been found to be elevated, with decreased serum levels of adiponectin. Psoriasis may induce systemic inflammation leading to insulin resistance, endothelial dysfunction, and the development of atherosclerosis and cardiovascular comorbidities in the psoriatic march.

Identification of serum biomarkers (CRP, soluble CD40 ligand, human matrix Gla protein and fetuin-A, serum YKL-40, GlycA, and complement C3) of systemic inflammation is important for assessing cardiovascular risk; adequate treatment for psoriasis may decrease the risk of cardiovascular comorbidities as a result of suppression of systemic inflammation [12].

A group of 74 patients with psoriasis and 65 healthy controls were studied by Vachatova et al. [83]. Individuals with insulin resistance, diabetes, and cardiovascular disease were included in the psoriasis group. None of them had treatments that could influence inflammatory reaction. Patients with psoriatic arthritis were excluded. The patients with psoriasis presented with higher diastolic blood pressure, MS, BMI, and hip circumference compared to the controls. Patients with psoriasis had higher levels of leptin, resistin, and lipoprotein-associated phospholipase A2 (Lp-PLA2) than controls. Among individuals with MS, those in the psoriatic patients group had higher CRP levels of leptin, resistin, and Lp-PLA2 than those of CG. Patients with psoriasis and MS had higher levels of leptin and Lp-PLA2 and lower levels of adiponectin than patients with psoriasis without MS.

The combination of psoriasis and MS increases the expression of inflammatory and anti-inflammatory cytokines and increases the risk of cardiovascular disease. Likewise, adipokines (leptin, adiponectin, and resistin) are involved in the pathogenesis of psoriasis and MS, and the levels of Lp-PLA2 indicate the presence of subclinical atherosclerosis and cardiovascular risk in patients with psoriasis.

Kiluk et al. [84] found that Lp-PLA2 is associated with an increased incidence of cardiovascular events but only a few studies have associated it with psoriasis.

Metabolomics may potentially explain the mechanisms of various diseases, facilitate early diagnosis, and identify possible therapeutic areas.

Wang et al. [85] presented a utility (mmPredict) to determine metabolomic biomarkers, which is complemented by current studies of metabolomic profiles that are based on patient data. This utility reveals the metabolites of psoriasis based on both genetic and phenotypic relevance (genome-phenome). It was shown in the study that mmPredict is more effective in identifying clinically relevant biomarkers in psoriasis than the genome-based approach. They are improving mmPredict to incorporate other types of data such as the phenotype of drugs and high-level diseases observed in humans.

Another study, conducted by Coimbra et al. [86], showed that inflammatory cytokines were involved in the pathogenesis of psoriasis such as IL-22, IL-17, IL-23, IL- 8, TNF-α, and endothelial growth factor.

Ellulu et al. and Rodríquez-Cerdeira et al. [87, 88] reported that reduced levels of adiponectin are associated with insulin resistance and several adverse vascular events and are inversely correlated with serum CRP in obese patients with diabetes and those with coronary artery disease. Risk factors for cardiovascular disease (obesity, insulin resistance, and dyslipidaemia) are associated with decreased levels of adiponectin and high levels of leptin. Elevated levels of leptin in patients with psoriasis are associated with overweight and obesity, but not with the severity of psoriasis.

Excess fatty tissue, due to obesity associated with psoriasis, contributes to atherogenic dyslipidaemia, and several studies have associated atherogenic dyslipidaemia with increased blood levels of total cholesterol, triglycerides, LDL, and apolipoprotein A, and low levels of HDL and apolipoprotein B in patients with psoriasis.

In a study by Asha et al. [89], ox LDL is considered a marker of hyperlipidaemia and atherosclerosis. A few studies have shown increased levels of oxLDL in patients with psoriasis, which was consistent with the BMI. oxLDL/LDL ratio a better predictor for atherosclerotic risk as compared to LDL

Advanced oxidation protein products (AOPPs) and adipokines are also involved in the pathogenesis of endothelial dysfunction and atherosclerosis.

In other study conducted by Haberka et al. [90], patients with non-severe psoriasis had significantly increased impaired endothelial function and CIMT, and significantly increased serum levels of AOPPs and visfatin. Increased oxidative stress and serum visfatin levels are associated with a chronic inflammatory state; increased AOPP levels are associated with endothelial dysfunction (FMD), vascular remodelling CIMT, and increased serum visfatin levels, suggesting that increased oxidative stress is the main pathomechanism involved in cardiometabolic complications found in non-severe psoriasis.

In patients with psoriasis with a risk allele of FTO gene rs9939609, Tupikowska-Marzec et al. [91] suggested that they have an increased risk for more severe psoriasis and obesity, and may develop obesity-induced insulin resistance and therefore, CV risk.

As Barrera et al. [92] Vit D ot only to reduce its clinical severity, but also for delineating the risk profile for co-morbidities cardiac risk factors that may result from psoriasis [93].

Recently, a study in women conducted by Hruska et al. [94] showed a strong relationship between single nucleotide polymorphism (SNP) rs4597342 within the3’untranslated region (3’UTR) of integrin alpha M (ITGAM) with psoriasis susceptibility and severity. The associated risk was observed specifically for allele T of rs4597342, which introduces a binding site for miR-21 that is upregulated during psoriasis [94]. These markers are also linked to IgA nephropathy [95] and type 1 diabetes [96], which could be interesting diagnostic markers for CVD

Treatment Biomarkers

Reviewing the literature, we observed that patients with psoriasis receive suboptimal and disconnected attention and it leads to a decrease in the quality of life. As proposed by Gulliver et al. [97], the concept of treat-to-target, which has already been adopted for other conditions such as diabetes and dyslipidaemia, would be very beneficial for patients with psoriasis.

Treatment regimens should be individualized according to the risk factors of the patient, especially those with cardiovascular risk. The use of effective treatments is very important to help improve patient outcomes.

According to Caiazzo et al. [98], the introduction of biological therapy has greatly improved our therapeutic expectations and long-term control of the disease, and there is epidemiological evidence that it can also improve cardiovascular risk, as in patients with rheumatoid arthritis. However, some adverse effects of treatment with agents that block TNF-α have been described in patients with advanced congestive heart failure. Thus, special caution is required when these drugs are used in these patients.

Interleukins have recently been introduced or are under study for the treatment of psoriasis. Krueger et al. [98] reported that ustekinumab and briakinumab (ABT-874) are human monoclonal antibodies that target the p40 subunit common to both IL-12 and IL-23. Ustekinumab and other IL-12/23 inhibitors can increase the risk of cardiovascular adverse events, including st roke, MI, and cardiovascular death [99]. The main inflammatory cytokines involved in CV are summarized in Table 1 [88, 100–109).

Table 1. Involvement of inflammatory cytokines in cardiovascular risk

Obesity

IL-17A expression is upregulated in obesity [88].

Cytokines such as TNF-α and IL-6 are responsible for the pro-inflammatory state of obesity [100].

Adipose tissue of individuals with obesity leads to the development of Th17 cells [88].

Diabetes

IL-8, IL-17, IL-18 and IL-6 levels are increased in diabetes, leading to insulin resistance. Increased levels of IL-6 and IL-1β also lead to insulin resistance [101].

TNF-α induces insulin resistance by reducing the tyrosine kinase activity of the insulin receptor [102, 103].

Atherosclerosis

Cytokine products of Th1 cells such as TNF-α lead to endothelial dysfunction and consequently cause T-cell movement to atherosclerotic plaques. IL-17 contributes to vascular and systemic inflammation in experimental atherosclerosis in murine models [104].

Modulating the Th-17/IL-17 pathway might play a key role in the progression of coronary atherosclerosis [105].

Heart failure

Increased serum/plasma levels of TNF-α and IL-6 promote metabolic syndrome [106].

TNF-α levels show a linear relationship with prognosis [107].

Congestive heart failure is inversely associated with IL-6 levels [108].

Non-alcoholic fatty liver disease

TNF-α exacerbates hepatic insulin resistance, resulting in increased free fatty acid synthesis and decreased free fatty acid oxidations, thereby promoting hepatic steatosis. Thus, the relationship between non-alcoholic fatty liver disease and diabetes mellitus type 2 is complex and bidirectional [109].

In a study by Papp et al. [110] and Spuls et al. [111] with anti-interleukin-17-receptor antibody treatment, they concluded that patients with plaque psoriasis significantly improved. These were especially effective in hard-to-treat areas such as the scalp and nails; significant differences were observed with placebo with ixekizumab treatment. There was no evidence of cardiovascular risk in the treated patients. A more recent study by Lockshin et al. [112] was also not conclusive in terms of CV risk assessment.

The participation of nails also as Ventura et al. [113], it correlates positively with the early onset of psoriasis in the populations of Europe and America, which would be a marker of initiation of treatment as soon as possible.

Shih et al. [114] reported that the treatment of psoriasis with anti-inflammatory biological therapy reduces levels of oxHDL together with a decrease in non-calcified plaque in one year, so they suggested that serum levels of OML could be useful as early markers in atherosclerosis. These were measured with coronary computed angiography, which allows the quantification and characterization of the non-calcified plaque in the coronary arteries. It will be very important to investigate the factors for the oxLDL induction of LOX-1 in psoriasis. LOX-1 receptor expression could be another novel treatment option for psoriasis and represents a very interesting and promising strategy.

In the vascular wall, oxLDL, cholesterol, and nicotine have the ability to induce IL-23 secretion by DCs. Abbas et al. [115] and Di Cesare et al. [116] reported a considerable associated between oxLDL and IL-23. In dyslipidaemia, oxLDL binding to LOX-1 could induce endothelial dysfunction, macrophage foam cell formation, and smooth muscle cell migration and proliferation. Additionally, the relationships among the oxLDL receptor, IL-23, and LOX-1 still need to be studied.

Glycoprotein acetylation (GlycA) is a recent biomarker for systemic inflammation that can predict CV events, risk of serious infection, or type 2 diabetes [117]. In addition, it has shown good results in the assessment of the pathology or in the response to treatments. To assess its use as a biomarker in psoriasis, a study by Joshi et al. [118] included patients with mild to moderate psoriasis and control patients, for a total of 412 participants. They observed that there is a correlation between GlycA and high sensitivity C-reactive protein (hsPCR) in patients with psoriasis. In addition, there is also a correlation between GlycA and the severity of psoriasis. There are other risk factors that correlate with GlycA in psoriasis, such as BMI, waist-hip ratio, metabolic syndrome, HDL cholesterol, and LDL cholesterol. GlycA is associated with vascular inflammation (LV) and coronary artery disease (CAD). However, CAD is not associated with hsPCR in patients with psoriasis, but in controls. This indicates that GlycA is associated with cardiovascular disease (CVD) independently of traditional risk factors. After treatment with anti-TNF therapy in a group of 16 patients with psoriasis, GlycA levels were observed to have decreased, compared with baseline; however, this decrease was not the same as that of hsPCR levels which was statistically non-significant. In the treated patients, aortic VI also decreased.

Each of the patients studied by Montaudié et al. [119] had a comorbidity, and 45% had two or more. When treatment with biological agents (infliximab, adalimumab, etanercept and ustekinumab) was used, a statistically significant decrease was observed only for the inflammatory parameters (CRP, P = 0.004) and the erythrocyte sedimentation rate (ESR, P = 0.002).

In a study by Jiang et al. [120], adipokine imbalance was associated with chronic inflammatory state and insulin resistance. Psoriasis seems to be an independent risk factor of CV complications. Biological therapy including efalizumab and alefacept, primarily disrupt the activation and migration of T cells, whereas agents like infliximab, etanercept, and adalimumab target TNF-a. Recently, agents, which target the p40 subunit shared by both IL-12 and IL-23, have been developed, as well as new anti-IL-17 agents and anti-IL-23p19 agents, was found to improve endothelial function and reduce carotid vascular remodelling [121, 122].

Several authors, such as Späh et al. [123] have demonstrated that antirheumatic drugs, such as methotrexate, reduce the risk of cardiovascular disease in patients with psoriasis. Other biological therapies used in the treatment of psoriasis such as etanercept, infliximab, and adalimumab, could have a beneficial effect on cardiovascular function in addition to reducing the levels of systemic markers of inflammation, such as ESR and CRP. [124]. Other authors as Ridker et al. ( CIRT study) no benefit founded with low-dose of methotrexate for prevention the atherosclerotic events or CVD [125]

However, new therapeutic targets are controversial, since recent studies on anti-IL12/23 and anti-IL17 suggested that they could increase CVD risk in psoriatic patients. Rungapiromnan et al. [126] compiled data from the Cochrane Library, MEDLINE, Embase, U.S. Food and Drug Administration, European Medicines Agency, pharmaceutical companies, and five clinical trial registers. They found that there were no adverse effects during the short, randomized, and controlled clinical trials

Finally, a study conducted by Lee et al. [127] with 60,028 patients with psoriasis were included and treated with Ustekinumab (Us) vs TNF Inhibitor (TNFi). No substantially different risk of incident of atrial fibrillation and major adverse cardiovascular events after initiation of Us vs TNFi was observed in this study

The main biomarkers of inflammation involved on diagnose, prognosis and treatment in patients with psoriasis and cardiovascular risk factors, are collated in Table 2 [12, 17, 49, 57, 64, 65, 67–84, 86–91, 92–94, 99, 114–120].

Table 2. Biomarkers of inflammation in patients with psoriasis and cardiovascular risk factors

TYPE

BIOMARKERS

REFERENCES

PROGNOSIS

HLA antigens (A30, B50, Cw6 and DR7.20)

[64]

Metabolic Syndrome

[83]

Oxidation lipid mediators (OxLp(a), OxHDL, OxLDL)

[57]

Monocytes (CD14+, CD16+) // Monocytes (CD14++, CD162)

[67]

Fluorodeoxyglucose (FDG)

[49]

S100A8/A9

[49]

Downregulation of CD16 and CD62L

[49]

Microparticles present in the atherosclerotic plaque breakdown

[68]

Homocysteine

[69, 70]

Tetrahydrofolate reductase (MTHFR)

[71]

Cytokines (CRP, IL-6, IL-10)

[65 72, 75]

AmygA

[73, 74]

Fetuin-A // Carotid intima-media thickness (CIMT)

[76, 77]

Adiponectin, resistin, leptin, vaspin

[17, 78, 79]

DIAGNOSIS

Chemerin

[80, 81]

CD15+ neutrofils

[80]

CD123+/BDCA-2+/ChemR23+ plasmatoid dendritic cells

[80]

CRP

[12, 82, 83]

Soluble CD40 ligand

[12]

Fetuin-A // Carotid intima-media thickness (CIMT)

[12, 90]

YKL-40

[12]

GlycA

[12]

Complement C3

[12]

Human matrix Gla protein

[12]

Adiponectin, resistin, leptin

[82, 83, 87, 88]

Lymphocytes (Th1, Th17)

[82]

Lp-PLA2

[83, 84]

Cytokines (IL-22, IL-17, IL-23, IL- 8, TNF-α, EGF)

[86]

oxLDL/LDL ratio

[89]

Advanced oxidation protein products (AOPPs)

[90]

Visfatin

[90]

FTO allele (rs9939609)

[91]

Vitamin D

[92, 93]

The major allele T of rs4597342 -ITGAM (only female)

[94]

TREATMENT

Interlekines (IL-12, IL-23, IL-17)

[99,110, 111, 115, 116]

Oxidation lipid mediators (OxHDL, OxLDL)

[114, 115, 116]

LOX-1

[114, 115, 116]

GlycA

[117]

CRP

[118]

Carotid intima-media thickness (CIMT)

[119]

ESR, CRP

[120]

Conclusions

The association between psoriasis and cardiovascular risk factors has been recently demonstrated by several groups, especially in young patients with severe forms of psoriasis. This association implies that psoriasis should not be exclusively considered as a cutaneous or joint disease, but rather as a chronic inflammatory disease with systemic involvement. Thus, in addition to specifically treating cutaneous symptoms in patients with psoriasis, managing classic risk factors involved in atherogenesis is paramount.

PCPs are in an important position to detect and respond to early indicators of CVD risk and comorbidities in their patients, they working collaboratively with dermatology health professionals and other specialists, PCPs can facilitate communication about the patient’s treatment preferences and personalized for each patient to optimize treatment in each case. Moreover, as mechanisms involved in psoriasis and CVD are further elucidated, future therapeutic strategies may involve inhibition of pro-inflammatory cytokines that are involved in both diseases.

Since psoriasis carries a risk of developing CVD that is probably due to systemic inflammation, there is a need for new prognostic, diagnostic, and treatment biomarkers. The purpose of these biomarkers is to improve diagnosis for intervention by reducing inflammation, which could reduce or eliminate CVD risk in psoriatic patients.

These biomarkers should be used particularly in primary care to detect and filter patients with psoriasis that have elevated risk of developing CVD.

Therefore, these biomarkers allow for rapid detection of high-risk psoriasis patients for myocardial infarction and heart failure to avoid complications. Since detection of these biomarkers is highly sensitive and specific, it should be routinely implemented in healthcare systems around the world.

Conflicts of Interest

The authors declare no potential competing interests with respect to the research, authorship and/or publication of this article.

Funding Statement

No funding was received.

Abbrevations

CV: Cardiovascular

CRP:   C-reactive protein

IFN:  Interferon

IL: Interleukin

LL37: Cathelicidin

PMN: Polymorphonuclear neutrophil

S-100: S-100 proteins

Th: T-helper cells

TGF-β: Transforming growth factor-beta

Tn: Naïve T lymphocyte

TNF: Tumor necrosis factor

CCL: CC chemokine ligands

CXCL: Chemokine (C-X-C motif) ligand

β-DEF: β-defensins

KC: Keratinocyte

mDC: Myeloid dendritic cell

pDC: plasmacytoid Dendritic Cells

LCN2: Antimicrobial peptide

NKT: Natural killer T cell

TSLP: Thymic stromal lymphopoietin

NO: Nitric oxide

pDC: Plasmacytoid dendritic cell

TSLP: Thymic stromal lymphopoietin

References

  1. Gudjonsson JE, Elder JT. Psoriasis. (2012) In: Goldsmith LA, Katz SI, editors. Fitzpatrick’s dermatology in general medicine. McGraw-Hill; p. 169–193.
  2. van de Kerkhof PCM, Nestle FO. (2012) Psoriasis. In: Bolognia JL, Jorizzo JL, editors. Dermatology. Elsevier; p. 135–156.
  3. Tampa M, Sarbu MI, MitranMI, Mitran CI, Matei C, Georgescu SR. (2018) The pathophysiological mechanisms and the quest for biomarkers in psoriasis, a stress-related skin disease. Dis Markers. 2018: 5823684. [Crossref]
  4. Surcel M, Huica R, Constantin C, Ursaciuc C, Neagu M. (2017) Biomarkers insights in psoriasis-regulatory cytokines. Current Biomarkers. 7(1): 3–11.
  5. Benson JM, Sachs CW, Treacy G, Zhou H, Pendley CE, Brodmerkel CM, et al. (2011) Therapeutic targeting of the IL-12/23 pathways: generation and characterization of ustekinumab. Nat Biotechnol. 29: 615–624.
  6. Fernández-Armenteros JM, Gómez-Arbonés X, Buti-Soler, Betriu-Bars A, Sanmartin-Novell V, Ortega-Bravo M, et al. (2019) Psoriasis, metabolic syndrome and cardiovascular risk factors. A population-based study. J Eur Acad Dermatol Venereol.33: 128–135. [Crossref]
  7. Kimball AB, Guerin A, Latremouille-Viau D, Yu AP, Gupta S, Bao Y, et al. (2010) Coronary heart disease and stroke risk in patients with psoriasis: retrospective analysis. Am J Med. 123: 350–357. [Crossref]
  8. Hu SC, Yuan SF, Chen GS, Tu HP. (2019) Increased incidence of ischaemic heart disease and cerebrovascular disease in psoriasis patients with depression: a nationwide retrospective cohort study. J Eur Acad Dermatol Venereol. 33(8): 1547–1554. [Crossref]
  9. Babino G, Fulgione E, Giorgio CM, Agozzino M, Alfano R, Argenziano G. (2019) Efficacy and safety of secukinumab in a psoriatic patient affected by comorbid metabolic disorders. Dermatol Ther. 32(3): e12858. [Crossref]
  10. Svecova D, Lubell MW, Casset-Semanaz F, Mackenzie H, Grenningloh R, Krueger JG. (2019) A randomized, double-blind, placebo-controlled phase 1 study of multiple ascending doses of subcutaneous M1095, an anti-interleukin 17A/F nanobody, in moderate-to-severe psoriasis. J Am Acad Dermatol. 81(1): 196–203. [Crossref]
  11. Boehncke S, Thaci D, Beschmann H, Ludwig RJ, Ackermann H, Badenhoop K, et al. (2007) Psoriasis patients show signs of insulin resistance. Br J Dermatol. 157(6): 1249–51.
  12. Hu SC, Lan CE. (2017) Psoriasis and cardiovascular comorbidities: focusing on severe vascular events, cardiovascular risk factors and implications for treatment. Int J Mol Sci. 18(10). [Crossref]
  13. Tablazon IL, Al-Dabagh A, Davis SA, Feldman SR. (2013) Risk of cardiovascular disorders in psoriasis patients: current and future. Am J Clin Dermatol. 14(1): 1–7. [Crossref]
  14. Alexandroff AB, Pauriah M, Camp RD, Lang CC, Struthers AD, Armstrong DJ. (2009) More than skin deep: atherosclerosis as a systemic manifestation of psoriasis. Br J Dermatol. 161(1): 1–7. [Crossref]
  15. Schäkel K, Schön MP, Ghoreschi K. (2016) Pahogenesis of psoriasis. Hautarzt. 67(6): 422–31. [Crossref]
  16. Boehncke WH. (2018) Systemic Inflammation and cardiovascular comorbidity in psoriasis patients: causes and consequences. Front Immunol. 9: 579. [Crossref]
  17. Bavoso NC, Pinto JM, Soares MMS, Diniz MDS, Teixeira Júnior AL. (2019) Psoriasis in obesity: comparison of serum levels of leptin and adiponectin in obese subjects – cases and controls. An Bras Dermatol. 94(2): 192–197. [Crossref]
  18. Gil-Guillén V, Orozco-Beltrán D, Maiques-Galán A, Aznar-Vicente J, Navarro J, Cea-Calvo L, et al. (2007) Agreement between REGICOR and SCORE scales in identifying high cardiovascular risk in the Spanish population. Rev Esp Cardiol. 60(10): 1042–50.
  19. Khera AV, Demler OV, Adelman SJ, Collins HL, Glynn RJ, Ridker PM, et al. (2017) Cholesterol efflux capacity, high-density lipoprotein particle number, and incident cardiovascular events: an analysis from the JUPITER trial (justification for the use of statins in prevention: an intervention trial evaluating rosuvastatin). Circulation. 135(25): 2494–2504. [Crossref]
  20. Ruiz-Villaverde G, López de la Torre-Casares M, Sánchez-Cano D, Villaverde-Gutiérrez C. (2006) Comparison of the Score and Framingham-DORICA models to estimate the cardiovascular risk in a cohort of hypertensive patients from southern Spain. Med Clin (Barc). 127(8): 314–5.
  21. Topel ML, Shen J, Morris AA, Al Mheid I, Sher S, Dunbar SB, et al. (2018) Comparisons of the Framingham and pooled cohort equation risk scores for detecting subclinical vascular disease in blacks versus whites. Am J Cardiol. 121(5): 564–569. [Crossref]
  22. Puig L. (2012) Cardiovascular risk and psoriasis: the role of biologic therapy. Actas Dermosifiliogr. 103(10): 853–62. [Crossref]
  23. QRISK®3–2018, risk calculator, already have a diagnosis of coronary heart disease (including angina or heart attack). https://qrisk.org/three/, 2018 (accessed 24 June 2019).
  24. Hippisley-Cox J, Coupland C, Vinogradova Y, Robson J, Brindle P. (2008) Performance of the QRISK cardiovascular risk prediction algorithm in an independent UK sample of patients from general practice: a validation study. Heart. 94(1): 34–9. [Crossref]
  25. Daudén E, Castañeda S, Suárez C, García-Campayo J, Blasco AJ, Aguilar MD. (2012) Integrated approach to comorbidity in patients with psoriasis. Actas Dermosifiliogr. 103 Suppl 1: 1–64.
  26. Cea-Calvo L, Vanaclocha F, Belinchón I, Rincón Ó, Juliá B, Puig L. (2016) Underdiagnosis of cardiovascular risk factors in outpatients with psoriasis followed at hospital dermatology offices: The PSO-RISK study. Acta Derm Venereol. 96(7): 972–973.
  27. Willard-Grace R, Chen EH, Hessler D, DeVore D, Prado C, Bodenheimer T. (2015) Health coaching by medical assistants to improve control of diabetes, hypertension, and hyperlipidemia in low-income patients: a randomized controlled trial. Ann Fam Med. 13(2): 130–8. [Crossref]
  28. Moore KJ, Sheedy FJ, Fisher EA. (2013) Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. 13(10): 709–21. [Crossref]
  29. Shin DW, Cho J, Yang HK, Park JH, Lee H, Kim H, et al. (2014) Impact of continuity of care on mortality and health care costs: a nationwide cohort study in Korea. Ann Fam Med. 12(6): 534–41. [Crossref]
  30. Rafieian-Kopaei M, Setorki M, Doudi M, Baradaran A, Nasri H. (2014) Atherosclerosis: process, indicators, risk factors and new hopes. Int J Prev Med. 5(8): 927–46. [Crossref]
  31. Balak DM, Hajdarbegovic E. (2017) Drug-induced psoriasis: clinical perspectives. Psoriasis (Auckl). 7: 87–94. [Crossref]
  32. Milaniuk S, Pietrzak A, Mosiewicz B, Mosiewicz J, Reich K. (2015) Influence of psoriasis on circulatory system function assessed in echocardiography. Arch Dermatol Res. 307(10): 855–61. [Crossref]
  33. Dattilo G, Imbalzano E, Casale M, Guarneri C, Borgia F, Mondello S, et al. (2018) Psoriasis and cardiovascular risk: correlation between psoriasis and cardiovascular functional indices. Angiology. 69(1): 31–37. [Crossref]
  34. Sutton-Tyrrell K, Najjar SS, Boudreau RM, Venkitachalam L, Kupelian V, Simonsick EM, et al. (2005) Elevated aortic pulse wave velocity, a marker of arterial stiffness, predicts cardio-vascular events in well-functioning older adults. Circulation. 111(25): 3384–90. [Crossref]
  35. Ardic I, Kaya MG, Yarlioglues M, Karadag Z, Dogan A, Yildiz H, et al. (2010) Impaired aortic elastic properties in normotensive patients with psoriasis. Blood Press. 19(6): 351–8.
  36. Gorga E, Scodro M, Valentini F, D’Ortona R, Arisi M, Sciatti E, et al. (2018) Echocardiographic evaluation of diastolic dysfunction in young and healthy patients with psoriasis: A case-control study. Monaldi Arch Chest Dis. 88(3): 934.
  37. Bulbul Sen B, Atci N, Rifaioglu EN, Ekiz O, Kartal I, Buyukkaya E, et al. (2013) Increased epicardial fat tissue is a marker of subclinical atherosclerosis in patients with psoriasis. Br J Dermatol. 169(5): 1081–6. [Crossref]
  38. Gisondi P, Ferrazzi A, Girolomoni G. (2010) Metabolic comorbidities and psoriasis. Acta Dermatovenerol Croat. 18(4): 297–304. [Crossref]
  39. Tang K, Lin J, Ji X, Lin T, Sun D, Zheng X, et al. (2019) Non-alcoholic fatty liver disease with reduced myocardial FDG uptake is associated with coronary atherosclerosis. J Nucl Cardiol. 2019.
  40. Schiffrin EL. (2002) Beyond blood pressure: the endothelium and atherosclerosis progression. Am J Hypertens. 15(10 Pt 2): 115S-122S. [Crossref]
  41. El-Mongy S, Fathy H, Abdelaziz A, Omran E, George S, Neseem N, et al. (2010) Subclinical atherosclerosis in patients with chronic psoriasis: a potential association. J Eur Acad Dermatol Venereol. 24(6): 661–6. [Crossref]
  42. Biyik I, Narin A, Bozok MA, Ergene O. (2006) Echocardiographic and clinical abnormalities in patients with psoriasis. J Int Med Res. 34(6): 632–9. [Crossref]
  43. Goyal A, Dey AK, Chaturvedi A, Elnabawi YA, Aberra TM, Chung JH et al. (2018) Chronic stress-related neural activity associates with subclinical cardiovascular disease in psoriasis: a prospective cohort study. JACC Cardiovasc Imaging. pii: S1936-878X(18)30920-3. [Crossref]
  44. Tawakol A, Ishai A, Takx RA, Figueroa AL, Ali A, Kaiser Y et al. (2017) Relation between resting amygdalar activity and cardiovascular events: a longitudinal and cohort study. Lancet. 389(10071): 834–845. [Crossref]
  45. Shiba M, Kato T, Izumi T, Miyamoto S, Nakane E, Haruna T, et al. (2019) Risk of myocardial infarction in patients with psoriasis: a cross-sectional patient-population study in a Japanese hospital. J Cardiol. 73(4): 276–279. [Crossref]
  46. Polic MV, Miskulin M, Smolic M, Kralik K, Miskulin I, Berkovic MC, et al. (2018) Psoriasis severity – A risk factor of insulin resistance independent of metabolic syndrome. Int J Environ Res Public Health. 15(7). [Crossref]
  47. Prodanovich S, Kirsner RS, Kravetz JD, Ma F, Martinez L, Federman DG. (2009) Association of psoriasis with coronary artery, cerebrovascular, and peripheral vascular diseases and mortality. Arch Dermatol. 145(6): 700–3.
  48. Eder L, Chandran V, Gladman DD. (2014) The Framingham Risk Score underestimates the extent of subclinical atherosclerosis in patients with psoriatic disease. Ann Rheum Dis. 73(11): 1990–6. [Crossref]
  49. Naik HB, Natarajan B, Stansky E, Ahlman MA, Teague H, Salahuddin T, et al. (2015) The severity of psoriasis associates with aortic vascular inflammation detected by FDG PET/CT and neutrophil activation in a prospective observational study. Arterioscler Thromb Vasc Biol. 35(12): 2667–76. [Crossref]
  50. Prey S, Paul C, Bronsard V, Puzenat E, Gourraud PA, Aractingi S, et al. (2010) Cardiovascular risk factors in patients with plaque psoriasis: a systematic review of epidemiological studies. J Eur Acad Dermatol Venereol. 24 Suppl 2: 23–30. [Crossref]
  51. Qureshi AA, Choi HK, Setty AR, Curhan GC. (2009) Psoriasis and the risk of diabetes and hypertension: a prospective study of US female nurses. Arch Dermatol. 145(4): 379–82. [Crossref]
  52. Brauchli YB, Jick SS, Meier CR. (2008) Psoriasis and the risk of incident diabetes mellitus: a population-based study. Br J Dermatol. 159(6): 1331–7. [Crossref]
  53. Herron MD, Hinckley M, Hoffman MS, Papenfuss J, Hansen CB, Callis KP, et al. (2005) Impact of obesity and smoking on psoriasis presentation and management. Arch Dermatol. 141(12): 1527–34. [Crossref]
  54. Uyar B, Akyildiz M, Solak A, Genc B, Saklamaz A, et al. (2015) Relationship between serum Fetuin-A levels and carotid intima-media thickness in Turkish patients with mild to moderate psoriasis. A case-control study. Acta Dermatovenerol Croat. 23(3): 171–7. [Crossref]
  55. Rosa DJ, Machado RF, Matias FA, Cedrim SD, Noronha FL, Gaburri D, et al. (2012) Influence of severity of the cutaneous manifestations and age on the prevalence of several cardiovascular risk factors in patients with psoriasis. J Eur Acad Dermatol Venereol. 26(3): 348–53.
  56. Gisondi P, Farina S, Giordano MV, Girolomoni G. (2010) Usefulness of the Framingham risk store in patients with chronic psoriasis. Am J Cardiol. 106(12): 1754–7. [Crossref]
  57. Sorokin AV, Kotani K, Elnabawi YA, Dey AK, Sajja AP, Yamada S, et al. (2018) Association between oxidation-modified lipoproteins and coronary plaque in psoriasis. Circ Res. 123(11): 1244–1254. [Crossref]
  58. Myśliwiec H, Baran A, Harasim-Symbor E, Choromańska B, Myśliwiec P, Milewska AJ, et al. (2017) Increase in circulating sphingosine-1-phosphate and decrease in ceramide levels in psoriatic patients. Arch Dermatol Res. 309(2): 79–86. [Crossref]
  59. Blegvad C, Nybo Andersen AM, Adam A, Zachariae C, Skov L. (2019) Psoriasis as a predictor of cardiometabolic comorbidity in women: a study based on the Danish national birth cohort Acta Derm Venereol. 99(3): 274–278. [Crossref]
  60. Pietrzak A, Czuczwar P, Mosiewicz J, Paszkowski T, Chodorowska G, Bartosinska J, et al. (2015) Cardiovascular disease in psoriatic post-menopausal women. J Eur Acad Dermatol Venereol. 29(6): 1231–4.
  61. Li WQ, Cho E, Weinstock MA, Mashfiq H, Qureshi AA. (2016) Epidemiological assessments of skin outcomes in the Nurses’ Health Studies. Am J Public Health. 106(9): 1677–83. [Crossref]
  62. Li W, Han J, Hu FB, Curhan GC, Qureshi AA. (2012) Psoriasis and risk of type 2 diabetes among women and men in the United States: a population-based cohort study. J Invest Dermatol. 132(2): 291–8. [Crossref]
  63. Li WQ, Han JL, Manson JE, Rimm EB, Rexrode KM, Curhan GC, et al. (2012) Psoriasis and risk of nonfatal cardiovascular disease in U.S. women: a cohort study. Br J Dermatol. 166(4): 811–8. [Crossref]
  64. Kundakçi N, Oskay T, Olmez U, Tutkak H, Gürgey E. (2002) Association of psoriasis vulgaris with HLA class I and class II antigens in the Turkish population, according to the age at onset. Int J Dermatol. 41(6): 345–8. [Crossref]
  65. Wongpiyabovorn J, Hirankarn N, Ruchusatsawat K, Yooyongsatit S, Asawanonda P, Poovorawan Y. (2008) Association of the interleukin-10 distal promoter (-2763A/C) polymorphism with late-onset psoriasis. Clin Exp Dermatol. 33(2): 186–9.
  66. Dinić M, Zečević RD, Hajduković Z, Mijušković M, Ðurić P, Jović Z. (2016) Psoriasis is the independent factor for early atherosclerosis: A prospective study of cardiometabolic risk profile. Vojnosanit Pregl. 73(12): 1094–1101. [Crossref]
  67. Golden JB, Groft SG, Squeri MV, Debanne SM, Ward NL, McCormick TS, et al. (2015) Chronic psoriatic skin inflammation leads to increased monocyte adhesion and aggregation. J Immunol. 195(5): 2006–18.
  68. Ryan C, Kirby B. (2015) Psoriasis is a systemic disease with multiple cardiovascular and metabolic comorbidities. Dermatol Clin. 33(1): 41–55. [Crossref]
  69. Tinelli C, Di Pino A, (2019) Ficulle E, Marcelli S, Feligioni M. Hyperhomocysteinemia as a risk factor and potential nutraceutical target for certain pathologies. Front Nutr. 6: 49. [Crossref]
  70. Wang WM, Jin HZ. (2017) Homocysteine: A potential common route for cardiovascular risk and DNA methylation in psoriasis. Chin Med J (Engl). 130(16): 1980–1986. [Crossref]
  71. Qi JH, Qi JH, Shi N, Chen YJ, Nie G. (2015) Association between MTHFR 677C/T polymorphism and psoriasis risk: A metaanalysis. Genet Mol Res. 14(2): 3869–76. [Crossref]
  72. Kaur S, Kingo K, Zilmer M. (2017) Psoriasis and cardiovascular risk-do promising new biomarkers have clinical impact? Mediators Inflamm. 2017: 7279818. [Crossref]
  73. Johnson TV, Abbasi A, Master VA. (2013) Systematic review of the evidence of a relationship between chronic psychosocial stress and C-reactive protein. Mol Diagn Ther. 17(3): 147–64. [Crossref]
  74. Paschoal RS, Silva DA, Cardili RN, Souza CDS. (2018) Metabolic syndrome, C-reactive protein and cardiovascular risk in psoriasis patients: a cross-sectional study. An Bras Dermatol. 93(2): 222–228. [Crossref]
  75. Uaratanawong R, Uaratanawong S, Chunhasewee C, Chawvavanich P. (2016) High sensitivity C-Reactive protein level and psoriasis severity in Thai patients. J Med Assoc Thai. 99(9): 1039–45 [Crossref]
  76. Yiu KH, Yeung CK, Zhao CT, Chan JC, Siu CW, Tam S, et al. (2013) Prevalence and extent of subclinical atherosclerosis in patients with psoriasis. J Intern Med. 273(3): 273–82.
  77. Ix JH1, Barrett-Connor E, Wassel CL, Cummins K, Bergstrom J, Daniels LB, et al. (2011) The associations of fetuin-A with subclinical cardiovascular disease in community-dwelling persons: The Rancho Bernardo Study. J Am Coll Cardiol. 58(23): 2372–9. [Crossref]
  78. Yazıcı D, Yavuz D, Öğünç AV, Sirikçi Ö, Toprak A, Deyneli O, et al. (2012) Serum adipokine levels in type 1 diabetic patients: association with carotid intima media thickness. Metab Syndr Relat Disord. 10(1): 26–31.
  79. Ataseven A, Kesli R. (2016) Novel inflammatory markers in psoriasis vulgaris: vaspin, vascular adhesion protein-1 (VAP-1), and YKL-40. G Ital Dermatol Venereol. 151(3): 244–50. [Crossref]
  80. Nakajima H, Nakajima K, Tarutani M, Sano S. (2013) Clear association between serum levels of adipokines and T-helper 17-related cytokines in patients with psoriasis. Clin Exp Dermatol. 38(1): 66–70. [Crossref]
  81. Albanesi C, Scarponi C, Pallotta S, Daniele R, Bosisio D, Madonna S, et al. (2009) Chemerin expression marks early psoriatic skin lesions and correlates with plasmacytoid dendritic cell recruitment. J Exp Med. 16;206(1): 249–58. [Crossref]
  82. Furue M, Tsuji G, Chiba T, Kadono T. (2017) Cardiovascular and Metabolic diseases comorbid with psoriasis: beyond the skin. Intern Med. 56(13): 1613–1619. [Crossref]
  83. Vachatova S, Andrys C, Krejsek J, Salavec M, Ettler K, Rehacek V, et al. (2016) Metabolic syndrome and selective inflammatory markers in psoriatic patients. J Immunol Res. 2016: 5380792. [Crossref]
  84. Kiluk P, Baran A, Świderska M, Maciaszek M, Flisiak I. (2019) Lp- PLA2 as a promising predictor of comorbidities in patients with severe psoriasis. J Dermatolog Treat. 1–7. [Crossref]
  85. Wang Q, McCormick TS, Ward NL, Cooper KD, Conic R, Xu R. (2018) Combining mechanism-based prediction with patient-based profiling for psoriasis metabolomics biomarker discovery. AMIA Annu Symp Proc. 2017: 1734–1743. [Crossref]
  86. Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha A, et al. (2010) Interleukin (IL)-22, IL-17, IL-23, IL-8, vascular endothelial growth factor and tumour necrosis factor-α levels in patients with psoriasis before, during and after psoralen-ultraviolet A and narrowband ultraviolet B therapy. Br J Dermatol. 163(6): 1282–90.
  87. Ellulu MS, Patimah I, Khaza’ai H, Rahmat A, Abed Y. (2017) Obesity and inflammation: the linking mechanism and the complications. Arch Med Sci. 13(4): 851–863. [Crossref]
  88. Rodríguez-Cerdeira C, Cordeiro-Rodríguez M, Carnero-Gregorio M, López-Barcenas A, Martínez-Herrera E, Fabbrocini G, et al. (2019) Biomarkers of inflammation in obesity-psoriatic patients. Mediators Inflamm. 2019: 7353420. [Crossref]
  89. Asha K, Singal A, Sharma SB, Arora VK, Aggarwal A. (2017) Dyslipidaemia & oxidative stress in patients of psoriasis: emerging cardiovascular risk factors. Indian J Med Res. 146(6): 708–713. [Crossref]
  90. Haberka M, Bańska-Kisiel K, Bergler-Czop B, Biedroń M, Brzezińska-Wcisło L, Okopień B, et al. (2018) Mild to moderate psoriasis is associated with oxidative stress, subclinical atherosclerosis, and endothelial dysfunction. Pol Arch Intern Med. 128(7–8): 434–439. [Crossref]
  91. Tupikowska-Marzec M, Kolačkov K, Zdrojowy-Wełna A, Słoka NK, Szepietowski JC, Maj J. (2019) The Influence of FTO polymorphism rs9939609 on obesity, some clinical features, and disturbance of carbohydrate metabolism in patients with psoriasis. Biomed Res Int. 2019: 7304345. [Crossref]
  92. Barrea L, Savanelli MC, Di Somma C, Napolitano M, Megna M, Colao A, et al. (2017) Vitamin D and its role in psoriasis: An overview of the dermatologist and nutritionist. Rev Endocr Metab Disord. 18(2): 195–205. [Crossref]
  93. Herrmann M, Farrell CL, Pusceddu I, Fabregat-Cabello N, Cavalier E. (2017) Assessment of vitamin D status – a changing landscape. Clin Chem Lab Med. 55(1): 3–26. [Crossref]
  94. Hruska P, Kuruczova D, Vasku V, Bienertova-Vasku J. (2019) MiR-21 binding site SNP within ITGAM associated with psoriasis susceptibility in women. PLoS One. 14(6): e0218323. [Crossref]
  95. Shi D, Zhong Z, Xu R, Li B, Li J, Habib U, et al. (2019) Association of ITGAX and ITGAM gene polymorphisms with susceptibility to IgA nephropathy. J Hum Genet. 64(9): 927–935.
  96. Törn C, Liu X, Hagopian W, Lernmark Å, Simell O, Rewers M, et al. (2016) Complement gene variants in relation to autoantibodies to beta cell specific antigens and type 1 diabetes in the TEDDY Study. Sci Rep. 6: 27887. [Crossref]
  97. Gulliver WP, Randell S, Gulliver S, Connors S, Bachelez H, MacDonald D, et al. (2016) Do biologics protect patients with psoriasis from myocardial infarction? A retrospective cohort. J Cutan Med Surg. 20(6): 536–541. [Crossref]
  98. Caiazzo G, Fabbrocini G, Di Caprio R, Raimondo A, Scala E, Balato N, et al. (2018) Psoriasis, cardiovascular events, and biologics: lights and shadows. Front Immunol. 9: 1668. [Crossref]
  99. Krueger GG, Langley RG, Leonardi C, Yeilding N, Guzzo C, Wang Y, et al. (2007) A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med. 356(6): 580–92. [Crossref]
  100. Popko K, Gorska E, Stelmaszczyk-Emmel A, Plywaczewski R, Stoklosa A, Gorecka D, et al. (2010) Proinflammatory cytokines Il-6 and TNF-α and the development of inflammation in obese subjects. Eur J Med Res. 15 Suppl 2: 120–2. [Crossref]
  101. Kaplanski G. (2018) Interleukin-18: Biological properties and role in disease pathogenesis. Immunol Rev. 281(1): 138–153. [Crossref]
  102. Alipourfard I, Datukishvili N, Mikeladze D. (2019) TNF-α downregulation modifies insulin receptor substrate 1 (IRS-1) in metabolic signaling of diabetic insulin-resistant hepatocytes. Mediators Inflamm. 2019: 3560819. [Crossref]
  103. Zhang C. (2008) The role of inflammatory cytokines in endothelial dysfunction. Basic Res Cardiol. 103(5): 398–406. [Crossref]
  104. Erbel C, Akhavanpoor M, Okuyucu D, Wangler S, Dietz A, Zhao L, et al. (2014) IL-17A influences essential functions of the monocyte/macrophage lineage and is involved in advanced murine and human atherosclerosis. J Immunol. 193(9): 4344–55. [Crossref]
  105. Cimmino G, Ciuffreda LP, Ciccarelli G, Calabrò P, Ferraiolo FA, Rivellino A, et al. (2017) Upregulation of TH/IL-17 pathway-related genes in human coronary endothelial cells stimulated with serum of patients with acute coronary syndromes. Front Cardiovasc Med. 4: 1. [Crossref]
  106. Mohammadi M, Gozashti MH, Aghadavood M, Mehdizadeh MR, Hayatbakhsh MM. (2017) Clinical significance of serum IL-6 and TNF-α levels in patients with metabolic syndrome. Rep Biochem Mol Biol. 6(1): 74–79. [Crossref]
  107. Heberto Herrera Garza E, Herrera Garza JL, Rodríguez González H, Treviño Treviño A, Ibarra Flores M, Torre Amione G. (2002) Importance of tumor necrosis factor-alpha in the pathogenesis of heart failure. Rev Esp Cardiol. 55(1): 61–6.
  108. Liu M, Chen J, Huang D, Ke J, Wu W. (2014) A meta-analysis of proinflammatory cytokines in chronic heart failure. Heart Asia. 6(1): 130–6. [Crossref]
  109. Mu W, Cheng XF, Liu Y, Lv QZ, Liu GL, Zhang JG, et al. (2019) Potential nexus of non-alcoholic fatty liver disease and type 2 diabetes mellitus: Insulin resistance between hepatic and peripheral tissues. Front Pharmacol. 9: 1566. [Crossref]
  110. Papp KA, Leonardi C, Menter A, Ortonne JP, Krueger JG, Kricorian G, et al. (2012) Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl J Med. 366(13): 1181–9. [Crossref]
  111. Spuls PI, Hooft L. (2012) Brodalumab and ixekizumab, anti-interleukin-17-receptor antibodies for psoriasis: a critical appraisal. Br J Dermatol. 167(4): 710–3; discussion 714–5. [Crossref]
  112. Lockshin B, Balagula Y, Merola JF. (2018) Interleukin 17, inflammation, and cardiovascular risk in patients with psoriasis. J Am Acad Dermatol. 79(2): 345–352. [Crossref]
  113. Ventura A, Mazzeo M, Gaziano R, Galluzzo M, Bianchi L, Campione E. (2017) New insight into the pathogenesis of nail psoriasis and overview of treatment strategies. Drug Des Devel Ther. 11: 2527–2535. [Crossref]
  114. Shih CM, Huang CY, Wang KH, Huang CY, Wei PL, Chang YJ, et al. (2018) Oxidized low-density lipoprotein-deteriorated psoriasis is associated with the upregulation of Lox-1 receptor and Il-23 expression in vivo and in vitro. Int J Mol Sci. 19(9). [Crossref]
  115. Abbas A, Gregersen I, Holm S, Daissormont I, Bjerkeli V, Krohg-Sørensen K, et al. (2015) Interleukin 23 levels are increased in carotid atherosclerosis: Possible role for the interleukin 23/interleukin 17 axis. Stroke. 46(3): 793–9. [Crossref]
  116. Di Cesare A, Di Meglio P, Nestle FO. (2009) The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 129(6): 1339–50. [Crossref]
  117. Fashanu OE, Oyenuga AO, Zhao D, Tibuakuu M, Mora S, Otvos JD, et al. (2019) GlycA, a novel inflammatory marker and its association with peripheral arterial disease and carotid plaque: the multi-ethnic study of atherosclerosis. Angiology. 70(8): 737–746. [Crossref]
  118. Joshi AA, Lerman JB, Aberra TM, Afshar M, Teague HL, Rodante JA, et al. (2016) GlycA is a novel biomarker of inflammation and subclinical cardiovascular disease in psoriasis. Circ Res. 119(11): 1242–1253. [Crossref]
  119. Montaudié H, Albert-Sabonnadière C, Acquacalda E, Fontas E, Danré A, Roux C, et al. (2014) Impact of systemic treatment of psoriasis on inflammatory parameters and markers of comorbidities and cardiovascular risk: results of a prospective longitudinal observational study. J Eur Acad Dermatol Venereol. 28(9): 1186–91. [Crossref]
  120. Jiang S, Hinchliffe TE, Wu T. (2015) Biomarkers of an autoimmune skin disease–Psoriasis. Genomics Proteomics Bioinformatics. 13(4): 224–33. [Crossref]
  121. Mansouri B, Patel M, Menter A. (2013) Biological therapies for psoriasis. Expert Opin Biol Ther. 13(12): 1715–30. [Crossref]
  122. Kamleh MA, Snowden SG, Grapov D, Blackburn GJ, Watson DG, Xu N, et al. (2015) LC–MS metabolomics of psoriasis patients reveals disease severity-dependent increases in circulating amino acids that are ameliorated by anti-TNFα treatment. J Proteome Res. 14(1): 557–66. [Crossref]
  123. Späh F. (2008) Inflammation in atherosclerosis and psoriasis: common pathogenic mechanisms and the potential for an integrated treatment approach. Br J Dermatol. 159 Suppl 2: 10–7. [Crossref]
  124. Wakkee M, Thio HB, Prens EP, Sijbrands EJ, Neumann HA. (2007) Unfavorable cardiovascular risk profiles in untreated and treated psoriasis patients. Atherosclerosis. 190(1): 1–9. [Crossref]
  125. Ridker PM, Everett BM, Pradhan A, MacFadyen JG, Solomon DH, Zaharris E, et al. (2019) Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 380(8): 752–762. [Crossref]
  126. Rungapiromnan W, Yiu ZZN, Warren RB, Griffiths CEM, Ashcroft DM. (2017) Impact of biologic therapies on risk of major adverse cardiovascular events in patients with psoriasis: systematic review and meta-analysis of randomized controlled trials. Br J Dermatol. 176(4): 890–901. [Crossref]
  127. Lee MP, Desai RJ, Jin Y, Brill G, Ogdie A, Kim SC. (2019) Association of ustekinumab vs TNF inhibitor therapy with risk of atrial fibrillation and cardiovascular events in patients with psoriasis or psoriatic arthritis. JAMA Dermatol. 155(6): 700–707. [Crossref]