Daily Archives: December 10, 2021

ENCODE at UCSC

Posted: December 10, 2021 at 7:27 pm

The Encyclopedia of DNA Elements(ENCODE) Consortium is an international collaboration of research groups funded by the National Human Genome Research Institute (NHGRI).The goal of ENCODE is to build a comprehensive parts list of functional elements in the human genome, including elements that act at the protein and RNA levels, and regulatory elements that control cells and circumstances in which a gene is active.

ENCODE results from 2007 and later are available from the ENCODE Project Portal,encodeproject.org.This covers data generated during the two production phases 2007-2012and 2013-present. The ENCODE Project Portal also hosts additionalENCODE access tools, and ENCODE project pages including up-to-date information about data releases, publications, and upcoming tutorials.

UCSC coordinated data for the ENCODE Consortium from its inception in 2003 (Pilot phase) to the end of the first 5 year phase of whole-genome data production in 2012. All data produced by ENCODE investigators and the results of ENCODE analysis projects from this period are hosted in the UCSC Genome browser and database.Explore ENCODE data using the image links below or via the left menu bar.All ENCODE data at UCSC are freely available for download and analysis.

Go here to see the original:
ENCODE at UCSC

Posted in Genome | Comments Off on ENCODE at UCSC

OCD: Using Genome Data to Predict Risk, Symptoms and Treatment Response – A Free Webinar from the Brain & Behavior Research Foundation – Yahoo…

Posted: at 7:26 pm

Webinar Presenter

Dr. Gwyneth Zai

Dr. Jeffrey Borenstein

New York, Dec. 07, 2021 (GLOBE NEWSWIRE) -- The Brain & Behavior Research Foundation (BBRF) is hosting a free webinar, OCD: Using Genome Data to Predict Risk, Symptoms and Treatment Response on Tuesday, December 14, 2021, from 2pm to 3pm ET. The presenter will be Gwyneth Zai, Assistant Professor at the University of Toronto and recipient of a 2016 Young Investigator Grant.

Dr. Zai will discuss how the human genome holds clues to understanding the heterogeneity and complexity of obsessive-compulsive disorder (OCD). She will explain her team's use of genome data to identify genetic variations that contribute to the risk of developing OCD and which may enable prediction of the response of individual patients to antidepressant medications. Jeffrey Borenstein, M.D., President and CEO of the Brain & Behavior Research Foundation and Host and Executive Producer of the public television series Healthy Minds, will be the moderator. Join by phone or on the web at bbrf.org/decemberwebinar.

This webinar is part of a series of free monthly Meet the Scientist webinars on the latest developments in psychiatry offered by the Brain & Behavior Research Foundation. Please use #BBRFWebinar when sharing or posting about our Meet the Scientist Webinars on social media.

The Brain & Behavior Research Foundation The Brain & Behavior Research Foundation awards research grants to develop improved treatments, cures, and methods of prevention for mental illness. These illnesses include addiction, ADHD, anxiety, autism, bipolar disorder, borderline personality disorder, depression, eating disorders, OCD, PTSD, and schizophrenia, as well as research on suicide prevention. Since 1987, the Foundation has awarded more than $430 million to fund more than 5,100 leading scientists around the world, which has led to over $4 billion in additional funding for these scientists. 100% of every dollar donated for research is invested in research. BBRF operating expenses are covered by separate foundation grants. BBRF is the producer of the Emmy nominated public television series Healthy Minds with Dr. Jeffrey Borenstein, which aims to remove the stigma of mental illness and demonstrate that with help, there is hope.

Story continues

Attachments

Originally posted here:
OCD: Using Genome Data to Predict Risk, Symptoms and Treatment Response - A Free Webinar from the Brain & Behavior Research Foundation - Yahoo...

Posted in Genome | Comments Off on OCD: Using Genome Data to Predict Risk, Symptoms and Treatment Response – A Free Webinar from the Brain & Behavior Research Foundation – Yahoo…

Psoriatic Arthritis vs. Osteoarthritis – Health Essentials from Cleveland Clinic

Posted: at 7:23 pm

You have psoriasis, and now youre experiencing joint pain. Does this mean you have psoriatic arthritis? Or could it be unrelated?

Cleveland Clinic is a non-profit academic medical center. Advertising on our site helps support our mission. We do not endorse non-Cleveland Clinic products or services.Policy

Osteoarthritis is the most common type of arthritis, and, sometimes, it can occur alongside psoriasis. Then again, psoriatic arthritis is also a possibility if you are experiencing skin symptoms, says rheumatologist Rochelle Rosian, MD.

How can you make sense of it all? We talked to Dr. Rosian to find out what these types of arthritis have in common, how they differ and how to treat them.

The word arthritis is kind of a catch-all term, Dr. Rosian explains. Its used to describe more than 100 different conditions that cause pain, swelling and joint damage. Osteoarthritis and psoriatic arthritis are two of those many conditions.

Osteoarthritis, or degenerative joint disease, is the most common type of arthritis. It occurs when the cartilage that cushions your bones wears down, leaving bone to rub against bone.

Since its caused by wear and tear on the joints over time, it usually develops in older adults. Unlike psoriatic arthritis, osteoarthritis doesnt involve inflammation or an overactive immune system.

Common features of osteoarthritis include:

The location of the pain depends on the type of osteoarthritis:

Psoriatic arthritis is a type of inflammatory arthritis. It occurs when your bodys immune system works overtime, creating inflammation throughout your body. In psoriatic arthritis, that inflammation targets the joints and the places where tendons and ligaments attach to bones.

Most people with psoriatic arthritis also have psoriasis, a disease that causes red, scaly patches of skin. Psoriatic arthritis can strike at any age, though symptoms usually develop between ages 30 and 50.

Psoriatic arthritis causes symptoms in your joints and beyond, including:

Joint symptoms:

Skin symptoms:

Other symptoms:

In osteoarthritis, X-rays may show signs like worn cartilage. In psoriatic arthritis, X-rays can show joint damage in later stages of the disease.

But they arent especially helpful in making a diagnosis in the early stages. X-rays can help make an arthritis diagnosis. But they arent always a slam dunk, Dr. Rosian says.

X-rays can also reveal bone spurs, which can develop in people with osteoarthritis and psoriatic arthritis. Those images may reveal differences between the two diseases:

To diagnose joint disease, your healthcare provider will probably consider several factors in addition to X-rays, including:

Getting an accurate diagnosis is important since psoriatic arthritis and osteoarthritis require different treatments.

Medications are often an essential part of treating arthritis. But the medications can differ depending on the type.

Treatments for osteoarthritis include:

Treatments for psoriatic arthritis include:

In addition to medication, you can take other steps to manage arthritis symptoms. These approaches can help whether you have psoriatic arthritis or osteoarthritis:

By making certain lifestyle changes and working with your doctor to develop a treatment plan, you can keep arthritis symptoms in check, says Dr. Rosian.

Originally posted here:
Psoriatic Arthritis vs. Osteoarthritis - Health Essentials from Cleveland Clinic

Posted in Psoriasis | Comments Off on Psoriatic Arthritis vs. Osteoarthritis – Health Essentials from Cleveland Clinic

Psoriasis Volunteering: Benefits and Pitfalls – Everyday Health

Posted: at 7:23 pm

The announcement of the 2021 Outstanding Volunteer Leader of the Year Award came at the end of the National Psoriasis Foundations virtual October Community Conference. I felt honored to be one of the nine finalists and thought that any of the other nominees would be deserving of this honor.

To my great surprise,I heard my name announced as the winner and saw my picture flash on the screen. My wife, Lori, gave me a hug, while those at the conference messaged me their congratulations.

Volunteering is a big part of who I am. Over the years, Ive given my time and energy to local food banks, churches, schools, and youth sports teams, and as a church minister I recruited and supported volunteers. Ive volunteered with the National Psoriasis Foundation (NPF) for years, hoping to improve the lives of those impacted by psoriatic disease.

A fellow NPF volunteer at the conference rightly noted that we dont do what we do for the recognition; we do it to help others. Still, Ive personally experienced many benefits from volunteering that I hope encourage you to volunteer as well.

Here are five ways volunteering has made my life better plus one pitfall to watch out for.

One great benefit of volunteering is meeting people who care about the same causes you do, with similar values. My first advocacy event with the NPF introduced me to a community of people from diverse backgrounds, all united by a desire to help the psoriatic disease community.

One experience that stands out was in April 2015, when I drove 30 minutes to the California State Capitol in Sacramento to join fellow NPF volunteers for the first annual California Lobby Day. Our mission was to talk to state legislators about bills related to access to medical care. The connections and friendships I made at the Lobby Day are ones I still greatly appreciate today.

I become particularly self-focused when my psoriasis is not doing well. Self-care is important during those times, but I also find myself losing perspective and imagining worst-case scenarios.

For me, volunteering can help remedy this by redirecting my focus outward. For instance, organizing the first Sacramento Team NPF Walk came at a tough time for me. I felt challenged by my work on many fronts and my psoriasis was flaring, which was causing me to stress about potentially changing biologics.

Putting my attention on the event led me to see my difficulties in a new light. I found joy in planning the Walk path, talking to dermatology offices, and inviting participants to join the fundraiser. All this gave me more energy to address my own personal concerns.

RELATED: How to Beat the Psoriasis-Stress Cycle

Finding ways to make a difference through volunteering can give your life meaning and purpose.

Im motivated by making a difference in the lives of others. Whether its serving a Thanksgiving meal to those who experience food insecurity or teaching a youth baseball player how to field a grounder, I want to uplift others. In the process, I find my own life is enriched.

Ive dedicated my NPF volunteer work to areas where I feel I can have the greatest impact, such as using education to reduce the stigma that often accompanies psoriasis. Through fundraising events, I also hope to assist in bringing about better treatments and a cure, and I strive to support fellow patients in advocating for themselves and others.

Volunteering can provide opportunities to develop or utilize your skill set. Ive grown my teaching skills as a youth coach, and my social media engagement has increased as an NPF Social Ambassador. Ive taught and trained volunteers on how to lead a meeting and mentor college students at church.

Im a reserved and introverted person, but volunteering has also taught me a lot about boldness and assertiveness. At work Ive cultivated skills in public speaking and leading teams, but asking legislators to support a bill that would help those in the chronic illness community felt different and even scary.

In some of those legislative meetings, I was the one who needed to give the ask. Over time it became more natural for me to assert myself in those situations and, consequently, in other areas of my life.

Its easy for me to get overwhelmed by the size and needs of the psoriasis community. When I think about the more than 8 million people in the United States and 125 million worldwide living with psoriatic disease, I feel small. Instead of doing something proactive, I sometimes find myself complaining.

Volunteering provides a positive direction to channel that unproductive energy. Instead of bemoaning the problem, I can be part of the solution. Joining a patient advocacy organization like the NPF brings me together with others who are rowing in the same direction. Together, we can bring about a cure for psoriasis and improve lives.

RELATED: You Yes, You! Can Become a Psoriasis Patient Advocate

While I love to volunteer, I do find myself overcommitted from time to time. I want to do it all, but when my health warrants more attention or work is especially busy, Ive learned when to pull back or say no.

Still, there are so many ways to contribute to the betterment of others through volunteering, and what compels me may be different than what excites you. If you have time and energy to offer, consider how you can get involved and experience the joy of giving to others.

Learn more about volunteering with the National Psoriasis Foundation.

See the original post here:
Psoriasis Volunteering: Benefits and Pitfalls - Everyday Health

Posted in Psoriasis | Comments Off on Psoriasis Volunteering: Benefits and Pitfalls – Everyday Health

Non-alcoholic fatty liver disease in patients with psoriasis | PTT – Dove Medical Press

Posted: at 7:23 pm

Introduction

Psoriasis is a chronic, immune-mediated inflammatory dermatological disease that affects nearly 150 million people worldwide. It is associated with a significant disease burden, a marked negative impact on patients quality of life,1 and places enormous pressure on health systems globally.24 In recent years attention has focused on the inflammatory processes underpinning the pathophysiological changes in psoriasis. In particular, cutaneous inflammation in psoriasis is linked to chronic systemic low-grade inflammation, which is thought to underlie the associations of psoriasis with comorbidities such as psoriatic arthritis, mood disorders including depression, and a range of cardiometabolic disorders that include myocardial infarction, hypertension, obesity, type 2 diabetes, non-alcoholic fatty liver disease (NAFLD), dyslipidemia, and hyperuricemia.3,5

One comorbidity of psoriasis that is increasingly recognized as clinically relevant is NAFLD.3,6,7 NAFLD is characterized by excessive hepatic fat storage and is strongly associated with obesity, type 2 diabetes, and metabolic syndrome. Over time, NAFLD can progress to a more severe, inflammatory disease termed non-alcoholic steatohepatitis (NASH) and eventually to the development of cirrhosis.8 NAFLD is the most common liver disease and its prevalence is rising rapidly. US studies have reported a 1035% prevalence in the general population with an average of between 20 and 30%.9 The global prevalence of NAFLD is estimated to be 25%.10,11 The presence of NASH in the general population is difficult to ascertain since it can be confirmed only by liver biopsy. However, rates between 1.5 and 6.5% have been reported.9,10,12 NASH is a potentially progressive disease that can lead to cirrhosis in 1225% of cases within a 10-year period, and it is associated with increased risks for hepatocellular carcinoma and cardiovascular- and liver-related deaths.8,13,14

NAFLD has been shown to occur 1.5 to 3 times more frequently in patients with psoriasis than in the general population.3,11,15 Prevalences of NAFLD ranging up to 65% have been reported in this patient population.16 The increased risk of NAFLD in psoriasis patients is not surprising, as the two diseases share a common link with the metabolic syndrome. However, there may be an association between psoriasis and NAFLD that is independent from the metabolic syndrome, hinging directly on the chronic low-grade systemic inflammatory burden characteristic of these two conditions.5 Importantly, when psoriasis co-occurs with NAFLD the disease severity and morbidity of both conditions may be greater.17

The aim of this review is to evaluate the recent literature on the clinical and pathophysiological associations linking psoriasis with NAFLD, independently of the metabolic syndrome, focusing on the chronic low-grade inflammation underpinning the two disorders. Second, we aimed to assess the potential hepatotoxic and hepatoprotective effects of currently approved systemic psoriasis therapies, in particular dimethyl fumarate (DMF), an oral small molecule with pleiotropic effects that could positively impact NAFLD, focusing on the chronic low-grade inflammation underpinning the two disorders. Second, we aimed to assess the potential hepatotoxic and hepatoprotective effects of currently approved systemic psoriasis therapies, in particular dimethyl fumarate (DMF), an oral small molecule with pleiotropic effects that could positively impact NAFLD.

For this narrative review, a targeted search of PubMed up to 14 July 2021 was conducted using the following search strategies: psoriasis with (a) non-alcoholic fatty liver disease/NAFLD (n = 96), (b) hepatotoxicity 1/1/2010 to 14/7/2021 (n = 72), and (c) hepatoprotection (n = 14); psoriasis treatment with (a) hepatotoxicity 1/1/2010 to 14/7/2021 (n = 68), and (b) hepatoprotection (n = 11). Relevant papers were identified by reviewing abstracts or full papers (if required), and by contributory articles cited in the selected references. The bibliography was augmented by key articles known to the authors and, finally, by individual drug data identified via a search of LiverTox (https://www.ncbi.nlm.nih.gov/books/NBK548744/).

NAFLD is the most common chronic liver disease worldwide and, in the majority of patients, is strongly associated with metabolic risk factors such as obesity, diabetes mellitus, and dyslipidemia. Individuals with metabolic syndrome have a 4- to 11-fold increased risk of developing NAFLD.18 NAFLD is characterized by fat deposition in the liver, in the absence of viral diseases such as hepatitis B or C, significant alcohol consumption, use of steatosis-stimulating drugs such as methotrexate, steroids, amiodarone and tamoxifen, or certain hereditary conditions including Wilsons disease and cholesteryl ester storage syndrome.19,20

NAFLD includes a wide spectrum of liver conditions with two main histological forms: simple hepatic steatosis (fat deposited, but no damage to liver cells) and NASH, which is characterized by hepatic inflammation that can lead to liver fibrosis. The aberrant pathophysiological processes underlying the progression to NASH are not fully understood, but are thought to include an imbalance of fatty acid metabolism leading to steatosis, and an elevated inflammatory response as a result of oxidative/metabolic stress and dysregulated cytokine production.21 Potential consequences of these processes are the development of lobular hepatitis with perivenular/pericellular (chicken wire) fibrosis, NAFLD-associated cirrhosis and liver failure and, albeit infrequently, hepatocellular carcinoma (Figure 1).5,22

Figure 1 Spectrum of non-alcoholic fatty liver disease (NAFLD). Data from these studies.5,22

In terms of clinical presentation, the majority of NAFLD cases are either asymptomatic or have nonspecific symptoms such as fatigue and abdominal pain, and/or abnormal liver function test results. NAFLD can be diagnosed if > 5% hepatic steatosis is shown by liver ultrasound in the absence of excessive alcohol use.8 The assessment for potential NASH requires additional testing using non-invasive assessment for liver fibrosis such as vibration-controlled transient elastography or magnetic resonance elastography. However, to confirm the diagnosis of NASH, histopathologic assessment of a liver biopsy is required.

Treatment of NAFLD is limited to optimal management of comorbid conditions such as type 2 diabetes and dyslipidemia, and lifestyle modifications aiming at 710% weight loss through caloric restriction and exercise.8 Weight loss and increased physical activity/exercise can help normalize liver enzyme levels, reduce hepatic inflammation and improve insulin resistance, steatosis and liver histology.23 There are currently no approved pharmacological therapies for NAFLD; oral vitamin E and pioglitazone are options that have shown modest clinical benefits.

The first published evidence in 2001 of a link between psoriasis and NAFLD involved three overweight/obese patients with NASH as confirmed by liver biopsy.24 Since then, various observational and controlled studies have highlighted an increased prevalence of NAFLD in patients with psoriasis.2529 For example, in a large Dutch population-based study (2292 participants aged 55 years) the prevalence of NAFLD was 46% in 118 patients with psoriasis and 33% in individuals without psoriasis.27 Importantly, while elderly study participants with psoriasis were 70% more likely to have NAFLD than those without psoriasis (crude odds ratio [OR] 1.70, 95% confidence interval [CI] 1.172.46), the increased risk was found to be independent of common risk factors such as smoking, alcohol consumption, and the presence of metabolic syndrome and its component disorders. A recent single-center cross-sectional study from Spain reported a 52% prevalence of NAFLD among a cohort of 71 patients with psoriasis.15 Of note, 14% of patients had liver fibrosis as diagnosed with transitional vibration-controlled elastography. A large population-based cohort study, using UK data from 197,130 patients with psoriasis and 1,279,754 matched controls, reported an elevated risk for incident cases of NAFLD among psoriasis patients without systemic therapy (adjusted hazard ratio [HR] 1.18, 95% CI 1.071.30) and an even higher risk for those receiving systemic therapy (adjusted HR 2.23, 95% CI 1.732.87) compared with the control group.30 The risk for incident NAFLD was also increased among patients with psoriatic arthritis, in particular for those receiving systemic therapy (adjusted HR 2.11, 95% CI 1.552.87). In line with these results, a 2015 systematic review and meta-analysis including 7 case-control studies found that patients with psoriasis had a 2-fold increased risk of NAFLD compared with controls (6 studies; n = 267,761; OR 2.15, 95% CI 1.572.94).31 The risk of NAFLD was significantly greater in patients with psoriatic arthritis (3 studies; n = 505 patients; OR 2.25, 95% CI 1.373.71) and in patients with severe psoriasis (2 studies; 51,930 patients, OR 2.07, 95% CI 1.592.71) compared to those with mild psoriasis. Recently, this meta-analysis was updated to include 2 additional studies (for a total of > 3 million patients and nearly 250,000 with NAFLD).32 The analysis was extended to investigate potential risk factors for NAFLD in psoriasis patients which included hypertension, male sex, hyperglycemia and obesity. Reaffirming the findings of the original systematic review, there was a strong association between psoriasis and NAFLD independent of confounders.

In several studies, the presence and severity of psoriasis were associated with a higher prevalence and greater severity of NAFLD, and NAFLD was a strong predictor of higher Psoriasis Area and Severity Index (PASI) scores.25,28,29,31,33 Using Control Attenuation Parameter to assess the degree of fatty liver and body surface area severity of psoriasis, Gandha et al confirmed a positive correlation between the two disorders.34 Furthermore, the progression to more severe forms of liver disease appeared to be higher in patients with psoriasis.35 Roberts et al reported that 48 of 103 (47%) psoriasis patients had NAFLD and 23 of 103 (22%) had biopsy-confirmed NASH, of whom 35% had stage 23 fibrosis.35 The prevalence of NASH was markedly higher than the 12% previously reported in patients with similar demographic characteristics but without psoriasis in the same medical center. Moreover, concomitant NAFLD in patients with psoriasis may confer a higher 10-year cardiovascular risk compared to psoriasis patients without NAFLD.36

In summary, the epidemiologic association between NAFLD and psoriasis is particularly strong given the high prevalence and increased incidence observed among patients with psoriasis. Importantly, this association is more evident in patients with severe psoriasis compared with those with mild psoriasis and is independent of common traditional risk factors such as smoking, alcohol consumption, and the presence of metabolic syndrome and its component disorders.5,31,32

Psoriasis and NAFLD are amongst a number of multifactorial disorders (including cardiovascular diseases and metabolic syndrome) whose pathogenesis is not fully understood, but involves complex interactions between genetic, immunological and environmental factors (Figure 2).5,3742

Figure 2 Common risk factors and associations between psoriasis (PsO) and non-alcoholic fatty liver disease (NAFLD). Reprinted from Prussick RB, Miele L. Non-alcoholic fatty liver disease in patients with psoriasis: a consequence of systemic inflammatory burden? Br J Dermatol. 2018;179(1):1629. 2018 The Authors. British Journal of Dermatology published by John Wiley & Sons Ltd on behalf of British Association of Dermatologists.5

The pathophysiology of psoriasis is characterized by sustained, self-amplifying inflammatory responses that lead to uncontrolled keratinocyte proliferation and dysfunctional differentiation. Disturbances in both innate and adaptive cutaneous immune responses are responsible for psoriatic inflammation.43 Activation of the innate immune system driven by endogenous signals and cytokines coexists with an auto-inflammatory response in some patients and with T cell-driven auto-immune reactions in others. Thus, psoriasis exhibits traits of an auto-immune disease on an auto-inflammatory background,44,45 with both mechanisms overlapping and possibly potentiating one another. T helper (Th)-1, Th17 and Th22 cells and some of their associated pro-inflammatory cytokines, such as interleukin (IL)-17A and IL-22 and tumor necrosis factor alpha (TNF-) are critically involved in sustaining and maintaining psoriasis.4548 The IL-23/IL-17 immune axis is considered to have a pivotal role in the pathogenesis of psoriasis; in line with this, IL-17- and IL-23-antagonists are highly effective treatments for psoriasis.49,50

The pathogenesis of NAFLD is complex and involves multiple pathways. Important pathophysiological mechanisms include genetic factors, insulin resistance and hyperinsulinemia, oxidative stress and hepatocyte lipotoxicity, hepatic inflammation, fibrosis, and gastrointestinal dysbiosis.21,38,5155 The various factors are encompassed in a multiple hit model for NAFLD development and progression.21,56 In the early stages of NAFLD, insulin resistancepossibly related to an increase in pro-inflammatory cytokinesplays a pivotal role by increasing the release of circulating free fatty acids, followed by abnormal accumulation of triglycerides in liver cells and hepatic steatosis. This may be followed by a phase in which simple steatosis transitions into steatohepatitis as a result of an increased inflammatory response. Additional pathways subsequently involved in NAFLD progression include mitochondrial dysfunction and liver apoptosis, increased oxidative stress, activation of the profibrogenic transforming growth factor- (TGF-) pathway, and hepatic stellate cell activation and injury.5

While the exact underlying mechanisms must still be fully clarified, both psoriasis and NAFLD are strongly associated with low-grade, chronic inflammation, peripheral insulin resistance, and increased levels of oxidative stress.40,57 Elevated insulin resistance and increased release of inflammatory cytokines are also characteristic of the metabolic syndrome and obesity. Adipose tissue produces adipocytokines (or adipokines) such as adiponectin, leptin and resistin, as well as pro-inflammatory cytokines which play important roles in the pathogenesis of both psoriasis and NAFLD.

Besides the important role of adipose tissue in mediating the interplay between skin and liver, (severe) psoriasis may have a direct impact on NAFLD, possibly via mechanisms beyond overweight and obesity. Indeed, NAFLD in the general population can also occur among individuals who are not obese and have a normal body mass index. These individuals are labelled as lean NAFLD.58 Interestingly, compared to healthy subjects, individuals with lean NAFLD have higher mean serum C-reactive protein (CRP) levels, suggesting that systemic inflammation might be one of the pathogenic factors.58

The secretion of pro-inflammatory cytokines from psoriatic tissue into the general circulation may reinforce the prevailing systemic pro-inflammatory milieu associated with NAFLD. IL-6, IL-17, TNF- and CRP produced by the liver (hepatokines) and psoriatic skin have reciprocal direct effects on these organs (Figure 3). The pro-inflammatory effects of adipocytokines and hepatokines are summarized in Table 1. Importantly, there may be a bi-directional relationship between psoriasis and NAFLD through pro-inflammatory pathways, postulated as the hepato-dermal axis.48 Circulating pro-inflammatory cytokines such as TNF- and IL-17 derived from psoriatic skin, upon reaching the liver, could impact liver inflammation and insulin resistance. Conversely, pro-inflammatory mediators stemming from hepatic inflammation could contribute to the onset or exacerbation of cutaneous inflammation in psoriasis. Pro-inflammatory immune modulators released by adipose tissue and the liver are involved in the promotion of hepatic fibrogenesis in NAFLD as well as psoriasis pathogenesis.5,40,48,59 TNF-, for example, is involved in psoriasis inflammation and has been shown to be an independent predictor of hepatic fibrogenesis and disease progression.60 Another relevant cytokine in this context is IL-17, which plays a central role in psoriasis pathogenesis. IL-17 is able to induce hepatic stellate cells activation and subsequent collagen production.50,61 By doing so, IL-17 facilitates the progression from simple liver steatosis to steatohepatitis.50,61,62

Table 1 Cytokine Levels and Effects of Adipocytokines and Hepatokines in Psoriasis and Non-Alcoholic Fatty Liver Disease (NAFLD)

Figure 3 Possible mechanisms linking dysfunctional visceral adipose tissue, psoriatic skin and steatosis.

Abbreviations: CRP, C-reactive protein; IL-6, interleukin-6; IL-17, interleukin-17; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; PAI-1, plasminogen activator inhibitor-1; TGF-, transforming growth factor-beta; TNF-, tumor necrosis factor alpha.

Notes: Reproduced from Mantovani A, Gisondi P, Lonardo A, et al. Relationship between non-alcoholic fatty liver disease and psoriasis: a novel hepato-dermal axis? Int J Mol Sci. 2016;17(2):217.48

High circulating (or hepatic) levels of proprotein convertase subtilisin kexin type-9 (PCSK9) have been shown to play a key role in muscle and liver lipid storage, adipose energy storage and hepatic fatty acids and triglycerides storage and secretion. These effects contribute to involvement of the enzyme in the pathogenesis of both NAFLD63 and psoriasis.64

Other shared pathways between psoriasis and NAFLD include modulation of lipid and glucose metabolism, which both play an important role in the development of metabolic syndrome and keratinocyte proliferation in psoriasis.

In summary, there is significant overlap in the pathophysiological factors underlying psoriasis and NAFLD, mostly relating to pathways involving inflammation, oxidative stress, and glucose and lipid metabolism.

Given the strong pathophysiological links between psoriasis and NAFLD and the high prevalence of NAFLD among psoriasis patients, dermatologists should always screen for possible liver disease in their psoriasis patients. A good understanding of the potential hepatic effects of systemic treatments prescribed for patients with psoriasis would also be clinically important. NAFLD-promoting drugs should be avoided, especially in high-risk patients. On the other hand, systemic therapies that reduce systemic inflammation may have a positive influence and mitigate the risk of developing NAFLD. Below, we summarize hepatotoxic and potential hepatoprotective effects of currently approved systemic psoriasis therapies with a focus on NAFLD.

Hepatotoxic risk is associated with the administration of a number of conventional drugs used in the treatment of psoriasis. Evidence for currently approved systemic psoriasis treatments is presented in Table 2.6587 Patients prescribed these medications should be monitored carefully for hepatotoxicity.

Table 2 Potential Hepatotoxic Effects of Some of the Most Commonly Used Systemic Therapies Used in the Treatment of Moderate to Severe Psoriasis

Perhaps the most substantive evidence, and concern, regarding potential hepatotoxicity relates to methotrexate.6570 Early signs of negative effects of methotrexate on liver function include elevated hepatic transaminase levels, but of even greater concern are methotrexate-induced histological changes including aggravation of underlying fatty liver to NAFLD and steatohepatitis with possible fibrosis and cirrhosis.71,72 These more serious adverse hepatic effects have been reported to occur in about 5% of patients treated with chronic, low-dose methotrexate, although rarely in patients without additional clinical risk factors for hepatotoxicity, such as pre-existing liver disease, excessive alcohol abuse, hepatitis B or C, central obesity and type 2 diabetes.71 Given the hepatotoxic concerns, use of methotrexate is preferably avoided in these high-risk psoriasis patient groups, including those with established NAFLD, and methotrexate is contraindicated if bilirubin values are > 5 mg/dl.73

Cyclosporin may cause dose-dependent, reversible increases in serum bilirubin and liver enzymes, with post-marketing experience reporting hepatotoxicity and liver injury.65,68,74 Cyclosporin can also potentially worsen NAFLD by increasing serum lipid levels.75 Close monitoring of liver function test enzymes and lipid parameters is recommended as abnormal values may necessitate dose reductions.68,74,76 Another frequently occurring adverse event associated with cyclosporin use is hypertension, which would be expected to negatively impact NAFLD.

Cases of liver enzyme increases ( 3 upper limit of normal [ULN]) and elevation of total bilirubin levels ( 2 ULN) have been reported infrequently for dimethyl fumarate (DMF), with resolution after treatment discontinuation. Assessment of serum aminotransferases (eg, alanine aminotransferase [ALT], aspartate aminotransferase [AST]) and total bilirubin levels are recommended prior to treatment initiation and during treatment.68,77 DMF is considered to be a possible rare cause of liver toxicity.78

Acitretin has been associated with transient increases in liver enzymes, but hepatotoxicity has been reported rarely.65 As acitretin frequently causes hyperlipidemia, use is preferably avoided in patients with or at high risk of NAFLD.11,75,79,80

No hepatotoxic effects have been documented for apremilast65 and no routine laboratory monitoring is required. Apremilast is considered to be an unlikely cause of apparent liver injury.78

There is evidence of hepatotoxic risk with TNF- inhibitors such as infliximab, adalimumab, certolizumab and etanercept65,8183 and guidelines advocate monitoring liver enzymes during treatment.65 Furthermore, compared with conventional systemic therapies for psoriasis (methotrexate and cyclosporin), treatment with TNF inhibitors (adalimumab, infliximab and etanercept) was found to be associated with significant increases in bodyweight and body mass index in a network meta-analysis (6 studies and 862 psoriasis patients).84 Increases in body weight might be associated with a reduction in the therapeutic response and/or exacerbations of comorbidities, although this needs to be confirmed in prospective studies. TNF inhibitors should be used cautiously in patients with or at high risk of NAFLD.

Other biologicals such as anti-IL17 agents (brodalumab, ixekizumab and secukinumab) and ustekinumab (anti-IL12/23) have been less widely investigated, but have generally exhibited minimal hepatotoxicity (Table 2). A retrospective study of 44 psoriasis patients receiving ustekinumab identified 6 cases (13.6%) of mild elevated transaminases, with no cases of severe hypertransaminasemia.85 Overall, the reported rate of mild-to-moderate serum aminotransferase elevations following ustekinumab therapy was 0.5% to 1.4%. There have been no reports of treatment-related symptomatic acute liver injury or jaundice linked to ustekinumab therapy.78 Interestingly, limited data found no increases in bodyweight in patients treated with ustekinumab.84,88

Analysis of over 3000 patients in clinical trials of the IL-17 antagonist secukinumab in psoriasis showed low rates of serum liver enzyme elevations compared to placebo and no cases of treatment-related liver injury. Post-marketing data have revealed no reports of liver injury due to secukinumab therapy.78 Similarly, clinical trials (> 3000 patients) and post-marketing studies of the anti-IL-17 biologic brodalumab have shown no evidence of liver enzyme elevation nor liver injury attributable to the agent.78 Comparable rates of serum liver enzyme elevations with the IL-17 antagonist ixekizumab and placebo were found in clinical trials, with no treatment-related cases of liver injury. Although post-marketing approval experience with ixekizumab is relatively limited, no cases of treatment-related liver injury have been reported.78 For newer anti-IL-23 biologics recently approved for the treatment of psoriasis (eg, guselkumab, risankizumab, and tildrakizumab), no evidence of acute liver injury or reactivation of hepatitis B or worsening of hepatitis C has been reported to date, but clinical experience with these agents is very limited (Table 2).

Assessment of hepatoprotection can be derived from several different research pathways including in vitro experiments, animal models and monitoring of potential biomarkers (Table 3).89108 Given the common etiology of inflammation between psoriasis and NAFLD, it might be postulated that systemic therapies for psoriasis that attenuate systemic inflammation may also have beneficial effects on NAFLD by targeting pro-inflammatory cytokines.109 However, to date, clinical data are scarce for any direct hepatoprotective effects of systemic psoriasis treatments.

Methotrexate has exhibited beneficial effects on cardiovascular inflammation91,92 as well as decreasing serum levels of PCSK9, which is a likely marker of improved lipid metabolism (Table 3).89 However, a large placebo-controlled RCT failed to show any benefits of methotrexate on the risk of cardiovascular events.110 Furthermore, the potential hepatotoxic risks associated with methotrexate therapy, as discussed earlier, would generally preclude its use in patients with NAFLD.

From the clinical study of Krahel et al,89 acitretin treatment was shown to increase PCSK9 levels, but there was no correlation between these elevated levels and markers of liver function such as transaminases in hepatic steatosis and NASH in high-risk patients (Table 3). Fibroblast growth factors (FGFs) 21 and 23 are markers for cardiometabolic disorders which are common in psoriasis. In one small trial, acitretin was reported to decrease FGF-21 by three-fold which was significantly greater than the reduction observed with methotrexate.90 In an earlier small clinical trial, it was shown that patients with chronic psoriasis treated with acitretin had reduced retinol-binding protein-4 levels and decreased insulin resistance.111 Given the link between psoriasis and increased insulin resistance/diabetes these changes could be clinically meaningful if confirmed in a larger study.111

There is a paucity of data on the potential hepatoprotective effects of cyclosporin. In a small group of patients with moderate to severe psoriasis, comorbidity with diabetes or metabolic syndrome did not affect the efficacy of cyclosporin.93 In another clinical study, cyclosporin significantly improved patients psoriasis symptoms and this was associated with increased transcription activity of TGF1 at the end of treatment in those with or without diabetes, and with or without metabolic syndrome.112 Through inhibition of the enzyme cyclophilin, cyclosporin has shown beneficial effects on liver fibrosis and cirrhosis in some patients. However, calcineurin-related toxicity has limited the possibility of long-term therapy and has required that dosages be kept low.113

Apremilast is a phosphodiesterase 4 inhibitor shown to be clinically effective in patients with psoriasis and/or psoriatic arthritis and also acts as a metabolic modulator. In individual cases, apremilast was shown to improve glucose metabolism114 and lipid profile.115 Similar findings were reported in a 1-year open observational study (Table 3).94

DMF, and its active metabolite monomethylfumarate (MMF), are of particular interest in the context of this review due to their unique multiple mechanisms of action (Figure 4).102,103,105,116,117 These include immunomodulatory and anti-inflammatory mechanisms that are potentially advantageous in psoriatic patients with comorbid NAFLD (Table 3). 116 For example, DMF/MMF regulates cellular responses to oxidative stress by modulating intracellular glutathione levels.102 DMF/MMF also reduces oxidative stress through activation of Nrf2 (nuclear factor erythroid 2related factor 2) which stimulates cytoprotective and anti-inflammatory factors such as HO-1 (heme oxygenase-1);103,106,116 and through inhibiting genes regulated by the transcription factor HIF-1 (hypoxia-inducible factor 1-alpha) and STAT3/STAT1 (signal transducer and activator of transcription) pathways.116 MMF is an agonist for hydroxy-carboxylic acid receptor 2 (HCA2/ GPR109A [G protein-coupled receptor 109A]) which inhibits neutrophil adhesion and recruitment by COX-1 (cyclooxygenase 1) and PGE2 (prostaglandin E2).116 Fumaric acid esters significantly reduced total cholesterol, low-density lipoprotein and apolipoprotein B levels in a prospective trial involving psoriasis patients.96 In animal models, DMF has shown potential to ameliorate acetaminophen-induced hepatic injury in mice,107 as well as liver ischemia/reperfusion injury in rats.104 Improvement of liver function and anti-oxidant status might prove to be a promising treatment approach in patients with psoriasis and comorbid NAFLD.

Figure 4 Proposed mechanisms of action of dimethyl fumarate/monomethyl fumarate (DMF/MMF).

Note: Data from these studies.102,103,105,116,117

Adalimumab was shown to reduce key markers of systemic inflammation including glycoprotein acetylation, CRP, IL-6 and TNF and this was associated with beneficial cardiovascular effects such as improved endothelial dysfunction as measured by flow-mediated dilation.95,96 TNF- inhibitors have been reported to improve insulin resistance in patients with rheumatoid arthritis118,119 and ankylosing spondylitis,118,120 but not in obese patients with either type 2 diabetes121 or metabolic syndrome.122,123 Seitz et al investigated the impact of TNF- inhibitors on the presence of liver fibrosis in patients with psoriatic arthritis and rheumatoid arthritis treated with methotrexate.124 Patients with psoriatic arthritis had a higher incidence of liver steatosis and hyperlipidemia and in this study. TNF- inhibitors exerted a protective effect against the development of liver fibrosis.

Secukinumab has been shown to have neutral effects on fasting plasma glucose, lipid parameters and liver enzymes, while reducing levels of CRP, a marker for systemic inflammation. It was also associated with a reduction in markers of oxidative stress. Secukinumab produced greater improvement in arterial elasticity, coronary artery function and myocardial deformation indices compared with methotrexate and cyclosporin. However, no data are available on the impact of these effects on liver function.97,98

Ustekinumab significantly decreases pro-inflammatory cytokines, such as TNF-, IL-1b, IL-17a and IL-6. VCAM-1 was significantly reduced by ustekinumab at 12 weeks, although this effect was not sustained at 1 year. Overall, ustekinumab transiently reduced aortic vascular inflammation at 12 weeks and, longer term, produced a more durable reduction in inflammatory cytokines associated with cardiovascular disease.101 No published studies have investigated the impact of this effect on liver inflammation and function.

NAFLD is an increasingly prevalent and clinically important comorbidity occurring in up to 65% of patients with psoriasis. The occurrence of NAFLD in psoriasis can have significant morbidity and mortality potential. There are multiple pathophysiological pathways that link psoriasis with NAFLD, in particular systemic inflammation and insulin resistance. Reducing systemic inflammatory burden may provide an opportunity to reduce the progression or even ameliorate NAFLD.

A number of conventional drugs used in the treatment of psoriasis are associated with hepatotoxic risk. Most evidence relates to use of methotrexate which, along with other NAFLD-promoting drugs, is best avoided in psoriasis patients with clinical risk factors for hepatotoxicity or established NAFLD. Despite the expectation that systemic psoriasis treatments might reduce the progression of or even ameliorate NAFLD by targeting pro-inflammatory cytokines, clinical data for direct hepatoprotective effects of these therapies are relatively scarce. Through its pleiotropic mechanisms of action contributing to its anti-inflammatory activity, DMF may have additional hepatoprotective effects of possible advantage in psoriatic patients with NAFLD.

However, real-world data on the effects of DMF in patients with psoriasis and concomitant NAFLD is currently lacking, and further studies with DMF and other systemic psoriasis therapies in routine clinical practice would be needed to inform therapeutic decision-making strategies for psoriasis patients with NAFLD.with DMF and other systemic psoriasis therapies in routine clinical practice would be needed to inform therapeutic decision-making strategies for psoriasis patients with NAFLD.

The authors thank Dr Rob Furlong and Dr Steve Clissold on behalf of Content Ed Net, Spain for editorial support, which was sponsored by Almirall, Barcelona, Spain, and performed in line with Good Publishing Practice (GPP-3) guidelines.

This article was supported by an unrestricted educational grant from Almirall, Barcelona, Spain.

DMWB is a consultant/speaker for AbbVie, Almirall, Celgene, Eli Lilly, Galderma, Janssen, LEO Pharma, Novartis, and Regeneron/Sanofi Genzyme. IK is an employee of Almirall, Barcelona, Spain. SP is a consultant/speaker for AbbVie, Almirall, Celgene, Eli Lilly, Galderma, Janssen, LEO Pharma, Novartis, UCB, and Pfizer. The authors report no other conflicts of interest in this work.

1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157(8):940946. doi:10.1001/jamadermatol.2021.2007

2. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263271. doi:10.1016/S0140-6736(07)61128-3

3. Balato N, Megna M, Palmisano F, et al. Psoriasis and sport: a new ally? J Eur Acad Dermatol Venereol. 2015;29(3):515520. doi:10.1111/jdv.12607

4. Gisondi P, Bellinato F, Girolomoni G, et al. Pathogenesis of chronic plaque psoriasis and its intersection with cardio-metabolic comorbidities. Front Pharmacol. 2020;11:117. doi:10.3389/fphar.2020.00117

5. Prussick RB, Miele L. Non-alcoholic fatty liver disease in patients with psoriasis: a consequence of systemic inflammatory burden? Br J Dermatol. 2018;179(1):1629. doi:10.1111/bjd.16239

6. Lnnberg AS, Skov L. Co-morbidity in psoriasis: mechanisms and implications for treatment. Expert Rev Clin Immunol. 2017;13:2734. doi:10.1080/1744666X.2016.1213631

7. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:10731113. doi:10.1016/j.jaad.2018.11.058

8. Sheka AC, Adeyi O, Thompson J, et al. Nonalcoholic steatohepatitis: a review. JAMA. 2020;323(12):11751183. doi:10.1001/jama.2020.2298

9. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34(3):274285. doi:10.1111/j.1365-2036.2011.04724.x

10. Younossi ZM. The epidemiology of nonalcoholic steatohepatitis. Clin Liver Dis (Hoboken). 2018;11(4):9294. doi:10.1002/cld.710

11. Belinchn-Romero I, Bellot P, Romero-Prez D, et al. Non-alcoholic fatty liver disease is associated with bacterial translocation and a higher inflammation response in psoriatic patients. Sci Rep. 2021;11(1):8593. doi:10.1038/s41598-021-88043-8

12. Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA. 2015;313(22):22632273. doi:10.1001/jama.2015.5370

13. Adams LA, Lymp JF, St Sauver J, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113121. doi:10.1053/j.gastro.2005.04.014

14. Harrison SA, Di Bisceglie AM. Advances in the understanding and treatment of nonalcoholic fatty liver disease. Drugs. 2003;63(22):23792394. doi:10.2165/00003495-200363220-00001

15. Magdaleno-Tapial J, Valenzuela-Oate C, Ortiz-Salvador JM, et al. Prevalence of non-alcoholic fatty liver and liver fibrosis in patients with moderate-severe psoriasis: a cross-sectional cohort study. Australas J Dermatol. 2020;61(2):105109. doi:10.1111/ajd.13175

16. Ortolan A, Lorenzin M, Tadiotto G, et al. Metabolic syndrome, non-alcoholic fatty liver disease and liver stiffness in psoriatic arthritis and psoriasis patients. Clin Rheumatol. 2019;38(10):28432850. doi:10.1007/s10067-019-04646-7

17. Olveira A, Herranz P, Montes ML. Psoriasis and fatty liver: a harmful synergy. Rev Esp Enferm Dig. 2019;111(4):314319. doi:10.17235/reed.2019.6263/2019

18. Hamaguchi M, Kojima T, Takeda N, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med. 2005;143(10):722728. doi:10.7326/0003-4819-143-10-200511150-00009

19. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology. 2012;142(7):15921609. doi:10.1053/j.gastro.2012.04.001

20. Golabi P, Otgonsuren M, de Avila L, et al. Components of metabolic syndrome increase the risk of mortality in nonalcoholic fatty liver disease (NAFLD). Medicine (Baltimore). 2018;97(13):e0214. doi:10.1097/MD.0000000000010214

21. Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci. 2013;14(10):2070420728. doi:10.3390/ijms141020704

22. Gisondi P, Galvan A, Idolazzi L, et al. Management of moderate to severe psoriasis in patients with metabolic comorbidities. Front Med (Lausanne). 2015;2:1. doi:10.3389/fmed.2015.00001

23. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(2):367378. doi:10.1053/j.gastro.2015.04.005

24. Lonardo A, Loria P, Carulli N. Concurrent non-alcoholic steatohepatitis and psoriasis. Report of three cases from the POLI.ST.E.N.A. study. Dig Liver Dis. 2001;33(1):8687. doi:10.1016/S1590-8658(01)80144-4

25. Gisondi P, Targher G, Zoppini G, et al. Non-alcoholic fatty liver disease in patients with chronic plaque psoriasis. J Hepatol. 2009;51(4):758764. doi:10.1016/j.jhep.2009.04.020

26. Gisondi P, Barba E, Girolomoni G. Non-alcoholic fatty liver disease fibrosis score in patients with psoriasis. J Eur Acad Dermatol Venereol. 2016;30(2):282287. doi:10.1111/jdv.13456

27. van der Voort EA, Koehler EM, Dowlatshahi EA, et al. Psoriasis is independently associated with nonalcoholic fatty liver disease in patients 55 years old or older: results from a population-based study. J Am Acad Dermatol. 2014;70(3):517524. doi:10.1016/j.jaad.2013.10.044

28. Madanagobalane S, Anandan S. The increased prevalence of non-alcoholic fatty liver disease in psoriatic patients: a study from South India. Australas J Dermatol. 2012;53(3):190197. doi:10.1111/j.1440-0960.2012.00905.x

29. Abedini R, Salehi M, Lajevardi V, et al. Patients with psoriasis are at a higher risk of developing nonalcoholic fatty liver disease. Clin Exp Dermatol. 2015;40(7):722727. doi:10.1111/ced.12672

30. Ogdie A, Grewal SK, Noe MH, et al. Risk of incident liver disease in patients with psoriasis, psoriatic arthritis, and rheumatoid arthritis: a population-based study. J Invest Dermatol. 2018;138(4):760767. doi:10.1016/j.jid.2017.10.024

31. Candia R, Ruiz A, Torres-Robles R, et al. Risk of non-alcoholic fatty liver disease in patients with psoriasis: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2015;29(4):656662. doi:10.1111/jdv.12847

32. Phan K, Onggo J, Charlton O, et al. Relationship between psoriasis and non-alcoholic fatty liver disease - Updated systematic review and adjusted meta-analysis. Australas J Dermatol. 2019;60(4):e352e355. doi:10.1111/ajd.13015

33. Yang YW, Keller JJ, Lin HC. Medical comorbidity associated with psoriasis in adults: a population-based study. Br J Dermatol. 2011;165(5):10371043. doi:10.1111/j.1365-2133.2011.10494.x

34. Gandha N, Wibawa LP, Jacoeb TNA, et al. Correlation between psoriasis severity and nonalcoholic fatty liver disease degree measured using controlled attenuation parameter. Psoriasis (Auckl). 2020;10:3944. doi:10.2147/PTT.S272286

35. Roberts KK, Cochet AE, Lamb PB, et al. The prevalence of NAFLD and NASH among patients with psoriasis in a tertiary care dermatology and rheumatology clinic. Aliment Pharmacol Ther. 2015;41(3):293300. doi:10.1111/apt.13042

36. Romero-Prez D, Belinchn-Romero I, Bellot P, et al. Nonalcoholic fatty liver disease puts patients with psoriasis at greater cardiovascular risk. Australas J Dermatol. 2019;60:e304e310. doi:10.1111/ajd.13098

37. National Guideline Centre (UK). Non-alcoholic fatty liver disease: assessment and management. London: National Institute for Health and Care Excellence (UK); 2016. (NICE Guideline, No. 49.) 5, Risk factors for NAFLD. Available from: https://www.ncbi.nlm.nih.gov/books/NBK384735. Accessed November 10, 2021.

38. Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24(7):908922. doi:10.1038/s41591-018-0104-9

39. Kamiya K, Kishimoto M, Sugai J, et al. Risk factors for the development of psoriasis. Int J Mol Sci. 2019;20:4347. doi:10.3390/ijms20184347

40. Ganzetti G, Campanati A, Molinelli E, et al. Psoriasis, non-alcoholic fatty liver disease, and cardiovascular disease: three different diseases on a unique background. World J Cardiol. 2016;8(2):120131. doi:10.4330/wjc.v8.i2.120

41. Grelj J, Mlinari-Raan I, Marko PB, et al. Polymorphisms in GNMT and DNMT3b are associated with methotrexate treatment outcome in plaque psoriasis. Biomed Pharmacother. 2021;138:111456. doi:10.1016/j.biopha.2021.111456

42. Heitmann J, Frings VG, Geier A, et al. Non-alcoholic fatty liver disease and psoriasis - is there a shared proinflammatory network? J Dtsch Dermatol Ges. 2021;19(4):517528.

43. Harden JL, Krueger JG, Bowcock AM. The immunogenetics of psoriasis: a comprehensive review. J Autoimmun. 2015;64:6673. doi:10.1016/j.jaut.2015.07.008

44. Liang Y, Sarkar MK, Tsoi LC, et al. Psoriasis: a mixed autoimmune and autoinflammatory disease. Curr Opin Immunol. 2017;49:18. doi:10.1016/j.coi.2017.07.007

See more here:
Non-alcoholic fatty liver disease in patients with psoriasis | PTT - Dove Medical Press

Posted in Psoriasis | Comments Off on Non-alcoholic fatty liver disease in patients with psoriasis | PTT – Dove Medical Press

Perception of the threat, mental health burden, and healthcare-seeking behavior change among psoriasis patients during the COVID-19 pandemic – DocWire…

Posted: at 7:23 pm

This article was originally published here

PLoS One. 2021 Dec 9;16(12):e0259852. doi: 10.1371/journal.pone.0259852. eCollection 2021.

ABSTRACT

This study aimed to investigate the perceived threat, mental health outcomes, behavior changes, and associated predictors among psoriasis patients during the COVID-19 pandemic. The COVID-19 has been known to increase the health risks of patients with psoriasis owing to patients immune dysregulation, comorbidities, and immunosuppressive drug use. A total of 423 psoriasis patients not infected with COVID-19 was recruited from the Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Chang Gung Memorial Hospital, and China Medical University Hospital from May 2020 to July 2020. A self-administered questionnaire was used to evaluate the perceived threat, mental health, and psychological impact on psoriasis patients using the Perceived COVID-19-Related Risk Scale score for Psoriasis (PCRSP), depression, anxiety, insomnia, and stress-associated symptoms (DAISS) scales, and Impact of Event Scale-Revised (IES-R), respectively. Over 94% of 423 patients with psoriasis perceived threat to be 1 due to COVID-19; 18% of the patients experienced psychological symptoms more frequently 1, and 22% perceived psychological impact during the pandemic to be 1. Multivariable linear regression showed that the higher psoriasis severity and comorbidities were significantly associated with higher PCRSP, DAISS, and IES-R scores. The requirement for a prolonged prescription and canceling or deferring clinic visits for psoriasis treatment among patients are the two most common healthcare-seeking behavior changes during the COVID-19 pandemic. Psoriasis patients who perceived a higher COVID-19 threat were more likely to require a prolonged prescription and have their clinic visits canceled or deferred. Surveillance of the psychological consequences in psoriasis patients due to COVID-19 must be implemented to avoid psychological consequences and inappropriate treatment delays or withdrawal.

PMID:34882690 | DOI:10.1371/journal.pone.0259852

Link:
Perception of the threat, mental health burden, and healthcare-seeking behavior change among psoriasis patients during the COVID-19 pandemic - DocWire...

Posted in Psoriasis | Comments Off on Perception of the threat, mental health burden, and healthcare-seeking behavior change among psoriasis patients during the COVID-19 pandemic – DocWire…

SARS: A Mystery Illness To Impact Air Travel – Aviation Week

Posted: at 7:21 pm

The SARS pandemic spread around the world in 2003 and we can see how, even as far back as 20 years ago, world health is a priority for air travel which relies on health information and government guidelines to restrict travel to stop the virus spreading.

SARS: A New Blow

In 2002 airlines were affected by the war on terrorism, the war in Iraq and what reporter Frances Fiorino wrote on page 59 of the April 7 issue of the magazine, germ warfare, against severe acute respiratory syndrome (SARS), a deadly atypical pneumonia that is spreading rapidly worldwide.

As such, the World Health Organisation (WHO) issued emergency travel advisory postponing travel to Hong Kong and Chinas Guangdong province where the virus was believed to have originated. Read more about this in our archive.

SARS In Europe

By April 28, we were reporting on news that European airlines were trying to allay potential passengers fears about the danger of air travel, and that WHO had conducted aggressive research and had confirmed the SARS virus as a member of the coronavirus family.

French health epidemiologist Jean-Baptiste-Brunet said: Medical detectives should be aware that SARS, which was carried from Asia to most of the world, is, of course, directly tied to air transportation, he added. But commercial transports are not the cause, just the conduit.

Read more of this article in which it discusses the effect of air conditioning and pressurized cabins.

SARS Redux

By 2005, we were reporting on having weathered the severe economic blows of SARS but the world is about to battle another new enemy, avian influenza virus, which while the strain causes severe illness in fowl, is now being transmitted from birds to humans, according to a WHO report. And it is spreading, with the fear that the vaccine may not necessarily work against the latest strain.

The image in the article shows that infrared monitors had been installed in Taiwan's Chiang Kai-shek International Airport to scan travelers for fever, a symptom of SARS.

Read more on this analysis.

Follow this link:

SARS: A Mystery Illness To Impact Air Travel - Aviation Week

Posted in Germ Warfare | Comments Off on SARS: A Mystery Illness To Impact Air Travel – Aviation Week

SomaLogics SomaScan Assay used in largest proteomic study to date, bridging the gap between genomics and disease – Yahoo Finance

Posted: at 7:19 pm

BOULDER, Colo., Dec. 09, 2021 (GLOBE NEWSWIRE) -- In a new study published in Nature Genetics, scientists at deCODE genetics, a subsidiary of Amgen, used SomaLogics (NASDAQ: SLGC) SomaScan Assay to measure blood proteins in 35,559 Icelanders and mapped them to 27 million genetic sequence variants. Using this vast amount of proteomic data, these researchers hope to demonstrate that combining protein measurements at population scale with genetic data on disease will dramatically impact understanding of human diseases and potential drug targets. This new study was the largest proteomic study published to date with 170 million protein measurements.

Less than 10% of human disease is driven by genetics. Plasma proteomics, the study of blood proteins, can help bridge the gap between genomics and disease discovery. This paper found that linking genes to proteins, and then to diseases can show patterns between the factors that cause a disease and the factors that are a consequence of a disease. This process may give a roadmap of how diseases develop and offer potential drug targets.

In this study, the plasma levels of 4,719 blood proteins were tested for genetic associations with 373 diseases and traits, producing 257,490 of these associations. SomaLogics SomaScan Assay was used to find genetic variant-protein target associations, called protein quantitative trail loci or pQTLs. In the study, 94% of the proteins measured using the SomaScan Assay showed an associated pQTL, resulting in more than 18,000 pQTLs. Ninety-three percent of these pQTLs are considered novel. The study also identified 938 genes encoding as potential protein drug targets for various diseases.

Our SomaScan Assay offers the ability to measure and identify the largest percentage of the human proteome at commercial scale on the market today and it proved to be exquisitely specific in this study, said SomaLogic Chief Executive Officer Roy Smythe, M.D. We hope that this study, and more like it, will help to provide the vital information that can be added to genetic data to create a more comprehensive understanding of human biology, and increasingly power more effective treatments for human disease.

Story continues

About SomaLogicSomaLogic (Nasdaq: SLGC) seeks to deliver precise, meaningful, and actionable health-management information that empowers individuals worldwide to continuously optimize their personal health and wellness throughout their lives. This essential information, to be provided through a global network of partners and users, is derived from SomaLogics personalized measurement of important changes in an individuals proteins over time. For more information, visit http://www.somalogic.com and follow @somalogic on Twitter.

Forward Looking Statements Disclaimer This press release contains certain forward-looking statements within the meaning of the federal securities laws with respect to the proposed business combination between SomaLogic and CM Life Sciences II and otherwise, including statements regarding the anticipated benefits of the business combination, the anticipated timing of the business combination, expansion plans, projected future results and market opportunities of SomaLogic. These forward-looking statements generally are identified by the words believe, project, expect, anticipate, estimate, intend, strategy, future, opportunity, plan, may, should, will, would, will be, will continue, will likely result, and similar expressions. Forward-looking statements are predictions, projections and other statements about future events that are based on current expectations and assumptions and, as a result, are subject to risks and uncertainties. Forward looking statements do not guarantee future performance and involve known and unknown risks, uncertainties and other factors. Many factors could cause actual future events to differ materially from the forward-looking statements in this press release, including factors which are beyond SomaLogics or CM Life Sciences IIs control. You should carefully consider the risks and uncertainties described in the Risk Factors section of the CM Life Sciences IIs registration statement on Form S-4 (File No. 333-256127) (the Registration Statement) and the definitive proxy statement/prospectus included therein. These filings identify and address important risks and uncertainties that could cause actual events and results to differ materially from those contained in the forward-looking statements. Forward-looking statements speak only as of the date they are made. Readers are cautioned not to put undue reliance on forward-looking statements, and SomaLogic and CM Life Sciences II assume no obligation and do not intend to update or revise these forward-looking statements, whether as a result of new information, future events, or otherwise. Neither SomaLogic nor CM Life Sciences II gives any assurance that either SomaLogic or CM Life Sciences II or the combined company will achieve its expectations.

SomaLogic Contact Emilia Costales 720-798-5054ecostales@somalogic.com

Investor ContactLynn Lewis or Marissa BychGilmartin Group LLCinvestors@somalogic.com

Visit link:
SomaLogics SomaScan Assay used in largest proteomic study to date, bridging the gap between genomics and disease - Yahoo Finance

Posted in Human Genetics | Comments Off on SomaLogics SomaScan Assay used in largest proteomic study to date, bridging the gap between genomics and disease – Yahoo Finance

A tiny primate may join the ranks of the world’s model organisms – The Economist

Posted: at 7:19 pm

TREE 2B, RANOMAFANA, is not an address recognised by Madagascars postal service. It is, though, someones home. The someone in question is a mouse lemur called Judah, the 349th participant to be enrolled into a project run by Mark Krasnow, a biochemist at Stanford University, in California.

Your browser does not support the

Enjoy more audio and podcasts on iOS or Android.

Judahs involuntary membership of the project began when he found himself trapped inside a metal box. He had been lured there by a bait of banana put there by Dr Krasnows collaborators, Haja Ravelonjanahary and Mahery Razafindrakoto of the ValBio research centre on the edge of Ranomafana National Park, 260km south of Antananarivo. Judahs captivity was temporary, for he was released back into his home at 2B about six hours later. But in the interim he was subjected to various indignities. He had his testes measured, a blood sample taken and he was made to do exercises to see how strong he was. He also had a tiny transponder inserted under his skin so that he could be identified next time he was caught.

Judah, and his 348 predecessors similarly trapped and released by biologists at ValBio, are among the first recruits to what is, on the face of it, an extraordinarily ambitious undertaking. For Dr Krasnows plan is to add mouse lemurs to the short and rather random list of so-called model organisms. These are species which, for various reasons, biologists know a lot about. And, since knowledge breeds knowledge, they tend to be the ones about which further knowledge accumulates.

Model organisms assist all sorts of biological research, but a lot of it is medical. And here there is a problem. Ideally, medical research would be done on species that resemble Homo sapiens. But working on human beings closest relativesapes and monkeysis increasingly hard to do. First, such large animals are expensive to keep. Second, that expense means they are often unavailable in the numbers needed for statistically significant work. Third, public opinion, at least in the West, is swinging against their use.

Mice, one common alternative to primates, are cheap, abundant and less prone to stir consciences. But they can only take you so far. Though mammals, they are not close relatives of people. Sometimes that lack of relatedness can be finessed by inserting human genes that are relevant to the matter under investigation. But even then, the underlying platform is still a rodent, not a primate. By contrast, a mouse lemur, though it looks and behaves a bit like a mouse, and is not much bigger, is indeed a primate, and so is much more similar to a human being than a rodent is.

Mice, moreover, have short lives, and thus high turnover. But mouse lemurs can live for 14 years in captivity and maybe ten in the wild. That is a nice compromise between a period brief enough to arrive at conclusions that are useful (and will result in career-enhancing research papers), and long enough to be more similar to a human beings life-history. Yet, like mice, mouse lemurs breed prolifically and quickly, with a gestation period of just two months and maturity achieved within six to eight months. And not just in a laboratory. In Madagascar there are millions of themfor, contrary to common perception, not all lemur species are endangered.

What is particularly intriguing for Dr Krasnow and his colleagues, though, is that, in captivity at least, mouse lemurs suffer several illnesses which affect humans too. These include Alzheimers and other neurodegenerative disorders, cardiac arrhythmias, metastatic uterine cancer, strokes and atherosclerosis, the furring of the arteries that can lead to a heart attack.

Model organisms tend to happen by accident. Yeast is used by brewers and bakers, so is an obvious topic for study. Fruit flies were picked by Thomas Morgan, an early geneticist, because they are easy to breed in large numbersand it helped that some of their cells have giant chromosomes which showed up well under the microscopes of the day. And mice were kept as pets by fanciers long before one saw the inside of a laboratory cage.

Dr Krasnows plan to add mouse lemurs to the list was slightly less accidental than these. It began in 2009, when he charged his daughter Maya, then still at school, and two of her friends to come up with a new model organism for studying primates as a summer project in his laboratory. After reviewing the gamut of the primate order, which contains about 500 species, and also looking at a few outliers such as tree shrews, Krasnow junior and her two compadres settled on mouse lemurs. Not only are these abundant and fast-breeding, they also do well in captivity, as a 60-year-old colony of them in France testifies.

Not one to ignore his daughters advice, Dr Krasnow investigated in more detail. In 2011, he organised a workshop of lemur biologists at the Howard Hughes Medical Institute, in Virginia, to kick the idea around. It found favour, and in particular it accelerated the completion of a genome-sequencing project for the animalsa sine qua non for any self-respecting model organism. It also introduced Dr Krasnow to the idea that fieldwork might be an important part of his proposal.

That, in some ways, is the most intriguing idea of the lot. Most biologists working with model organisms make a fetish of control. Mice, in particular, are often bred deliberately to be as genetically similar to one another as possible, within a given line. Dr Krasnow has the opposite plan. Genetic analysis is now so cheap that every animal involved in a project can be sequenced. Made visible in this way, diversity is as much an opportunity as a problem, for that information can be correlated not only with obvious, medically relevant stuff, such as disease manifestation, but also with behaviourand behaviour expressed in the wild, not just in the restricted environment of a laboratory.

That insight led to collaboration with Patricia Wright, a primatologist at the State University of New York, Stony Brook, who helped encourage the Malagasy government to found Ranomafana, and who has been working there for decades. And that led to the lemur-trapping project now joined by Judah. One early discovery from the genetic analyses made possible by this project (admittedly, one that is not of much obvious medical use) is that what appeared to be one species of brown mouse lemur, the species Dr Krasnow and Dr Wright thought they were investigating, is actually two. They live in the same range and are indistinguishable to the human eye. But they can clearly tell each other apart because their genetics show that they diverged several million years ago, and do not interbreed.

Dr Krasnow does, however, have high hopes of the medical side. In particular, as they age, mouse lemurs in captivity sometimes develop the plaques and tangles of abnormal protein seen in human Alzheimers patients. At the same time, they develop behavioural abnormalities, such as forgetfulness. Nothing similar happens naturally in mice. Nor do mice develop the sorts of heart arrhythmias seen in people. But mouse lemurs do. In fact, he and his colleagues have now identified nine types of arrhythmia in their lemurs, each of which corresponds to one found in people.

Though the animals will not be subjected to invasive sampling while alive, the ability to identify them individually in the wild means that their behaviour can be studied, to see if it changes as they age in ways similar to ageing in people. What else might be discovered from this behavioural work remains to be seen, for this is an old-fashioned experiment of the sort that is not testing a specific hypothesis but, rather, searching for leads to pursue.

Meanwhile, back in the lab, and thanks to a technique called single-cell RNA expression profiling, Dr Krasnow and his Stanford colleague Stephen Quake have built a near-complete atlas of lemur cell typesabout 750 in all. This permits a whole new level of investigation. For example, they were able to identify a metastatic cell in the lung of an animal that had had to be put down because it had cancer, as deriving from that animals uterus.

It could all fall flat on its face, of course. For one thing, the field data may shed no light on disease-relevant biology after all. Most of the illnesses that Dr Krasnow is interested in manifest themselves in later life. In humans, such diseases are associated with behaviours which evolution did not foresee, such as consuming processed food or sitting at a desk all day. Since being locked up in a cage and fed a reliable supply of food is equally unnatural, that may also be true for lemurs. It is therefore by no means clear that looking at wild lemurs will add anything. Moreover, illnesses like Alzheimers are not exactly life-elongating. In the wild, any individual manifesting them would probably get short shrift from natural selection. Indeed, there is a whole body of theory which suggests the very reason they manifest only in old age is because, in a state of nature, a human being would probably have died or been killed before they had had a chance to appear.

There is also the political side of things. Though researchers on other species are unlikely to be hostile in principle to mouse lemurs joining the model-animal-research party, whether they will co-operate with the group of newcomers in the far corner who are talking animatedly about the critters remains to be seen. Model animals do, however, require a consensus that that is what they areand this consensus is best built by lots of people studying lots of different aspects of them. So if not enough people join the mouse-lemur clique, the project will be doomed.

Another potential threat is that, although mouse lemurs do not truly share the mini-me human lookalikeness of monkeys and apes, they are still pretty cute. Those opposed to animal experiments of any sorteven the carefully non-invasive work being done by Dr Krasnow and Dr Wrightcould probably make something of that. And the very similarity of physiology to humans that makes the lemurs an attractive subject of study might also be used to argue that they should not be used in research.

Still, it is a bold idea, and certainly worth pursuing. Perhaps the cross-fertilisation of laboratory and field studies in this way will, indeed, turn out to be the wave of the future. In army terms, mouse lemurs are now at boot camp, undergoing basic training. Whether they will pass muster remains to be seen.

To enjoy more of our mind-expanding science coverage, sign up to Simply Science, our weekly newsletter.

This article appeared in the Science & technology section of the print edition under the headline "New Model Army"

View post:
A tiny primate may join the ranks of the world's model organisms - The Economist

Posted in Human Genetics | Comments Off on A tiny primate may join the ranks of the world’s model organisms – The Economist

Seven Bridges Launches Unified Patient Network to Facilitate Clinical Research with Aim to Advance Precision Medicine and Improve Patient Care -…

Posted: at 7:19 pm

The UPN aims to accelerate the scientific advancement and clinical implementation of precision medicine.

The UPN will operate as a collaborative group of nonprofit academic health centers and other health systems participating in clinical research that is enabled by the UPN through biopharma funding. The initiative aims to advance medical science and identify the genetic roots of human health and disease, by building a large database of research participants' de-identified clinical and genomic data that will be available for research purposes by researchers at UPN member health systems and biopharma companies.

"The UPN aims to accelerate the scientific advancement and clinical implementation of precision medicine, in a manner that provides truly unprecedented return-of-value to our health system members and their research participants, via clinical whole genome sequencing, genetic screening, genetic counseling, research tools, data assets, collaborative interoperability and a significant incremental funding stream, at no charge to the health systems or patients. With the ability to extend invitations to participate to patients across multiple health systems, UPN will be able to provide biopharma researchers unprecedented access to highly harmonized de-identified whole genome and longitudinal EHR data regarding highly specific cohorts drawn from thousands of research participants," said William Moss, CEO of Seven Bridges and the UPN.

"Our unique approach enables us to simultaneously optimize clinical and scientific research value on demand, without making it needlessly difficult to combine de-identified sequencing data and EHR content for large populations, resulting in a highly efficient operating model," Moss continued.

The UPN will operate across many disease states and therapeutic areas, including rare, complex neurodegenerative, psychiatric and autoimmune diseases and disorders, as well as cancer, cardiology and common diseases such as diabetes. The network will begin by aggregating very specific cohorts, measured on the order of thousands of research participants. Ultimately, the UPN's goal is to include over five million sequenced patient volunteers in its active network.

Patients who volunteer for clinical research studies conducted as part of the UPN will need to provide informed consent to participate and can opt at any time to have their de-identified genetic and clinical data removed from the network's database. The privacy of research participants will be strictly protected. Only de-identified genomic and clinical electronic health record (EHR) content will be made available via thehighly secure database. Such data will be used, as part of institutional review board (IRB)-approved research studies, to understand how genes contribute to or protect against various diseases and influence how well patients respond to treatment. In some cases, genomic sequencing may reveal genetic alterations that could change the course of a patient's treatment.

Washington University School of Medicine in St. Louis and its affiliated health system, BJC HealthCare, is the first academic health system to join the UPN as a founding member. The network will expand to include other health systems and consented research participants from those institutions.

"Washington University has a long-standing commitment to advance precision medicine and bring more personalized treatments to our patients," said David H. Perlmutter, MD, Executive Vice Chancellor for Medical Affairs, the George and Carol Bauer Dean, and the Spencer T. and Ann. W. Olin Distinguished Professor at the School of Medicine. "The UPN will be an important part of making this a reality by providing a platform to aggregate clinical and genomic data from research participants and share de-identified data with researchers. The UPN strategy takes another important step in positioning our communities for a new era of precision medicine, with more personalized diagnoses and treatments across many diseases."

With the launch of the UPN, Seven Bridges has assembled a world-class executive leadership team, including Chief Clinical and Research Officer Dr. David Ledbetter. Dr. Ledbetter was previously executive vice president and chief scientific officer at Geisinger Health System where he was the principal investigator for the MyCode biobank and precision health program that exceeded 175,000 patients with exome sequence data linked to rich, longitudinal EHR and other clinical data. He has also been a professor of human genetics at Emory University School of Medicine, the University of Chicago School of Medicine, and Baylor College of Medicine.

"Previous experience from large population genomics projects have shown that healthcare data combined with genomics data can rapidly accelerate knowledge to help prevent disease or to improve patient outcomes, as well as identify new drug targets forbiopharma pipelines.Until now, these valuable data sets have been confined to single health systems rather than aggregated and shared across multiple health systems, or have been siloed by individual commercial entities. The Unified Patient Network will unlock the long-promised benefits of our national investments in health IT and population scale genomics," said David Ledbetter, PhD, Chief Clinical and Research Officer for the UPN. "This unique multi-sided network will bring these stakeholderstogether with the aim of advancing precision medicine through a genomics-enabled learning health system, whereby patients can have their genomes sequenced free of charge, giving researchers greater insights into patient health risks, and biopharmaceutical companies to more easily identify cohorts of patients as part of drug discovery efforts, thereby lowering everyone's costs."

Phillip Payne, PhD, Associate Dean for Health Information and Data Science and Chief Data Scientist at Washington University School of Medicine said,"By bringing health systems together, we can enroll more patients into UPN studies, helping to speed innovative research while also protecting patients' identities and confidentiality. Genome sequencing is expensive and out of reach for most patients, but the UPN is providing such sequencing to research volunteers free of charge. This opens up the technology to many more people, including those in under-resourced communities, and is a huge win from an access and affordability standpoint."

The UPN will receive funding from biopharma companies that request access to research participants' de-identified genomic and health information for the companies' own research. A portion of that funding will be returned to the health systems in the UPN, Payne said, to support efforts to improve their patients' access to medical care and drive the institutions' research and teaching missions.

The UPN has assembled a dynamic team of partners and supporters to advance the high level of collaboration required to build, grow and sustain the network. The ecosystem includes Seven Bridges, Genome Medical, Amazon Web Services, the Broad Institute of MIT and Harvard, Illumina and others.

Partnering closely with Seven Bridges, the UPN is leveraging the Seven Bridges highly-secured research and development ecosystem as the interoperability infrastructure for the network community, and enabling exploration and analysis for complex cohort stratification across populations of millions of patients, via the ARIA scientific intelligence system. The content will be made available only to credentialed researchers as part of IRB-approved research studies, as mutually agreed to by the UPN and the health system members, by leveraging Seven Bridges' proven security, authentication and authorization protocols and technologies.

The UPN is also working with Genome Medical, the leading telehealth provider of genetics and genomics care. "We are pleased that our genomic specialists and technology-enabled clinical support tools will expand access to the benefits of genomic science and medicine within the network," said Lisa Alderson, CEO and co-founder of Genome Medical. "By helping patients and their clinicians better understand and interpret genomic data, health care can best meet the needs of individual patients."

For information on UPN, please visit linkedin.com/company/unifiedpatientnetwork or unifiedpatientnetwork.org.

About Seven Bridges

Seven Bridges enables researchers to extract meaningful insights from genomic and phenotypic data in order to advance precision medicine. The Seven Bridges Ecosystem consists of a compliant analytic platform, intelligently curated content, transformative algorithms, unprecedented access to federated data sets, and expert on-demand professional services. This holistic approach to bioinformatics is enabling researchers at the world's leading academic, biotechnology, clinical diagnostic, government, medical centers, and pharmaceutical entities to increase R&D efficiency, enhance the hypothesis resolution process, isolate critical biomarkers, and even turn a failing clinical trial around while also reducing computational workflow times and data storage costs. To learn more, visit sevenbridges.comor follow us on LinkedInand Twitter.

Media ContactValerie Enes[emailprotected]+1 408-497-8568

SOURCE Seven Bridges

See the original post here:
Seven Bridges Launches Unified Patient Network to Facilitate Clinical Research with Aim to Advance Precision Medicine and Improve Patient Care -...

Posted in Human Genetics | Comments Off on Seven Bridges Launches Unified Patient Network to Facilitate Clinical Research with Aim to Advance Precision Medicine and Improve Patient Care -…