Cause of rapid loss of vaccination protection in autoimmune diseases identified

People who are treated with TNF-α inhibitors for their autoimmune disease such as Crohn’s disease or rheumatoid arthritis lose their vaccination protection significantly earlier than average. The mechanism underlying the early decrease in antibody levels has now been eludicated by a scientific team from MedUni Vienna. In view of the results, principal investigator Ursula Wiedermann-Schmidt emphasises the importance of regular boosters for those affected. The research work has just been published in the specialist journal eBioMedicine.

The study was conducted by the Center for Pathophysiology, Infectiology and Immunology in cooperation with the Department of Gastroenterology and Hepatology of the University Department of Internal Medicine III. Patients with inflammatory bowel diseases (IBD) such as Crohn’s disease or ulcerative colitis and healthy controls were administered a SARS-CoV-2 mRNA vaccination and a booster after six months. Subsequent analyses showed that people receiving TNF-α blocker therapy had significantly lower antibody levels than healthy subjects and IBD patients receiving another form of treatment (α4β7-integrin antagonists).

TNF-α blockers are anti-inflammatory and immunosuppressive drugs from the group of biologics that are not only used for inflammatory bowel diseases, but also for other autoimmune diseases such as rheumatoid arthritis or psoriatic arthritis. According to the research team, the significantly faster loss of vaccine protection observed in the study is due to the fact that the strong inflammatory situation in these patients – despite the use of TNF-α inhibitors – inhibits the production of memory B cells in the lymph nodes. These are the cells of the immune system that are responsible for the production of long-lived plasma cells as well as antibodies and thus for long-term vaccine protection against already known pathogens – an essential prerequisite for the quality and duration of the protective effect of vaccinations.

Check of vaccination status with diagnosis of disease
“In our study we were able to elucidate the exact mechanism why only short-lived plasma cells are formed under TNF-α blocker therapy, so that antibody protection only lasts for a short time,” says corresponding author Ursula Wiedermann-Schmidt, Head of the Centre for Pathophysiology, Infectiology and Immunology and the Outpatient Clinic for Vaccinations, Travel and Tropical Medicine at MedUni Vienna. The study results apply not only to SARS-CoV-2 vaccinations, but in principle to all vaccinations. “For this group of patients, it is therefore necessary to maintain short-term protection through repeated booster vaccinations,” says Wiedermann-Schmidt. Special attention must be paid to vaccinations that are administered for the first time under TNF-α blocker therapy – here the early antibody waning and loss of protection can be most pronounced. Vaccinations performed already before the start of TNF-α blocker therapy would most likely retain their protective effect. In principle, when diagnosing an autoimmune disease (and other diseases under immunosuppressive therapy), the entire vaccination status should be ascertained as soon as possible and missing vaccinations should be supplemented before starting TNF-α blocker therapy (as well as other immunosuppressive therapies).

Potential treatment of autoimmune diseases revealed in the new study like multiple sclerosis and rheumatoid arthritis.

Diagram showing the results of mice that were treated with PEP


Mice that have neuroinflammation caused by autoimmunity were treated with PEP. The results found that PEP-treated mice showed improved signs of recovery compared to mice not treated with PEP. CREDI Tsung-Yen Huang (OIST)

Scientists in Japan have revealed a chemical compound that could be used for the treatment of various autoimmune diseases like multiple sclerosis and rheumatoid arthritis. These diseases occur when the body’s immune response goes wiry. The immune system, which normally attacks pathogens and infections, instead attacks healthy cells and tissues. For the millions of people who suffer from autoimmune diseases worldwide, the result can be debilitating—rheumatoid arthritis causes excessive joint pain, while multiple sclerosis can disable one’s brain and spinal cord function.

“The key to the development of autoimmune diseases, and thus the way to inhibit this development, lies in our cells, but the underlying mechanism has always been unclear,” stated Prof. Hiroki Ishikawa, who leads the Immune Signal Unit at the Okinawa Institute of Science and Technology (OIST). “Now, our recent research has shed light on a compound that could suppress the development of these diseases.”

Prof. Ishikawa went on to explain that this research, published in Cell Reports, could lead to the development of treatments for autoimmune diseases.

The research focused on T helper 17 cells, or Th17 cells. Th17 cells are a type of T cell—a group of cells, which form major parts of the immune system. These cells, which exist in high numbers in our guts, evolved to help us fight invasive pathogens but, sometimes, they’re overactivated and mistake normal, healthy tissue as pathogens, resulting in autoimmunity. The generation of Th17 cells requires glycolysis, a metabolic process in which glucose is broken down and converted to energy to support the metabolic needs of cells. Glycolysis is essential for the growth of not only Th17 cells but also a variety of cells in our body.

“What’s interesting in that excessive glycolysis seems to suppress Th17 cell activity,” stated first author, Mr. Tsung-Yen Huang, a PhD candidate in the Immune Signal Unit. “So, we hypothesized that molecules produced during glycolysis may inhibit the cells.”

Enter phosphoenolpyruvate, or PEP for short. This chemical compound is a metabolite produced when glucose is converted to energy. Since it is part of such an important process, PEP is generated every day in our bodies. The researchers found that treatment with PEP can inhibit the maturation of TH17 cells, leading to resolution of inflammatory response.

Mr. Huang explained how this was, at first, a confusing result, as it went against all other research on the topic, but he decided to persevere and take a closer look at what could be occurring.

The research led them to a protein called JunB, which is essential for the maturation of Th17 cells. JunB promotes Th17 maturation by binding to a set of specific genes. The researchers found that PEP treatment inhibits the generation of Th17 cells by blocking JunB activity.

Armed with this knowledge, the researchers went on to treat mice that had neuroinflammation caused by autoimmunity with PEP. This disease is very similar to multiple sclerosis and these mice showed positive signs of recovery. The scientists have now filed a patent to continue with this research.

“Our results show the clinical potential of PEP,” explained Mr. Huang. “But first we need to increase its efficiency.”

In the past, researchers who were interested in developing a treatment for autoimmune diseases, often looked at inhibiting glycolysis and thus Th17 cells. But glycolysis is essential to various types of cells in the body and inhibiting it could have significant side-effects. PEP has the potential to be used as a treatment without resulting in such side-effects.

Specialized garbage disposal cell, implicated in autoimmune disease, tracked for the first time.

Australian researchers have made a fundamental discovery about what happens in lymph nodes, shedding light on the causes of immune-related diseases like lupus.

2-Photon microscope image of the germinal centre inside a lymph node, showing B cells (green) moving around and a tingible body macrophage (red) grabbing the dead and dying B cells

2-Photon microscope image of the germinal centre inside a lymph node, showing B cells (green) moving around and a tingible body macrophage (red) grabbing the dead and dying B cells CREDIT Garvan

For almost 140 years, the origin and behaviour of an enigmatic cell type inside lymph nodes, called a tingible body macrophage, has remained a mystery. Now, for the first time, scientists at the Garvan Institute of Medical Research have tracked the cell’s lifecycle and function, with implications for our understanding of autoimmune disorders.

Autoimmune disease, which occurs when the immune system attacks the body, affects 5% of Australians and has a high chronic health burden worldwide, yet its causes are poorly understood.

“In living organisms, death happens all the time – and if you don’t clean up, the contents of the dead cells can trigger autoimmune diseases,” says lead author Professor Tri Phan, Head of the Intravital Microscopy and Gene Expression (IMAGE) Lab and Co-Lead of the Precision Immunology Program at Garvan.

2-Photon microscope image of the germinal centre inside a lymph node, showing tingible body macrophages (red)

3. 2-Photon microscope image of the germinal centre inside a lymph node, showing tingible body macrophages (red) CREDIT Garvan

Macrophages in many parts of the body are responsible for clearing foreign material like bacteria and viruses, but the researchers discovered that these tingible body macrophages, found inside lymph nodes, specialise in cleaning up the immune system’s own waste: the B cells that proliferate when we fight infection.

During an immune response, a massive number of B cells are made inside the lymph nodes and then tested for their ability to neutralise the infection. B cells that fail the test are destined to die, but on the way out, they can trigger the body to attack itself. The contents of these cells – especially those in the cell’s central nucleus – are inflammatory and can inadvertently activate some B cells to make antibodies against that waste, leading to autoimmunity. Removing this waste is therefore a critical housekeeping function.

The new research is published in the journal Cell.

Insights into a microscopic ecosystem

The scientists used state-of-the-art intravital imaging techniques at the ACRF INCITe Centre to observe how the macrophages form within the lymph nodes and how they behave in real time. Their analysis shows that, unlike other immune cells, tingible body macrophages do not chase their targets, but disperse evenly and lie in wait. When a dead or dying B cell comes close, the macrophage reaches out and wraps around the target, pulling it in to be ingested.

2-Photon microscope image of the germinal centre inside a lymph node, showing B cells (green) moving around and tingible body macrophages (red) evenly dispersed to grab the dead and dying cells

2-Photon microscope image of the germinal centre inside a lymph node, showing B cells (green) moving around and tingible body macrophages (red) evenly dispersed to grab the dead and dying cells CREDIT Garvan

“We know so very little about tingible body macrophages because it was not possible until now, with next-generation two-photon microscopes, to get inside the microstructures inside the lymph nodes of a living animal and watch the cells in action in real time. That’s why it’s taken 140 years – from when tingible body macrophages were first described in 1885 – to get where we are now,” says Professor Phan.

“A lot of what we do is like shooting a David Attenborough documentary but at a microscopic scale – capturing the hidden life of these rare cells ‘in the wild’, to show how these cellular ecosystems work to keep us healthy,” Abigail Grootveld, PhD student at Garvan and co-first author of the study.

“This research is exciting because it helps us to understand causes of autoimmune conditions like lupus. Understanding why somebody gets the disease in the first place and why it keeps coming back, is an important step towards future treatments for these diseases,” says Wunna Kyaw, PhD student at Garvan and co-first author of the study.

In systemic lupus, the immune system struggles to control the production of its fighter T cells and B cells. Their overactivity causes inflammation, autoantibodies and long-term damage throughout the body. This research shows that tingible body macrophages, with their B cell clean-up function, could be responsible for setting the chain of events in motion if they fail.

So far, the study has examined what happens with the macrophages in animal models of a healthy system. The researchers’ next step is to expand the experiment to an autoimmune model, to see if they can rescue the failing system and prevent autoimmunity at its root cause.

Meet VEXAS syndrome! The study offers the first glimpse of how previously unknown illness

Study offers first glimpse of how many suffer from previously unknown illness
Study offers first glimpse of how many suffer from previously unknown illness

According to a new study, about 13,200 men and another 2,300 women in the United States over age 50 are estimated to have VEXAS syndrome. Long considered a mystery illness until its genetic basis was identified in 2020, the latest findings, led by NYU Grossman School of Medicine researchers, offer the first indication of how common the illness is domestic.

Although a rare disorder, the syndrome carries a high mortality rate, with up to half of people, primarily men, dying within five years of diagnosis. The syndrome most often involves unexplained fevers and low blood oxygen levels in people diagnosed with other diseases, such as rheumatoid arthritis, lupus, and blood cancer. Some symptoms have been linked to an overactive immune system, which can cause inflammation and classify the syndrome as an autoimmune condition.

Researchers hope their findings will raise awareness of the disorder among physicians, particularly because high-dose steroids, JANUS kinase inhibitors, and bone marrow transplantation have proven effective in controlling some symptoms.

“Now that we know VEXAS syndrome is more common than many other types of rheumatologic conditions, physicians need to add this condition to their list of potential diagnoses when confronted by patients with persistent and unexplained inflammation and low blood cell counts, or anaemia,” says geneticist and study lead investigator David Beck, MD, PhD. Beck, an assistant professor in the Department of Medicine and the Department of Biochemistry and Molecular Pharmacology at NYU Langone Health, also led the federal research team initially identifying the shared UBA1 mutation among VEXAS patients.

Statistically, this corresponded to one in 4,269 American men over age 50 and one in 26,238 women over age 50 having or are likely to develop the syndrome. Researchers say this is a higher prevalence figure than many other inflammatory conditions, including vasculitis and myeloid dysplasia syndrome.

“Our study offers the first glimpse of just how common VEXAS syndrome is in the United States, particularly among men, who also happen to be the most to die from it,” says Beck, who is leading several clinical research efforts into VEXAS syndrome at NYU Langone’s Center for Human Genetics and Genomics.

Previous research, led by Beck, traced the syndrome’s origins to a mutation, or change in the letter code that makes up DNA, in the gene UBA1 (short for ubiquitin-like modifier activating enzyme 1.) The enzyme usually assists in protein breakdown.

VEXAS stands for many of its biological characteristics: vacuoles in blood cells, the E1 enzyme, X-linked, autoinflammatory, and somatic.

For the study, researchers analyzed the electronic medical records of adult patients who volunteered to participate in the Geisinger MyCode Community Health Initiative. The program has been collecting data for more than 25 years from patients in Geisinger’s 10-plus hospitals in Central and Northeastern Pennsylvania. Almost all study participants who agreed to have their blood DNA tested were white; half were over the age of 60.

Beck says the team next plans to analyze patient records in more racially diverse groups, especially among those with higher rates of rheumatologic and blood disease, to better understand who is most at risk of VEXAS syndrome. They also plan to look for additional genetic causes, test new therapies for the syndrome, and develop a simple blood test for UBA1 to make it easier to diagnose.

Evidence of autoimmunity’s origins uncovered via new approach

TCR similarities


A T cell receptor that recognizes a human protein fragment (left) is remarkably similar to one that recognizes a bacterial protein fragment (right), and to two receptors capable of recognizing both human and bacterial protein fragments (middle). A study by researchers at Washington University School of Medicine in St. Louis and colleagues at Stanford University and Oxford University supports the idea that some T cells that react to microbes also may react to normal human proteins, causing autoimmune disease. The findings promise to accelerate efforts to improve diagnostic tools and treatments for autoimmune diseases. CREDIT Xinbo Yang and K. Chris Garcia

Autoimmune diseases are thought to be the result of mistaken identity. Immune cells on patrol, armed and ready to defend the body against invading pathogens, mistake normal human cells for infected cells and turn their weapons on their own healthy tissues. In most cases, though, finding the source of the confusion — the tiny fragment of normal human protein that looks dangerously similar to a protein from a pathogen — has been challenging for scientists. That missing piece of the puzzle has hampered efforts to develop effective diagnostics and specific therapies for many autoimmune conditions.

That finally may be changing. A team involving researchers from Washington University School of Medicine in St. Louis, Stanford University School of Medicine and Oxford University has developed a way to find crucial protein fragments that drive autoimmunity, as well as the immune cells that respond to them. The findings, published Dec. 7 in Nature, open a promising pathway to diagnose and treat autoimmune diseases.

“Of all genes, the HLA genes have the greatest amount of variation across the human population. There are many, many autoimmune diseases that are associated with specific variants of the HLA genes, and in most cases we don’t know why,” said co-senior author Wayne M. Yokoyama, MD, the Sam J. Levin and Audrey Loew Levin Professor of Arthritis Research at Washington University. “This paper outlines a strategy for figuring out why certain HLA variants are linked to certain diseases. It also provides strong evidence that cross-reactivity between human and microbial proteins drives autoimmunity in at least two diseases and probably many others. Now that we understand the underlying drivers, we can start focusing on the approaches that are most likely to yield benefits for patients.”

The autoimmune diseases ankylosing spondylitis, which involves arthritis in the spine and pelvis, and acute anterior uveitis, which is characterized by inflammation in the eye, are both strongly associated with an HLA variant called HLA-B*27. The link between ankylosing spondylitis and HLA-B*27 was discovered 50 years ago — making it one of the first such associations identified between disease and HLA variants — and it remains one of the strongest known associations between any disease and an HLA variant.

The HLA family of proteins is involved in helping immune cells detect invading pathogens and distinguishing between microbial and human proteins, and is highly variable across individuals. HLA proteins function like hands that pick up fragments of whichever proteins are lying about — microbial or human — and show them to immune cells called T cells to figure out if they’re a sign of danger (microbial) or not (human).

T cells don’t recognize protein fragments by themselves; they recognize the fragment plus the hand that holds it. Scientists have long assumed that the combination of this particular hand — HLA-B*27 — plus a bit of an unknown human protein was being misidentified as dangerous in people with either of the two diseases, triggering autoimmune attacks in the eye or the spine. But for decades, they couldn’t find the fragment. Some scientists began to speculate that the misidentification hypothesis was wrong and some other reason accounted for the association between HLA-B*27 and the two diseases.

Co-corresponding author K. Christopher Garcia, PhD, and co-first author Xinbo Yang, PhD, of Stanford Medicine, along with co-corresponding authors Geraldine M. Gillespie, PhD, and Andrew J. McMichael, PhD, and co-first author Lee Garner, PhD, of Oxford University, collaborated with Yokoyama and co-first author Michael Paley, MD, PhD, of Washington University on a novel way to find the elusive fragment. The research team identified certain T cells that were abundant in the blood and joints of people with ankylosing spondylitis, and in the eyes of people with uveitis.

Garcia and Yang then devised a way to identify protein fragments that drive a T cell response when combined with HLA-B*27, and mapped the fragments against the human genome and five bacterial genomes to identify proteins from which the fragments may have originated. Using that approach, they were able to narrow down the millions of possibilities to a very short list of human and microbial proteins. Then, they determined the structures of the detector molecules — known as T cell receptors — on T cells from both groups of patients and compared them. The similarities were striking.

“This study reveals the power of studying T cell specificity and activity from the ground up; that is, identifying the T cells that are most active in a given response, followed by identifying what they respond to,” Garcia said. “Clearly these patient-derived TCRs are seeing a spectrum of common antigens, and that may be driving the autoimmunity. Proving this in humans is very difficult, but that is our future direction and could lead to therapeutics.”

The findings reveal key aspects of the biological mechanisms underlying ankylosing spondylitis, anterior uveitis and potentially many other autoimmune diseases.

“By combining recently developed technologies, we have revisited an old hypothesis that asks if the traditional antigen-presenting function of HLA-B*27 contributes to disease initiation or pathogenesis in the autoimmune conditions ankylosing spondylitis and uveitis,” Gillespie said. “Our findings that T cells at the sites of pathology recognize HLA-B*27 bound to both self and microbial antigens adds a very important layer of understanding to these complex conditions that also feature strong inflammatory signatures. Our hope is that this work will one day pave the way for more targeted therapies, not only for these conditions but ultimately, for other autoimmune diseases.”

By providing strong support for the idea that T cells that react to microbes also may react to normal human proteins, the findings promise to accelerate efforts to improve diagnostic tools and treatments for autoimmune diseases.

“For ankylosing spondylitis, the average time between initial symptoms and actual diagnosis is seven to eight years,” said Paley, an assistant professor of medicine, of ophthalmology, and of pathology & immunology. “Shortening that time with improved diagnostics could make a dramatic impact on patients’ lives, because treatment could be initiated earlier. As for therapeutics, if we could target these disease-causing T cells for elimination, we could potentially cure a patient or maybe even prevent the disease in people with the high-risk genetic variant. There’s a lot of potential for clinical benefit here.”