Biography
Professor Robert A. Harris
Professor Robert A. Harris (Bob) was born in Harpenden in Southern UK in 1966. He conducted a Bsc.Hons undergraduate degree at Portsmouth Polytechnic, majoring in Parasitology in 1987. PhD studies at University College London studying innate immune agglutinins in Schistosoma host snail species with Terry Preston and Vaughan Southgate as supervisors culminated with a thesis defence in early 1991. A 2.5 year postdoc at the London School of Hygiene & Tropical Medicine in Paul Kaye’s research group ensued, with focus on understanding the intracellular fate of Leishmania spp. protozoans in macrophages. Bob was awarded a Wellcome Trust postdoctoral fellowship that permitted his relocation to the Karolinska Institutet (Stockholm, Sweden) in the spring of 1994. A postdoc period was spent split between the labs of Anders Örn and Tomas Olsson, in which he studied Trypanosoma cruzi and Trypanosoma bruceii protozoan proteins. Bob became an Associate Professor at the Karolinska Institutet in 1999, heralding his establishment as a PI. Bob started to work with autoimmune diseases in 1996 and began study of therapy using live parasite infections or parasite molecules. His research group has developed autoantigen-specific vaccines, defined the effects of post-translational biochemical molecules on autoantigenicity and developed a macrophage adoptive transfer therapy that prevents pathogenesis in several experimental disease models. He became Professor of Immunotherapy in Neurological Diseases in 2013. In recent years research focus has centred on understanding the immunopathogenesis of incurable neurodegenerative diseases, with particular emphasis on development of immunotherapies directed at microglial cells as potential therapeutic paradigms.
Bob Harris CV July 2020
ERIK HERLENIUS GROUP
Development of autonomic control
About
Immature or deficient autonomic control is a common problem in infants born at a premature age and is of central importance in apneas, secondary hypoxic brain damage and sudden infant death syndrome.
PER ERIKSSON GROUP
Research
For better understanding of disturbances in respiratory control we study early development of cardiorespiratory control, brainstem neural networks and its associations with normal and pathological breathing. The conceptual change introduced by our recent data that endogenous prostaglandins are central pathogenic factors in respiratory disorders and the hypoxic response, open new diagnostic and therapeutic avenues that should significantly better the diagnostics and treatment of newborns and adult patients.
Inflammation is a major culprit in breathing disorders and we hypothesize that by using a newly developed urinary prostaglandin biomarker we can screen, detect and protect against inflammation related breathing disorders.
Our collaborative efforts enable us to move from a clinical problem to molecular understanding of the disease and studies are performed in patients, animal & in vitro models.
Our research is focused on the development of autonomic control with normal and paediatric patients as the target. Autonomic dysfunction in breathing and circulatory control often has its origin in neurodevelopment disorders. Furthermore, our basic research in developmental neuroscience how neural activity and stem cells form activity dependent networks is vital for the development of therapeutic interventions.
Read more
Contact: communication@cmm.se


CENTER FOR MOLECULAR MEDICINE
Published: 2023-08-15

Carmen Gerlach and Anthonie Zwijnenburg. Photo: Magdalena Lindén.
Your immune system protects you from severe diseases by fighting pathogenic germs. These battles are carried out by the different types of white blood cells, such as B and T cells. In particular, CD8 T cells are specialized in the killing of infected cells and as such are nicknamed ‘killer’ T cell. They can detect infected cells and tumor cells in our body and eliminate them. Activation lets T cells develop into so-called memory T cells that protect us long-term from severe disease upon re-infection with the pathogen. Vaccinations actively induce this process and use it to provide protection.
During an immune response, T cells with different specializations are necessary. Which of these states are formed will determine how the immune system will fight an infection for instance. Some T cells live longer, others proliferate, some make more cytokines, and others can kill better. This functional diversity is regulated by T cell differentiation. The different T cell states are also connected to the clinical outcomes of interventions such as adoptive T cell therapy, and transplantation. Tools to precisely determine T cell differentiation states are therefore of clinical relevance when T cells contribute to immune control and/or disease.
Researchers from CMM have performed a meticulous study of the transitions between CD8+ T cell differentiation states. High detailed analysis showed a gradual, not step-wise, process. However, the currently used markers did not reflect this graded nature of CD8+ T cell differentiation. The researchers found that the expression level of the chemokine receptor CX3CR1 predicts the skills of the individual T cells. Importantly, CX3CR1 expression levels identify functionally comparable T cell states in mice and humans. The resulting article was recently published in the journal Immunity.
“Human and mouse T cell immunology have been somewhat separate worlds. It was difficult to know which T cell subsets in humans corresponded to which T cell subset in mice, because although we gave the subsets the same names in both species (for example ‘effector memory’), the subsets were defined by completely different molecules. We wanted to find a common language between human and mouse T cell immunology so that it becomes possible to translate and interpret T cell properties (‘skills’) from one species to another”, says CMM Team Leader Carmen Gerlach, principal investigator and last author of the study.
The analyses revealed that graded expression of the chemokine receptor CX3CR1 mirrored the CD8+ T cell differentiation gradient and the corresponding T cell skills in a way that was comparable in humans and mice. Measuring the CX3CR1 expression gradient by flow cytometry is convenient and can be easily applied in clinical and fundamental science studies. Thus, measuring graded CX3CR1 expression provides a strategy to translate the behavior of distinct T cell differentiation states across species.
“The chemokine receptor CX3CR1 serves as an easy to use and effective measure to translate from one into the other species. Very similar to how the Rosetta Stone enabled translation between two different languages. We envision that our discovery allows for the deciphering of novel immunological mechanisms”, says Anthonie Zwijnenburg, PhD student in the Gerlach lab and first author.
First author Anthonie Zwijnenburg led the study and performed the largest part of it. Carmen Gerlach is the principal investigator. Second author Jyoti Pokharel, PhD student in the Gerlach lab, also made a substantial contribution to the study. All members of the Gerlach lab at CMM have contributed to the study in some way. CMM contributors beyond the Gerlach lab were Elena Hoffer and Liv Eidsmo from the Eidsmo lab.
The study was performed in collaboration with other researchers from Karolinska Institutet, some of which are also affiliated with Umeå University and the University of Copenhagen.
Funding was obtained from the Swedish Research Council, Ragnar Söderbergs Stiftelse, Jeansson Stiftelser, the Knut and Alice Wallenberg Foundation, Cancerfonden, Karolinska Institutet, the Marianne and Marcus Wallenberg Foundation, Region Stockholm, the Center for Innovative Medicine and Psoriasisfonden.
Publication
Zwijnenburg AJ, Pokharel J, Varnaitė R, Zheng W, Hoffer E, Shryki I, Comet NR, Ehrström M, Gredmark-Russ S, Eidsmo L, Gerlach C. Immunity. 2023 Jul 14:S1074-7613(23)00282-0. doi: 10.1016/j.immuni.2023.06.025. Online ahead of print.
Published: 2023-08-10

Jon Lampa. Photo: Karolinska Institutet.
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease which causes pain, fatigue, functional impairment, and frequently progressive joint destruction.
Early treatment is associated with improved outcome in patients with recently diagnosed RA, but the optimal firstline treatment is debated.
These are the 48-week results from the NORD-STAR study, a four-arm, investigator-initiated study in collaboration between the Nordic countries and the Netherlands.
The results show that early, aggressive treatment with the biological drugs abatacept and certolizumab pegol in the diagnosis of rheumatoid arthritis provides better clinical efficacy compared to conventional treatment reinforced with initial cortisone therapy. However, there was no difference between the arms in terms of radiological progression, which was generally very low with all four types of treatment.
The study is coordinated from Karolinska Institutet and Karolinska University Hospital. Adjunct Professor and CMM Team Leader Jon Lampa is the senior author. These results are important and may have implications for future international guidelines for the treatment of early RA.
The article was accepted on 7 June for publication in Annals of the Rheumatic Diseases.
The 24-week results were published in 2020 in BMJ (Hetland ML et al, BMJ 2020 Dec 2;371:m4328.).
Publication
Certolizumab pegol, abatacept, tocilizumab or active conventional treatment in early rheumatoid arthritis: 48-week clinical and radiographic results of the investigator-initiated randomised controlled NORD-STAR trial. Østergaard M, van Vollenhoven RF, Rudin A, et al. (see the paper for a complete list of authors), Annals of the Rheumatic Diseases Published Online First: 09 July 2023. doi: 10.1136/ard-2023-224116.
Published: 2023-08-10

Gustavo Monasterio. Photo: Private.
CMM researcher Gustavo Andrés Monasterio Ocares is the recipient of the 2023 IADR Osteology Foundation New Investigator Award in Oral Tissue Regeneration.
He officially received the award during the Opening Ceremonies of the 101st General Session of the IADR, which was held in conjunction with the 9th Meeting of the Latin American Region and the 12th World Congress on Preventive Dentistry, that took place on June 21, 2023, in Bogotá, Colombia.
Gustavo Monasterio earned his degree as a Doctor of Dental Surgery (DDS) at the University of Chile, Santiago, where he completed his degree in 2017. He then decided to pursue research and enrolled in the PhD program in Oral and Dental Sciences at the same university. He defended his PhD thesis on 2020, where, using a mouse model of periodontitis, he studied the immunogenicity and virulence of a periodontitis-associated oral pathobiont. This work was published in an international peer-reviewed immunology journal.
Gustavo Monasterio is currently leading his own research line focused in understanding the consequences of intestinal disorders on oral homeostasis. In his research line, he combines his background on oral immunology/microbiology with all the technology and tools that have been developed and optimized at the Karolinska Institute to study mechanisms of initiation and resolution of intestinal disorders. He is currently a postdoctoral fellow in the Eduardo Villablanca Group at Karolinska Institutet and CMM.
The award, amounting to $75,000 will support Gustavo Monasterio’s project titled “Investigating Regenerative Pathways in Salivary Glands to Mitigate Colitis-induced Oral Pathology”.
Read the press release from the International Association for Dental Research (IADR) here.
Published: 2023-08-01

Ingrid Kockum's research group. Top to bottom, left to right: Soumeen Jin, Thomas Moridi, Yuan Jiang, Klementy Shchetynsky, Ali Manouchehrinia, Quianwen Liu, Alexandra Gyllenberg, Ingrid Skelton Kockum, Pernilla Stridh. Photo: Magdalena Lindén
Multiple sclerosis (MS) is a neurological disease that usually occurs between the ages of 20 and 40 and is more common in women. In MS, the immune system mistakenly attacks the brain and spinal cord, resulting in symptom flares known as relapses as well as longer-term degeneration, known as progression. Despite the development of effective treatments for relapses, none can reliably prevent the accumulation of disability.
Previous studies have identified genetic factors that increase the risk of developing MS and lead to immune system dysfunction that can be treated to some extent, slowing down the disease.
But these risk factors don’t explain the large variance in disease severity.
The new genome-wide association study, led by researchers from UCSF (USA), the University of Cambridge (UK), and Karolinska Institutet (Sweden), points to a genetic variant that increases disease severity. Inheriting this genetic variant from both parents accelerated the time to needing a walking aid by almost four years.
“This is a breakthrough finding because it provides the first real progress in understanding long-term disability in MS, where patients gradually lose their mobility and independence,” says CMM Group Leader Ingrid Kockum, professor at the Department of Clinical Neuroscience, Karolinska Institutet, and principal investigator for the European part of the study. “This gives us an opportunity to develop new treatments to control progression, which is urgently needed by so many people suffering from MS.”
The variant sits between two genes called DYSF and ZNF638 with no prior connection to MS. These are normally active in the central nervous system and involved in repairing damaged cells and controlling viral infections, respectively.
“The gene variant that causes more severe MS thus appears to influence mechanisms in the brain and spinal cord, unlike genes that increase the risk of developing MS, where the immune system is central,” says CMMer Pernilla Stridh, researcher in Ingrid Kockum’s Group and coauthor of the paper. “This implies that controlling progression might require neuroprotective agents in addition to the current anti-inflammatory standard treatments.”
The researchers also found that high education was related to slower disease progression, supporting the idea that neurocognitive reserve and brain resilience play a key role in MS severity.
The work was the result of a large international collaboration of more than 70 institutions from around the world, where two large MS research consortia joined forces: The International Multiple Sclerosis Genetics Consortium (IMSGC) and The MultipleMS Consortium.
The study included more than 22,000 people with multiple sclerosis, many of which were recruited in Sweden through the National MS studies GEMS, EIMS, IMSE, and COMBAT-MS that are coordinated at Karolinska Institutet.
“One of the strengths of these studies is that they can be combined with the Swedish MS registry which contains unique data gathered by neurologists throughout Sweden,” explains Dr Stridh. “This has been crucial to being able to track disease severity over time.”
This work was supported in part by funding from EU’s Horizon 2020 (MultipleMS project, coordinated by Ingrid Kockum, Karolinska Institutet), National Institutes of Health (International Multiple Sclerosis Genetics Consortium), the National Multiple Sclerosis Society, MS Canada, deCODE Genetics/Amgen, the Swedish Research Council, Margaretha af Ugglas foundation, and the Swedish Brain Foundation. Some of the authors have received fees/funding from pharmaceutical companies, see the scientific paper for a complete list of competing interests.
Publication
Locus for severity implicates CNS resilience in progression of multiple sclerosis. International Multiple Sclerosis Genetics Consortium, MultipleMS Consortium (see the paper for a complete list of authors), Nature, online 28 June 2023, doi: 10.1038/s41586-023-06250-x.
Research Briefing: https://doi.org/10.1038/d41586-023-01787-3
Published: 2023-06-30
Atherosclerosis is a major cause of cardiovascular disease, which in turn is the most common cause of death worldwide and in Sweden.
In the late 1980's, the presence of activated T lymphocytes in human atherosclerotic plaques was discovered by Göran K Hansson and colleagues at Karolinska Institutet, leading to the conclusion that atherosclerosis is an inflammatory disease. In the 1990’s, due to a research mistake, researchers in Kevin J Tracey’s lab in New York discovered that by stimulating the vagus nerve, the production of the

Kristina Sparreljung and Peder Olofsson. Photo Kristian Pohl.
potent pro-inflammatory cytokine, TNF, could be turned down. In other words, they found that the nervous system communicates with the immune system and regulates inflammation in the body and called it “the inflammatory reflex”.
Peder Olofsson performed his PhD studies in the lab of Göran K Hansson, where he studied inflammation in atherosclerosis, and later became a researcher in Kevin J Tracey’s lab at the Feinstein Institute in New York, where he worked on understanding the mechanisms of neural control of
inflammation. With this background, Peder Olofsson has developed his research within the fields of inflammation neuroscience and bioelectronic medicine. He and his colleagues have detailed mechanisms of how peripheral nerves and neurotransmitter-releasing immune cells regulate immune responses, inflammation, and its resolution. They have also developed new methodology to selectively activate peripheral nerves, including non-invasive technology, which is already used for first-in-human investigation.
With the help of the Swedish Heart-Lung Foundation's large research grant of SEK 15 million, Peder Olofsson and co-applicant, CMMer associate professor Stephen Malin, will map how nerves and neural signals interact with blood vessels in atherosclerosis and vascular inflammation.
"Understanding inflammation and immune responses requires neuroscience. This research program will detail neural reflexes that regulate vascular inflammation, and aims to provide insights that may enable both new pharmacological and electronic approaches to treatment of excessive inflammation and atherosclerosis.”, says Peder Olofsson.
“Over two million people in Sweden live with cardiovascular disease. Peder Olofsson's research has the potential to provide groundbreaking knowledge on how we can reduce atherosclerosis and thereby give people more healthy years," says Kristina Sparreljung, Secretary General of the Swedish Heart-Lung Foundation, in a press release from the foundation.
Published: 2023-06-22

Certain immune T cells called tissue-resident memory cells are formed locally in the skin and other tissue, and protect against infections that they have encountered before. Some of them express proteins that enable them to kill infected cells. These “memory killer cells” can also contribute to the inflammatory skin disorders vitiligo and psoriasis. Recent research has shown that they are also involved in the body’s immune response to various cancers.
Varying responses to treatment
The memory killer cells have been shown to respond to immunotherapy,
a Nobel Prize-winning cancer therapy involving the tweaking/activation of the immune system. Immunotherapy is normally administered as a complement to other cancer treatments, and there is considerable
variation in how patients respond to it.
“We don’t know so much about how and why memory killer cells are formed in the skin and what it means for cancer patients,” says Professor
Yenan Bryceson at the Department of Medicine (Huddinge), Karolinska
Institutet. “Finding out how these cells develop enables us to contribute to the development of more efficacious immunotherapy for diseases like melanoma.”
On the photo (top left to bottom right): Liv Eidsmo, Yenan T. Bryceson, Wenning Zheng (second author), Beatrice Zitti (shared first author) and Elena Hoffer (shared first author). Photo: Tessa Campbell.
The study charted the development of memory killer cells in human skin, performed as a collaborative effort between Karolinska Institutet researchers Beatrice Zitti and Elena Hoffer (PhD student in Liv Eidsmo's Group at CMM). The researchers isolated T cells from the skin and blood of healthy volunteers and used advanced techniques to examine gene activity and expression of different proteins. This allowed them
to identify T cells in the blood with the potential to develop into memory killer cells in skin or other tissues. After knocking out specific genes, they could also demonstrate which genes are required for the maturation of memory killer cells in tissue.
More effective immunotherapy
The researchers then went on to study tumour samples from melanoma patients and found that those with a higher rate of survival also had a larger accumulation of epidermal memory killer cells.
“We’ve been able to identify several factors that control the formation of memory killer cells, which play an important part in maintaining a healthy skin,” says Liv Eidsmo, CMM Group Leader, dermatologist and professor at the University of Copenhagen in Denmark and researcher at Karolinska Institutet in Sweden, who led the study with Professor Bryceson. “There’s a fine balance between effective protection against tumours and infections in the skin and contribution to inflammatory diseases like vitiligo and psoriasis.”
The researchers now aim to harness their findings to optimise the immunotherapy-induced T-cell response to make it even better at eliminating cancer cells in tissues.
The study was conducted in collaboration with the Karolinska University Hospital, Nordiska Kliniken and Vrinnevi Hospital. It was financed by grants from Novartis, the EU (Marie Skłodowska-Curie Actions), KI Foundations and Funds, the Swedish Research Council, the Ragnar Söderberg Foundation, the Swedish Medical Society, Region Stockholm (ALF scheme), the Swedish Psoriasis Foundation, the Swedish Dermatology Foundation, the Swedish Cancer Society, the Göran Gustafsson Foundation, Stockholm City Council, the Karolinska Institutet Centre for Innovative Medicine (CIMED) and the Knut and Alice Wallenberg Foundation. There are no reported conflicts of interest.
This text is based on an article from the Karolinska Institutet website, published by Felicia Lindberg.
Publication
Human skin-resident CD8+ T cells require RUNX2 and RUNX3 for induction of cytotoxicity and expression of the integrin CD49a. Beatrice Zitti, Elena Hoffer, Wenning Zheng, Ram Vinay Pandey, Heinrich Schlums, Giovanna Perinetti Casoni, Irene Fusi, Lien Nguyen, Jaanika Kärner, Efthymia Kokkinou, Anna Carrasco, Jessica Gahm, Marcus Ehrström, Staffan Happaniemi, Åsa V. Keita, Charlotte R.H. Hedin, Jenny Mjösberg, Liv Eidsmo, Yenan T. Bryceson. Immunity, online 2 June 2023, doi: 10.1016/j.immuni.2023.05.003.
CMMer PhD student Elena Hoffer on the published study:
What do you think is most exciting about the findings in the paper?
Understanding how the CD49 positive T cells actually develop opens up the possibility of affecting this cell population that is correlated with a better survival in melanoma patients. I hope we can use these insights in the development of a new therapy in the future.
What in the working process has been most valuable for you as a PhD student?
For me, the most amazing part has been to see how the collaboration made science much more impacting. The authors of the paper were placed in several different countries and despite the distances and other challenges, I think we have really managed to make most of it. Science is much better and more fun when we collaborate. Nothing is a one-man show. That has been the lesson for me. Finally, I want to say that I enjoyed working on this project. I want to give particular credit to Beatrice Zitti who is a great planner and organizer and she has started this study 5 years ago and led it in an outstanding way.
Published: 2023-05-24

CMMer Göran K Hansson, professor of experimental cardiovascular research at the Department of Medicine, Solna, is awarded the Grand Silver Medal for his far-reaching contributions to cardiovascular medicine, Karolinska Institutet and the national and international discourse in medicine.
For three decades, Göran K Hansson has developed the now highly active research area of atherosclerosis immunology. He has made several of the seminal contributions that established atherosclerosis as an inflammatory disease – insights that have created a plethora of new possibilities for improved clinical protocols in cardiovascular prevention and therapy.
In addition to his leadership in cardiovascular medicine, Göran K Hansson has served on several boards and committees of Karolinska Institutet, including the Board for research (1995-2005), and he represented KI on the board of the Karolinska Hospital (2002-2003). He was one of the founding PIs of the Center for Molecular Medicine and he led the Linnaeus Centre for Research on Inflammation and Cardiovascular Disease, CERIC, from its start in 2008.
Olivia Thomas and Mattias Bronge, shared first authors of the study.
Photos: Erik Holmgren.
Göran K Hansson has also had central roles in the Nobel system for nearly 25 years. He served as the secretary general of the Royal Swedish Academy of Sciences during 2015-2021, and he served on the board of directors of the Nobel Foundation for 13 years (2009- 2022), also as its vice chair.
In his positions at the Karolinska Institutet and beyond, Göran K Hansson has taken a prominent role in promoting science in society, including commentaries and editorials in the daily press, participating in debates and news programmes, maintaining an active blog about science, and appointing an independent expert panel at the Royal Swedish Academy of Sciences during the Covid-19 pandemic. He remains an exceptionally respected voice promoting reason and science in society.
This text is based on a press release from Karolinska Institutet where you can read more about the 2023 awardees and about Karolinska Institutet's medals.
Published: 2023-05-19


Olivia Thomas and Mattias Bronge, shared first authors of the study.
Photos: Erik Holmgren.
A study published in Science Advances by researchers at CMM, further explains how the Epstein-Barr virus can trigger multiple sclerosis or drive disease progression. The results show that some individuals have antibodies against the virus that mistakenly attack a protein expressed in the central nervous system.
Epstein-Barr virus (EBV) infects most people early in life and then remains in the body, usually without causing symptoms. The link between EBV and the neurological disease multiple sclerosis (MS) was discovered many years ago and has puzzled researchers ever since. Increasing evidence, including two papers published in Science and Nature last year, suggests that EBV infection precedes MS and that antibodies against the virus may be involved. However, the molecular mechanisms seem to vary between patients and remain largely unknown.
“MS is an incredibly complex disease, but our study provides an important piece in the puzzle and could explain why some people develop the disease,” says the shared first author of the paper Olivia Thomas, postdoctoral researcher in the Group of Hans Grönlund at CMM and at the Department of Clinical Neuroscience, Karolinska Institutet. “We have discovered that certain antibodies against the Epstein-Barr virus, which would normally fight the infection, can mistakenly target the brain and spinal cord and cause damage.”
Misdirected antibodies
The researchers analysed blood samples from more than 700 patients with MS and 700 healthy individuals. They found that antibodies that bind to a certain protein in the Epstein-Barr virus, EBNA1, can also bind to a similar protein in the central nervous system called CRYAB. The protein is specifically expressed under conditions of cellular stress, eg. inflammation, where its role as a chaperone protein is to prevent protein aggregation. It is also expressed in many other tissues in the body. The misdirected, cross-reactive antibodies may damage the nervous system and cause severe symptoms in MS patients, including problems with balance, mobility and fatigue. The antibodies were present in about 23 percent of MS patients and 7 percent of control individuals.
“This shows that, whilst these antibody responses are not required for disease development, they may be involved in disease in up to a quarter of MS patients,” says Olivia Thomas. “This also demonstrates the high variation between patients, highlighting the need for personalised therapies. Current therapies are effective at reducing relapses in MS but unfortunately, none can prevent disease progression.”
T cells may also be involved
The researchers also found that there is likely a similar cross-reactivity among T cells of the immune system.
“We are now expanding our research to investigate how T cells fight EBV infection and how these immune cells may damage the nervous system in multiple sclerosis and contribute to disease progression,” says Mattias Bronge, affiliated researcher at the Department of Clinical Neuroscience, Karolinska Institutet and shared first author of the paper.
The study was financed by Sweden’s innovation agency Vinnova, the Swedish Research Council, the Swedish Brain Foundation, Karolinska Institutet, MS Forskningsfonden, Neuro, and Region Stockholm. Co-author Hans Grönlund is the inventor of a current patent filed by NEOGAP Therapeutics AB and is the founder and co-owner of this company. Birce Akpinar, Ola B. Nilsson, Erik Holmgren and Guro Gafvelin hold positions at NEOGAP Therapeutics AB. Roland Martin is a co-founder, co-owner and an employee of Cellerys, a spin-out from the University of Zürich, and is a coinventor and patent holder for several patents. Roland Martin and Tomas Olsson have received grants and fees from several companies. See the scientific paper for a complete list of the authors’ potential conflicts of interest.
This text is based on an article from the Karolinska Institutet website.
Epstein-Barr virus
The herpesvirus EBV is one of the most widespread viruses in humans. Over 90 percent of the world’s population are infected with EBV and carry the virus for life as a latent, usually asymptomatic, infection. Most people are infected as children with few or no symptoms but in young adults, the virus often causes infectious mononucleosis, also known as glandular fever or kissing disease.
Publication:
“Cross-reactive EBNA1 immunity targets alpha-crystallin B and is associated with multiple sclerosis”. Olivia G. Thomas, Mattias Bronge, Katarina Tengvall, Birce Akpinar, Ola B. Nilsson, Erik Holmgren, Tara Hessa, Guro Gafvelin, Mohsen Khademi, Lars Alfredsson, Roland Martin, André Ortlieb Guerreiro-Cacais, Hans Grönlund, Tomas Olsson, Ingrid Kockum. Science Advances, online 17 May 2023, doi: 10.1126/sciadv.adg3032. https://doi.org/10.1126/sciadv.adg3032
Published: 2023-04-05
Patients who experience serious cardiovascular events, such as heart failure, often have impaired blood vessel function. Now, researchers from CMM have discovered that certain T cells in human blood can produce acetylcholine, which may help regulate blood pressure and inflammation. The study, which is published in PNAS, also demonstrates a possible association between these immune cells in seriously ill patients and the risk of death.
It is known that acetylcholine regulates blood flow in mice, and that it is produced by T cells. In humans, the source of blood acetylcholine has been unclear. The new study, based on analyses of blood from healthy blood donors, now shows that human T cells can also release acetylcholine. The type of immune cells are known as ChAT+ T cells.
“This corroborates previous findings in different model systems and may contribute to the development of treatments for cardiovascular disease and inflammatory diseases,” says shared first author Laura Tarnawski in a press release from Karolinska Institutet.

Vladimir Shavva
Photo: Patricia Mendes
Laura Tarnawski
Photo: Private
Peder S. Olofsson
Photo: MedTechLabs
The research group, headed by Peder S. Olofsson who is the principal investigator in this study, focuses on the communication between the brain and the immune system. Acetylcholine is a neurotransmitter with important functions in the brain and neuromuscular system and this study sheds light on its role in the human immune system.
“Our new study shows that acetylcholine in the blood can be secreted by immune cells, which can regulate inflammation in the blood vessels,” says Vladimir Shavva, the other first author of the article published in PNAS.
Moreover, by studying 33 patients with severe circulatory failure who had been admitted for intensive care, the researchers found that higher relative blood levels of ChAT+ T cells were associated with reduced risk of death. The findings encourage further investigation of ChAT+ T cells in different groups of patients. This could contribute to new diagnostic and therapeutic opportunities for seriously ill patients with excessive inflammation.
The study was financed by the Swedish Research Council, the Swedish Heart Lung Foundation, the Knut and Alice Wallenberg Foundation, MedTechLabs, the ALF project scheme, the Lars Hierta Memorial Foundation, the Gösta Fraenckel Foundation, the Loo and Hans Osterman Foundation and the Foundation for Geriatric Diseases at Karolinska Institutet. Peder Olofsson is a shareholder of Emune AB. Co-author Michael Eberhardson has received lecture and consultancy fees from AbbVie, Merck (MSD), Takeda, Ferring, Orion Pharma, Otsuka, Tillotts, ITH, Novartis, Pfizer, Bristol Myers Squibb and Janssen and research grants from AbbVie and MSD. All other authors report no conflicts of interest.
This text is based on a press release from Karolinska Institutet which you can read here.
Publication
”Cholinergic regulation of vascular endothelial function by human ChAT+ T cells”. Tarnawski L, Shavva VS, Kort EJ, Zhuge Z, Nilsson I, Gallina AL, Martínez-Enguita D, Heller Sahlgren B, Weiland M, Caravaca AS, Schmidt S, Chen P, Abbas K, Wang FH, Ahmed O,Eberhardson M, Färnert A, Weitzberg E, Gustafsson M, Kehr J, Malin SG, Hult H, Carlström M, Jovinge S, Olofsson PS. PNAS (Proceedings of the National Academy of Sciences), online 29 March 2023, doi: 10.1073/pnas.2212476120.
Published: 2023-03-22

Letian Zhang and Ning Xu Landén. Photos: Private
Researchers at CMM have identified HOXC13-AS, a human skin-specific long noncoding RNA (lncRNA), that plays a crucial role in epidermal differentiation. The study is published in the journal Cell Death & Differentiation, highlighting lncRNA's physiological importance in the epidermal barrier's maintenance and reconstruction.
barrier's maintenance and reconstruction.
When the skin gets injured, the epidermal keratinocytes switch their states from homeostasis to regeneration to reconstruct the epidermal barrier. However, the dynamic gene expression and regulatory mechanisms underpinning this switch are enigmatic. Such knowledge is required to understand the pathological mechanism underlying failed re-epithelization in chronic non-healing wounds. Although not coding for proteins, many lncRNAs have been identified as important regulators in fundamental biological processes, but their role in epidermal differentiation remains largely unexplored.
In a new study published in Cell Death & Differentiation (03 March 2023), researchers from CMM and Karolinska Institutet performed RNA sequencing in human skin and wounds and identified a lncRNA, HOXC13-AS, specifically expressed in human skin and its expression was significantly downregulated when the skin gets injured. The study reveals that HOXC13-AS promotes keratinocyte differentiation, thus crucial for the epidermal barrier function. Mechanistically, HOXC13-AS interacts with COPA (COPI Coat Complex Subunit Alpha) proteins that participate in the retrograde transport of cargo proteins from the Golgi to the endoplasmic reticulum (ER).
"Our study demonstrates that the interaction between HOXC13-AS and COPA proteins interferes with the retrograde transport, which leads to ER stress and keratinocyte differentiation", says the first author, Dr. Letian Zhang, at CMM and the Department of Medicine Solna, who successfully defended his Ph.D. recently.
"The specific expression in human skin and the critical function in regulating ER stress make HOXC13-AS a potential therapeutic target for a range of cutaneous diseases showing chronic ER stress," says Ning Xu Landén, CMM Group Leader and Senior Lecturer at the Department of Medicine, Solna, who is the PI leading this study.
The study was supported by:
Swedish Research Council
Ragnar Söderbergs Foundation
Welander and Finsens Foundation (Hudfonden)
LEO foundation
Cancerfonden
Ming Wai Lau Centre for Reparative Medicine
Karolinska Institute
Open access funding was provided by Karolinska Institutet.
This text is based on an article published on the Karolinska Institutet website.
Human skin specific long noncoding RNA HOXC13-AS regulates epidermal differentiation by interfering with Golgi-ER retrograde transport. Letian Zhang, Minna Piiponen, Zhuang Liu, Dongquing Li, Xiaowei Bian, Guanglin Niu, Jennifer Geara, Maria A. Toma, Pehr Sommar, Ning Xu Landén. Cell Death Differ (2023). https://doi.org/10.1038/s41418-023-01142-z
Published: 2023-03-16
Thirteen researchers from CMM have received funding in the latest call from Reumatikerförbundet, the Swedish Rheumatism Association. The awarded research aims at increasing the knowledge about different aspects of rheumatic diseases.
Vivianne Malmström receives SEK 200 000 during two years for the project: Lymfocyters betydelse vid inflammatorisk reumatisk sjukdom.
Per-Johan Jakobsson receives SEK 250 000 during two years for the project: Tidig diagnostik och tidig behandling vid reumatisk sjukdom.
Lina Marcela Diaz Gallo Norlén receives SEK 115 000 for the project: Identifiering av homogena undergrupper av patienter med systemiska autoimmuna sjukdomar genom att integrera flera datakällor för att bidra till utvecklingen av personlig medicin.

Photo: Unsplash
Bence Rethi receives SEK 140 000 for the project: Autoantikroppar och utveckling av reumatoid artrit – molekylära mekanismer och nya behandlingsmöjligheter.
Jon Lampa receives SEK 150 000 for the project: Kvarstående smärta och trötthet trots antireumatisk behandling, en epidemiologisk och mekanistisk kartläggning vid tidig RA.
Karin Lundberg receives SEK 150 000 for the project: Om uppkomst och funktion av autoantikroppar i reumatoid artrit – studier av en möjlig koppling till mikrober.
Karine Chemin receives SEK 175 000 for the project: T-cells-signaturer vid reumatisk sjukdom, ett steg mot precisionsmedicin.
Caroline Grönwall receives SEK 205 000 for the project: Effekt av autoantikroppar och autoreaktiva B-celler i reumatiska sjukdomar.
Helena Erlandsson Harris receives SEK 275 000 for the project: Att förstå orsakerna till ledgångsreumatism hos barn.
Ioannis Parodis receives SEK 230 000 for the project: Prediktion av skov, behandlingssvar och långtidsutfall vid systemisk lupus erythematosus (PREDiCT-SLE).
Lars Alfredsson receives SEK 350 000 for the project: Betydelsen påverkbara levnadsvanor/arbetsmiljöfaktorer för insjuknande och förlopp av reumatoid artrit.
Ingrid Lundberg receives SEK 350 000 for the project: Sjukdomsmekanismer och behandlingseffekter vid kronisk reumatisk muskelinflammation, myosit.
Marie Wahren-Herlenius receives SEK 350 000 for the project: Sjögrens syndrom – kliniska och immunologiska studier.
The complete list of research projects is found on the Reumatikerförbundet homepage.
Read the article from Karolinska Institutet here (in Swedish).
Published: 2023-03-14

Sebastian Ols. Photo: Daryl Boey
Sebastian Ols, currently postdoctoral researcher in Karin Loré’s CMM Group, is one of the recipients of the Sven Gard Scholarship 2023. The prize is given yearly for the best PhD theses at Karolinska Institutet in the field of virology.
Sebastian Ols has received the 2023 prize for his dissertation ”Cellular and molecular mechanisms for induction of broad anti-viral B cell responses through vaccination.” The thesis focused on the characterization of immune responses by B-cells against viruses, elicited by vaccination, and the mechanisms underlying the induction of antibody responses of broad reactivity.
The Sven Gard Scholarship was established in 2004. Part of the funds in Karolinska Institutet's foundation for virus research (which was founded by professor Sven Gard) would be used to pay attention to the past year's best thesis in virology. The purpose of the foundation is to promote virus research at Karolinska Institutet, primarily that conducted by junior, unpromoted researchers.
The reviewers in the foundation consider Sebastian Ols’ thesis to provide improved knowledge on how antiviral B-cell responses can be modulated by the selection of vaccine platforms and they found the thesis is comprehensive and well written.
This text is based on an article published on the Karolinska Institutet website.
Published: 2023-03-16
The first detailed description of the microbiota and immune cells among asymptomatic Helicobacter pylori-infected individuals has been published in the journal JCI Insight. The study was led by Eduardo Villablanca, Group Leader at CMM and at the Department of Medicine, Solna, together with Juan Du, docent at the Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet.
Around half of the world’s population is infected by the Helicobacter Pylori (H. Pylori) bacterium. Only some of the infected individuals develop symptoms and related conditions such as gastric ulcers, whereas a majority are asymptomatic. This last group, however, seems to have a higher risk to develop cancer.
Eduardo Villablanca, Juan Du and their research teams wanted to analyze the gastric microbiome and immune cell composition in asymptomatic H.pylori-infected individuals in order to get a more detailed insight into the microenvironment in the asymptomatic stomach.

Eduardo Villablanca.
Photo: Patricia Torregosa
Juan Du.
Photo: John Sennett
Using single cell RNA sequencing and flow cytometry the researchers showed how the gastric microenvironment, which undergoes deep microbial and immunological changes during chronic H. pylori infection.
The study has increased the knowledge of stomach physiology, with respect to the gastric microbiome and identified potential bacterial function and effects on the host immunity in H. pylori-infected asymptomatic individuals. The data will be a valuable resource to interrogate pathology of gastric diseases, both for research and clinical setting, to follow H. pylori infected asymptomatic individuals
The study was financed by the Swedish Foundation for Strategic Research, Svenska Läkaresällskapet, Karolinska Institutet, Erling-Persson Foundation, Swedish Research Council, Formas, Cancerfonden and Wallenberg Academy Fellow program.
Read more in the news article published on the Karolinska Institutet webpage.
Publication
Chiara Sorini, Kumar P. Tripathi, Shengru Wu, Shawn M. Higdon, Jing Wang, Liqin Cheng, Sanghita Banerjee, Annika Reinhardt, Taras Kreslavsky, Anders Thorell, Lars Engstrand, Juan Du, and Eduardo J. Villablanca. Metagenomic and single-cell RNA-Seq survey of the Helicobacter pylori–infected stomach in asymptomatic individuals. JCI Insight. 2023;8(4):e161042. https://doi.org/10.1172/jci.insight.161042.
CS, KPT, and SW contributed equally to this work. JD and EJV contributed equally to this work.
Published: 2023-02-21
Researchers from CMM have identified T cells that contribute to the destruction of kidney tissue in patients with ANCA-associated vasculitis (AAV). The finding suggests the possibility of T cell screening to identify individuals at high risk of disease relapse despite treatment, a problem affecting many patients with AAV.
Ravi Kumar, postdoctoral researcher in Vivianne Malmström’s Group at CMM and at the Division of Rheumatology, Department of Medicine Solna, is the first author of the study that was recently published in the journal Kidney International.
In an article published on the Karolinska Institutet website,
Ravi Kumar answered some questions about the new findings.

Co-authors (from left to right): Bruno Raposo, Marcelo Afonso, Iva Gunnarsson, Ravi Kumar, Vivianne Malmström and Sanjay Boddul.
Publication
Identification of proteinase 3 autoreactive CD4+T cells and their T-cell receptor repertoires in anti-neutrophil cytoplasmic antibodies associated vasculitis.
Ravi Kumar Sharma, Niyaz Yoosuf, Marcelo Afonso, Andrea Scheffschick, Aune Avik, Alice Bartoletti, Begum Horuluoglu, Juan Sebastian Diaz Boada, Sanjay Kumar Boddul, Asta Dögg Jonasdottir, Björn Lövström, Hanna Brauner, Bruno Raposo, Karine Chemin, Annette Bruchfeld, Iva Gunnarsson, Vivianne Malmström.
Kidney International, online 15 Feb 2023. DOI: https://doi.org/10.1016/j.kint.2023.01.023.
Published: 2022-12-06

Researchers at CMM have shown that B cells, which are critical to the proper functioning of the immune system, can sometimes do more harm than good. In a publication in Immunity, Eduardo Villablanca and his research group show that the numbers of B cells increase greatly after bowel damage and prevent the tissue from healing. The results can be of signific-ance to the treatment of inflammatory bowel disease.
The number of B cells greatly increases after bowel damage, preventing the interaction between stromal and epithelial cells which is needed for the tissue to heal. Illustration: Villablanca lab
B cells are a type of white blood cell that have an important function in the immune system, in part by producing the antibodies that attack bacteria and viruses. Previous research has shown that people with chronic inflammatory bowel disease (IBD), such as Crohn’s disease or ulcerative colitis, have many more B cells in their intestines than healthy individuals. It has therefore been proposed that B cells might affect the severity of these diseases. Researchers at CMM have now tried to discover if, and if so how, B cells contribute to IBD.
Sharp increase during healing
“We’ve been able to show that the B cell population increases sharply in the colon during the healing of colonic lesions, and that these cells mainly accumulate in areas where the damage is severe,” says principal investigator Eduardo Villablanca, associate professor at the Department of Medicine (Solna), Karolinska Institutet and CMM Group Leader. “This prevents, in turn, the interaction between two other cell types – stromal and epithelial cells – which is needed for the tissue to heal.”
The researchers studied an experimental model of colitis and tissue from patients with ulcerative colitis, using a range of methods to analyse cell populations. Focusing particularly on how B cells affect healing in the intestinal mucosa, they found that mice lacking B cells recovered much more quickly after bowel damage than regular mice. The finding that the B cells seem to do more harm than good in colonic inflammation can be of significance to the treatment of IBD.
“We’ve been able to show that the B cell population increases sharply in the colon during the healing of colonic lesions, and that these cells mainly accumulate in areas where the damage is severe,” says principal investigator Eduardo Villablanca, associate professor at the Department of Medicine (Solna), Karolinska Institutet and CMM Group Leader. “This prevents, in turn, the interaction between two other cell types – stromal and epithelial cells – which is needed for the tissue to heal.”
The researchers studied an experimental model of colitis and tissue from patients with ulcerative colitis, using a range of methods to analyse cell populations. Focusing particularly on how B cells affect healing in the intestinal mucosa, they found that mice lacking B cells recovered much more quickly after bowel damage than regular mice. The finding that the B cells seem to do more harm than good in colonic inflammation can be of significance to the treatment of IBD.
Drugs that affect B cells
“There are already approved drugs that affect the B cell response and that are used for other diseases,” says Gustavo Monasterio, postdoc Eduardo Villablanca’s Group at CMM and one of the leading authors. “We now want to test if depleting B cells at specific time windows could also work with IBD. We also need to find out if the accumulation of B cells can have a long-term beneficial effect, such as by changing the composition of bacteria in the gastrointestinal tract.”
The study was supported by grants from the Swedish Research Council, the Swedish Cancer Society, the Knut and Alice Wallenberg Foundation (the Wallenberg Academy Fellow programme) and the German research foundation DFG. Eduardo Villablanca has received research grants from the pharmaceutical company F. Hoffmann-La Roche and co-author Camilla Engblom is scientific consultant for the biotech company 10X Genomics Inc. Julio Saez-Rodriguez receives funding from Glaxo Smith Kline and Sanofi and consultancy fees from Travere Therapeutics.
This text is based on an article from Karolinska Institutet's website.

Villablanca's research group with Eduardo Villablanca in the middle. Photo: Eduardo Villablanca.
Publication
”B cell expansion hinders the stroma-epithelium regenerative crosstalk during mucosal healing”. Annika Frede, Paulo Czarnewski, Gustavo Monasterio, Kumar P. Tripathi, David A Bejarano, Ricardo O. Ramirez Flores, Chiara Sorini, Ludvig Larsson, Xinxin Luo, Laura Geerlings, Claudio Novella-Rausell, Chiara Zagami, Raoul Kuiper, Rodrigo A Morales, Francisca Castillo, Matthew Hunt, Livia Lacerda Mariano, Yue O. O. Hu, Camilla Engblom, Ana-Maria Lennon-Dumenil, Romy Mittenzwei, Nadine Hövelmeyer, Joakim Lundeberg, Julio Saez-Rodriguez, Andreas Schlitzer, Srustidhar Das, Eduardo J. Villablanca. Immunity, online 2 December 2022, doi: 10.1016/j.immuni.2022.11.002.
Published: 2022-12-06

Hildur Arnardottir. Photo: Stefan Zimmermann.
Sweden’s innovation agency, Vinnova, awarded SEK 4.5 million to CMMer Hildur Arnardottir for a project aimed at personalized cardiovascular prevention using omega-3 fatty acids.
Hildur Arnardottir at the Department of Medicine, Solna and Magnus Bäck's CMM Group is awarded SEK 4.5 million from Vinnova in the European call ERA PerMed 2022: Personalised Prevention.
The title of the project is "Optimizing omega-3 supplementation to resolve inflammation in a personalized medicine cardiovascular disease prevention".
Cardiovascular diseases are the most common cause of death. Opti-mizing prevention is therefore of utmost importance, but personalized medicine has not yet reached clinical practice for cardiovascular diseases. Omega-3 fatty acids are traditionally considered to protect against cardiovascular disease but clinical trials have shown contra-dictory results. The aim of this project is to create a personalized decision-making tool to determine whether omega-3 treatment will optimally achieve cardiovascular health for individual patients.
This text is based on an article from Karolinska Institutet's website.
Published: 2022-12-06
Eduardo Villablanca, Group Leader at CMM, has received a FORMAS grant of SEK 3 million. He and his team will study the immunotoxic effects of PFAS in the intestinal mucosal immune responses.
Eduardo Villablanca research group will study the immunotoxic effects of PFAS, which is a pollutant highly present in the environment, also in Sweden, and it is almost impossible to get rid of.
PFAS exposure has been associate with many autoimmune disorders including inflammatory bowel diseases (IBD). In addition, PFAS has recently been associated with impaired response to COVID vaccines. In this Formas project, they will investigate the effect of PFAS in intestinal mucosal immune responses and study the responses to vaccines.

“This grant means that we for the first time will be able to study the effect of this pollutants in mucosal immune responses and vaccines, which would not be possible without FORMAS support. This project would give us the chance to transition our research into sustainability issues and find the causal links between exposure to pollutants and autoimmune diseases”, says Eduardo Villablanca.
More information:
Formas is a government research council for sustainable development. They fund research and innovation, develop strategies, perform analyses and conduct evaluations. Their areas of activity include the environment, agricultural sciences and spatial planning. Formas conduct evidence syntheses analyses that aim to make it easier for Sweden to achieve our environmental objectives.
This text is based on an article from Karolinska Institutet's website.
Eduardo Villablanca. Photo: Magnus Bergström.
Published: 2022-11-22

Ning Xu Landén. Photo: Ulf Sirborn
CMM Group Leader Ning Xu Landén has been granted DKK 4.2 million by the LEO Foundation for research on wound healing.
The project seeks to improve wound healing by identifying key regulators of cellular transition from inflammation to proliferation, a cardinal event during normal skin wound healing which is lacking in chronic wounds.
Ning Xu Landén and her research group will approach this by mapping the spatiotemporal changes, both genetic, molecular and cellular, happening during the healing of acute wounds. Using this mapping, they will then aim to identify the core genetic changes and intercellular crosstalk which regulates the inflammation to proliferation transition. Once identified, these changes and intercellular crosstalk will be characterized in more detail.
The goal is to identify the “master” regulators of inflammation-to-proliferation transition in order to improve and accelerate wound healing and thus minimize the risk of development of chronic wounds. If successful, this project could pave the way for a novel approach to wound healing which may also eventually reduce subsequent scarring.
This text is based on an article from Karolinska Institutet and a press release from LEO Foundation.
Published: 2022-11-17

Top left to bottom right: Richard Rosenquist Brandell (Photo: Rick Guidotti), Eduardo Villablanca (Photo: Magnus Bergström), Per-Johan Jakobsson (Photo: Karolinska Institutet), Emma Tham (Photo: Private).
The Swedish Cancer Society (Cancerfonden) has announced the 2022 grants supporting cancer research in Sweden. Four researchers from CMM received funding.
Richard Rosenquist Brandell received SEK 6,000,000 for the project “Unlocking the complex molecular landscape in chronic lymphocytic leukemia: clues towards precision medicine”. In addition, Richard Rosenquist Brandell received SEK 600,000 in support of the Swedish CLL Group, an interdisciplinary working group for clinicians and researchers focusing on chronic lymphocytic leukemia, the most common adult leukemia.
Eduardo Villablanca received SEK 3,000,000 for the project titled “Functional characterization of mechanisms promoting intestinal regeneration but tumorigenesis”.
Per-Johan Jakobsson received SEK 2,400,000 for the project titled “Stromal-cancer cell interplay and immunomodulatory functions of prostaglandin E2 in the tumor microenvironment, a novel target for therapeutic intervention”.
Emma Tham received funding in the category of “Research months” consisting of three paid months per year during three years for working on the project titled “Cancer genetics for improved diagnosis and personalised follow-up”.
Published: 2022-11-09

Fourteen researchers from CMM have received funding from the Swedish Research Council (Vetenskapsrådet) in the last call. The grants were announced on the 3rd of November. In total 143 resarchers from Karolinska Institutet were awarded grants from the Swedish Research Council in this call.
Here follows a list of CMM researchers, the amount of the grant and the Swedish titles of their awarded projects, within the two categories Medicine and Health and Natural sciences and technical sciences, respectively.
Photo: Pixabay
Medicine and Health
Kristina Broliden is awarded a total of SEK 3,000,000 for three years for the project “Genital candidainfektion som en riskfaktor för HIV infektion – identifiering av molekylära mekanismer”.
Giedre Grigelioniene is awarded a total of SEK 3,000,000 for two years for the project “Kartläggining av det molekylära landskapet vid medfödda skelettsjukdomar”.
Johan Grunewald is awarded a total of SEK 4,800,000 for four years “Kartläggning av etiologin vid sarkoidos kan förhindra sjukdomsutveckling med kronisk inflammation och fibros”.
Maja Jagodic is awarded a total of SEK 4,800,000 for four years for the project “Epigenetik i multipel skleros: mot en bättre förståelse av patogena mekanismer, skräddarsydda behandlingar och sjukdomshantering”.
Catharina Lavebratt is awarded a total of SEK 3,600,000 for three years for the project “Blodkärlsinflammation vid medicinering av ADHD hos barn?”.
Vivianne Malmström is awarded a total of SEK 3,000,000 for three years for the project “Autoreaktiva B- och T-celler i reumatisk sjukdom – immunologisk uppföljning och intervention”.
Outimaja Mäkitie is awarded a total of SEK 6,000,000 for four years for the project “Genetiska orsaker och bakomliggande mekanismer i tidigt debuterande osteoporos”.
Tomas Olsson is awarded a total of SEK 2,400,000 for three years for the project “Orsaker och patogenes vid multipel skleros, prevention och precis terapi”.
Onur Parlak is awarded a total of SEK 6,000,000 for four years for the project “Epidermal bioelektronik för infektionsövervakning i mänskliga hudsår”.
Martin Schalling is awarded a total of SEK 4,800,000 for four years for the project “Bipolär sjukdom – translationella och prospektiva studier med fokus på litiumbehandling”.
Cecilia Söderberg-Naucler is awarded a total of SEK 2,400,000 for three years for the project “Studier av immunometabolism för identifiering av nya behandlingsmöjligheter för kroniska inflammatoriska sjukdomar”.
Jenny Löfgren is awarded a total of SEK 5,550,000 for three years in support for a halftime-employment as researcher in a clinical environment. The research focus is within the theme of “Global Surgery in Stockholm”.
Natural sciences and technical sciences
Vladana Vukojevic is awarded a total of SEK 3,568,000 for four years for the project “Kvantitativ avbildning av snabba dynamiska processer via massivt parallell fluorescens korrelationsspektroskopi”.
Published: 2022-10-25

Roland Martin
Photo:University of Zurich
Professor Roland Martin is leaving a permanent position as Head of the MS Center at the University Hospital Zurich to continue his groundbreaking research at the Department of clinical neuroscience, and hosted by research groups at CMM.
The presence of Roland Martin in the MS consortium will mark the coming direction with intention to cure MS by precise, personalized diagnostics and immunotherapy says Hans Grönlund, CMM Group Leader and research group leader of Therapeutic immune design Unit, who will host Roland Martin. Professor Martin is a world leading expert on the role of the adaptive autoimmunity in MS, and his willingness to join our teams is also a sign of our international competitiveness worldwide, says CMM Group Leader Tomas Olsson, professor at the Neuroimmunology Unit, department of Clinical Neuroscience.
In the near future, the aim is to understand better the antigen specificities of autoreactive T cells in MS and other diseases with techniques developed by Roland Martin and coworkers. Furthermore, collaborative work to prepare for a phase 2 clinical trial aiming to cure MS by correcting pathogenic T cell responses is soon to start.
This text is based on a news article from Karolinska Institutet.
Published: 2022-10-14




Marie Wahren-Herlenius
Photo: Saga Rebecka Herlenius
Olle Kämpe
Photo: Sara Öster
Björn Reinius
Photo: Ulf Sirborn
Nils Landegren
Photo: Ulf Sirborn
Autoimmune diseases affect approximately 5% of the population and women are greatly overrepresented. The Knut and Alice Wallenberg Foundation has now granted SEK 39.1 million to a project that aims to map the general differences in the immune system between men and women at the molecular level and to reveal the specific mechanisms that lead to the development of autoimmune disease. In particular, the researchers will study the genetic variations that can be linked to the development of SLE and Sjögren's syndrome to understand why these variations more often lead to disease in women, although they are found in men at equal frequencies.
The project is titled “Sex matters in autoimmune disease” and will be led by Marie Wahren-Herlenius, CMM Group Leader and professor at the Department of Medicine, Solna, together with co-applicants CMM Group Leader professor Olle Kämpe and senior researcher Björn Reinius, both at Karolinska Institutet, and assistant professor Nils Landegren, at Uppsala University and Karolinska Institutet, and affiliated to Olle Kämpe's Group at CMM.
This article is based on press releases from the Knut and Alice Wallenberg Foundation and Karolinska Institutet.
Published: 2022-10-05

Magnus Bäck
Photo: Creo Media Group
Intravenous treatment with omega-3 fatty acids in elderly hospitalised patients in intensive care due to COVID-19 seems to have positive effects on the ability of the immune system to cope with the virus, according to a study by researchers from CMM and others from Karolinska Institutet. In the future, the results, published in the journal Clinical and Translational Medicine, could lead to a complementary, cost-effective treatment for COVID-19.
In patients with COVID-19, the immune system and the body’s activation of white blood cells are over-activated. It can lead to a so-called systemic inflammatory storm, which worsens the disease state and can cause complications such as sepsis and heart failure.
Researchers at Karolinska Institutet, among others, have through this study shown that omega-3 fatty acids can stimulate active healing of inflammation, without inhibiting the immune response. By accelerating the healing of the inflammation without compromising the body’s immune system, it could be possible to counteract the most serious complications of COVID-19, the authors believe.
Stimulated inflammation-healing molecules
The study was conducted in 2020, at an early stage of the pandemic when there were no vaccines available. It looked at 22 elderly hospitalised COVID-19 patients, one-half of whom were randomly assigned to intravenous treatment with omega-3 fatty acids for five days and the other half to intravenous administration of corresponding volumes of saline.
The treatment effect was found by mapping inflammatory biomarkers and immunological reactions.
“First, we showed that fatty acid metabolism to inflammation-healing molecules was stimulated in those patients treated with omega-3 fatty acids. By isolating immune cells before, during, and after treatment, we were able to show that immune function improved,” says Magnus Bäck, CMM Group Leader, senior consultant in cardiology and professor at the Department of Medicine, Solna, Karolinska Institutet, and the study’s corresponding author.
The biochemical analyses were carried out in collaboration with Craig Wheelock's research group at the Institute of Environmental Medicine, Karolinska Institutet.
Planning further studies
Researchers are now planning for larger clinical studies, which will be needed to show whether the course of the disease in severe COVID-19 is improved through treatment with omega-3 fatty acids.
“It is important that even our weakest and frailest patients have the opportunity to participate in studies when the enemy, in this case, COVID-19, is on the attack and that they can fight the disease with the help of the medicine,” says Dorota Religa, senior consultant and professor in geriatrics at the Department of Neurobiology, Care Sciences and Society, Karolinska Institutet.
“Stimulating the healing of inflammation with omega-3 fatty acids has the potential to lead to a new, cost-effective low-risk treatment for COVID-19, as a complement to existing treatment,” says Magnus Bäck.
The study was funded by King Gustaf V’s and Queen Victoria’s Masonic Foundation. There are no reported conflicts of interest or industry sponsors.
Publication
“Immunomodulation by intravenous omega-3 fatty acid treatment in older subjects hospitalized for COVID-19: a single-blind randomized controlled trial”, Hildur Arnardottir, Sven-Christian Pawelzik, Philip Sarajlic, Alessandro Quaranta, Johan Kolmert, Dorota Religa, Craig E. Wheelock, Magnus Bäck. Clinical and Translational Medicine, Letter to the editor, online 19 September 2022, doi: 10.1002/ctm2.895.
This article is based on a press release from Karolinska Institutet.
Published: 2022-09-22

Richard Rosenquist Brandell
Photo: Rick Guidotti
In Sweden the key to the successful implementation of precision medicine in healthcare has been the bottom-up approach where academia and health-care have joined forces to build a national infrastructure. This work has been de-scribed in a recent publication in Nature Medicine by 42 authors from the Clinical Genomics platform at SciLifeLab and Genomic Medicine Sweden (GMS). CMMer Richard Rosenquist Brandell, the Director of GMS, is the corresponding author.
Together, Clinical Genomics and GMS have established an innovative framework for continuously developing, adapting and implementing precision medicine in healthcare – for all patients. At the seven locations in Sweden with a faculty of medicine and regional healthcare provider with university hospital care both Clinical Genomics and GMS have established nodes to facilitate the development and optimization of new technologies and methods and their introduction into healthcare throughout the country.
“It is thanks to the great commitment of all enthusiasts around Sweden that we have been able to establish a national infrastructure for precision medicine. We are now observing how whole-genome sequencing and other broad genetic analyses are used more and more in healthcare, with increased opportunities for targeted treatment and follow-up,” says Richard Rosenquist Brandell, director of GMS, Professor at the Department of Molecular Medicine and Surgery, Karolinska Institutet, CMM Group Leader and senior consultant at Karolinska University Hospital.
Clinical Genomics and GMS have together with the healthcare system implemented whole-genome sequencing in the clinic for rare diseases. Similar projects are also underway in childhood cancer and leukemias. The infrastructures have jointly developed a number of national broad gene panels for cancer diagnostics that also contain pharmacogenes for dose adjustment. Other focus areas are whole-genome analyses for various infectious diseases and pandemic preparedness.
This article is based on news articles from Karolinska Institutet's and Genomic Medicine Sweden's websites.
Publication
Implementing precision medicine in a regionally organized healthcare system in Sweden. Thoas Fioretos, Valtteri Wirta, Lucia Cavelier, Eva Berglund, Mikaela Friedman, Michael Akhras, Johan Botling, Hans Ehrencrona, Lars Engstrand, Gisela Helenius, Therese Fagerqvist, David Gisselsson, Sofia Gruvberger-Saal, Ulf Gyllensten, Markus Heidenblad, Kina Höglund, Bo Jacobsson, Maria Johansson, Åsa Johansson, Maria Johansson Soller, Maréne Landström, Pär Larsson, Lars-Åke Levin, Anna Lindstrand, Lovisa Lovmar, Anna Lyander, Malin Melin, Ann Nordgren, Gunnel Nordmark , Paula Mölling, Lars Palmqvist, Richard Palmqvist, Dirk Repsilber, Per Sikora, Bianca Stenmark, Peter Söderqvist, Henrik Stranneheim, Tobias Strid, Craig E. Wheelock, Mia Wadelius, Anna Wedell, Anders Edsjö, Richard Rosenquist. Nature Medicine, online 19 september, 2022, doi:10.1038/s41591-022-01963-4
Published: 2022-09-19

Fredrik Piehl
Photo: Stefan Zimmermann
Early intervention with rituximab, a drug used in the treatment of rheumatoid arthritis (RA), can reduce the risk of deterioration in myasthenia gravis, an autoimmune disease that causes loss of muscle control. This is according to a randomised clinical study led by researchers at Karolinska Institutet and CMM and published in the journal JAMA Neurology.
“Patients with new onset myasthenia who received rituximab as a complement to standard of care showed greater improvement compared with patients who were given a placebo,” says the study’s principal investigator Fredrik Piehl, CMM Team Leader and professor at the Department of Clinical Neuroscience, Karolinska Institutet.. “They also needed fewer adjuvant treatments and lower doses of cortisone than the placebo group. These are encouraging results that give hope for a more effective strategy for controlling new onset myasthenia more quickly, even if larger studies will be needed to assess the long-term effects of the treatment.”
In myasthenia gravis, the immune system attacks the receptors between nerves and muscles, causing abnormal muscle weakness and fatigue. It often starts around the eye muscles but usually spread to other muscles of the body. The disease tends to progress in flare-ups and since there is no curative treatment, intervention is primarily aimed at dampening the immune system and treating the symptoms. Around 25 per 100,000 people live with the disease in Sweden, the majority of whom are women.
There is only one approved drug for myasthenia, Soliris, but the treatment is costly, which means that very few patients – none to date in Sweden – have benefited from it. Instead, many patients are treated with cortisone, which can cause side effects, and older tablet treatments that tend to lack scientific support.
The current study included 47 adult patients who had been diagnosed with myasthenia over the past year. Twenty-five of them were randomly assigned to a one-off treatment with 500 mg rituximab, a tried and tested drug used to treat rheumatoid arthritis, and 22 to a placebo group. The study was conducted at seven clinics in Sweden and the patients were followed for up to 48 weeks.
After four months, 71 percent of the rituximab group had attained good control of their disease according to a well-established 13-item rating scale, compared with 29 percent of the placebo group. Later follow-ups at six, nine and twelve months produced similar results.
The rituximab group also received on average lower doses of cortisone and needed fewer adjuvant treatments. However, they also reported more adverse reactions, most of which were mild. One patient with previously diagnosed heart disease in the group died from a myocardial infarction with cardiac arrest. Three patients in the placebo group needed hospital care during the study period, two for life-threatening conditions related to a deterioration of their myasthenia.
The researchers note that the study is relatively small with an imbalance in some of the baseline characteristics between the two groups, which is a limitation. At the same time, the results are promising and motivate further studies.
“The use of rituximab for myasthenia in Sweden increased even before the study results were finalized,” says Fredrik Piehl. “It is also a treatment that neurologists in Sweden are very familiar with thanks to the widespread and somewhat debated off-label prescribing for multiple sclerosis (MS). We’ll now, in a way similar to that with MS, analyse the long-term benefit-risk balance of the treatment with the help of national data collected via the Swedish myasthenia registry and national health registries. We also need to find markers that can predict the course of the disease at an early stage.”
The study was financed by the Swedish Research Council. Some of the researchers have received grants and fees from various pharmaceutical companies, including some that market rituximab, outside the scope of this study.
Publication: “Efficacy and Safety of Rituximab for New-Onset Generalized Myasthenia Gravis: The RINOMAX Randomized Clinical Trial.” Fredrik Piehl, Ann Eriksson-Dufva, Anna Budzianowska, Amalia Feresiadou, William Hansson, Max Albert Hietala, Irene Håkansson, Rune Johansson, Daniel Jons, Ivan Kmezic, Christopher Lindberg, Jonas Lindh, Fredrik Lundin, Ingela Nygren, Anna Rostedt Punga, Rayomand Press, Kristin Samuelsson, Peter Sundström, Oskar Wickberg, Susanna Brauner, Thomas Frisell. JAMA Neurology, online Sept. 19, 2022, doi: 10.1001/jamaneurol.2022.2887
This article is based on a press release from Karolinska Institutet.
Published: 2022-09-22

Göran K. Hansson
Photo: Markus Marcetic
CMMer Professor Göran K. Hansson has been awarded the European Society of Cardiology's gold medal. He is honored for his research efforts and, above all, for his discoveries on the role of inflammation in cardiovascular disease. The gold medal is the society's premier award and is awarded for outstanding contributions to cardiovascular medicine and was awarded at the congress in Barcelona.
Göran K. Hansson is Professor of Cardiovascular Research at the Department of Medicine Solna and works at BioClinicum and the Center for Molecular Medicine. For a number of years, he led the Linnaeus Center for Research on Inflammation and Cardiovascular Disease at KI, and during the years 2015-2022 he was the Secretary General of the Royal Swedish Academy of Sciences.
The European Society of Cardiology is the world's largest organization for cardiologists and cardiovascular researchers. At the congress in Barcelona, over 15,000 participants gathered to discuss new findings in cardiovascular medicine. The gold medal was awarded at the opening session of Barcelona's giant conference
center.
This article is based on a press release from Karolinska Institutet.
Published: 2022-07-27

Ingrid Lundberg
Photo: Vasan Kandaswamy
CMM Group Leader Ingrid Lundberg, professor at the Department of Medicine in Solna, is awarded the Grand Silver Medal for her efforts on behalf of Karolinska Institutet in pioneering scientific work on the rheumatic disease myositis, an area in which she is currently the world’s leading researcher.
Ingrid Lundberg's myositis research has altered understanding, diagnosis and treatment of this disease all over the world. She began her research on inflammatory muscle disease by observing patients suffering from myositis during her doctoral studies at Karolinska Institutet and Huddinge hospital in the late 1980´s when she saw the poor medical prognosis and great lack of knowledge relating to the disease, as well as observing how individual patients who engaged in physical exercise recovered, even though textbooks at the time strongly advised against movement and exercise. Two of the most clinically relevant research results that Ingrid Lundberg's research group have produced over the last 25 years are how physical exercise relieves myositis and how the disease develops over time. This has led to greatly improved and earlier diagnosis and treatment of the disease.
Ingrid Lundberg has made unique efforts to ensure that this research is also put into clinical practice in Sweden and abroad, and has carried out wide-ranging activities for Karolinska Institutet nationally and internationally. In this way, she has contributed to Karolinska Institutet's international reputation by creating a clinically rooted research environment at the university to which researchers and clinics across the globe turn for advice and collaboration regarding myositis disease.
Ingrid also has a long-term commitment to female researchers nationally and internationally. At Karolinska Institutet, she has served as chair for the Centre for Gender Medicine and has in many other ways supported and mentored female researchers in a range of different subjects.
Karolinska Institutet awards medals to people who have made special contributions to support KI. The medals are available in three categories: gold medal, grand silver medal and silver medal. Medals are awarded in connection with various academic ceremonies. The nominations are reviewed by the President, the Vice President, the University Director, the Academic Vice President for Research, the Academic Vice President for Doctoral Education, and the Academic Vice President for Higher Education.
Ingrid Lundberg has received the Grand Silver Medal 2022 together with Jan Andersson, professor emeritus of infectious diseases at the Department of Medicine, Huddinge and Stefan Einhorn, professor at the Department of Oncology-Pathology.
This article is based on the press release from Karolinska Institutet about the Grand Silver Medals 2022.
Published: 2022-06-21

CMM Group Leader and professor at the Department of Molecular Medicine and Surgery at Karolinska Institutet, is the recipient of the Mayo Clinic Cancer Center and Karolinska Institutet Cancer Research Award 2022.
The research project "Multi-Omics Studies of BTK Inhibitor Resistance in Chronic Lymphocytic Leukemia" will be headed by Professor Richard Rosenquist Brandell in collaboration with Dr. Neil E. Kay at the Mayo Clinic in Rochester, Minnesota.
The project includes multi-omic analysis in patients with chronic lymphocytic leukemia, who develop drug resistance against precision therapy with BTK inhibitors.
This article is based on a press release in Swedish from Karolinska Institutet.
Richard Rosenquist Brandell
Photo: Rick Guidotti
Published: 2022-06-15

The repurposing of FDA-approved drugs for alternative diseases is a faster way of bringing new treatments into the clinic. Researchers at CMM together with collaborators have repurposed a cancer drug for treatment of neuroinflammatory diseases such as multiple sclerosis. A novel drug carrier was also developed to facilitate drug delivery to target myeloid cells. These pre-clinical findings are described in a paper in the journal EMBO Reports.
Microglia are an organ-specific type of macrophage in the central nervous system. In the majority of chronic neurodegenerative disease conditions, such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease and chronic multiple sclerosis (MS), dysfunctional microglia play an important role. Modifying the activation of these disease-promoting microglia is an attractive therapeutic principle.
Keying Zhu (photo: private), Bob Harris (photo:Ulf Sirborn).
“The biotechnology industry has realized the potential for microglia-targeting strategies, and at least 20 new companies have started during recent years,” says Professor Bob Harris Group Leader at CMM and the Department of Clinical Neuroscience, Karolinska Institutet. “Compared to novel drug discovery programs that can take 20 years before a new medicine is approved, using existing prescribed drugs can halve that time.”
The researchers used in silico drug screening to identify candidates for microglial modulation and selected a Topoisomerase 1 (TOP1) inhibitor for further study. TOP1 was highly expressed in neuroinflammatory conditions both in mice and in tissues from MS patients, and TOP1 inhibition using camptothecin (CPT) and its FDA-approved analog topotecan (TPT) reduced inflammatory responses in microglia and macrophages in in vitro cultures, as well as ameliorating neuroinflammatory diseases in vivo.
Old drugs become new drugs
“The data-mining of open access databases is an approach that is both time and economically efficient, and there is so much data available nowadays,” says first author Keying Zhu, PhD student in Bob Harris' Group at CMM and the Department of Clinical Neuroscience, Karolinska Institutet. “We were lucky to identify four compounds with the properties we wished for, and one of these proved to be promising for our continued investigations, ultimately demonstrating significant therapeutic effect in our experimental model of MS.”
To specifically target microglia and macrophages, a nanosystem using β-glucan-coated DNA origami (MyloGami) loaded with TPT (TopoGami) was developed in collaboration with Professor Björn Högberg’s group at the Department of Medical Biochemistry and Biophysics at Karolinska Institutet. MyloGami had enhanced specificity for myeloid cells and also prevented the degradation of the DNA origami scaffold. Myeloid-specific TOP1 inhibition using TopoGami significantly suppressed the inflammatory response in microglia and mitigated MS-like disease progression.
The study was financed by the Swedish Research Council, the Swedish Neurofonden Foundation, the StratNeuro funding for Collaborative Neuroscience Projects at Karolinska Institutet, the Swedish MS Research Foundation, Allitd Litt Sterkere (Norwegian ALS Association), Ulla Carin Lindquists foundation, and KI Doctoral funding.
This article is based on a press release from Karolinska Institutet.
Publication
“Myeloid cell-specific topoisomerase 1 inhibition using DNA origami mitigates neuroinflammation.” Zhu K, Wang Y, Sarlus H, Geng K, Nutma E, Sun J, Kung SY, Bay C, Han J, Min JH, Benito-Cuesta I, Lund H, Amor S, Wang J, Zhang XM, Kutter C, Guerreiro-Cacais AO, Högberg B, Harris RA. EMBO Reports, online May 20, 2022, doi: 10.15252/embr.202154499
Published: 2022-06-01


April S. Caravaca (photo: private), Peder S. Olofsson (photo:MedTechLabs).
The nervous system is known to communicate with the immune system and regulate inflammation in the body. Researchers from CMM now show how electrical activation of a specific nerve can promote healing in acute inflammation. The finding, which is published in the journal PNAS, opens new ways to accelerate resolution of inflammation.
The way the body regulates inflammation is only partly understood. Previous research by Peder Olofsson’s group at Karolinska Institutet and CMM and other research groups has shown that electrical stimulation of the vagus nerve can reduce inflammation. Such nerve stimulation has been used with encouraging results in clinical studies of patients with inflammatory bowel disease and rheumatoid arthritis. However, how nerve signals regulate active resolution of inflammation was unclear.
“We have now studied effects of signals between nerves and immune cells at the molecular level,” says April S. Caravaca, a researcher in Peder Olofsson’s group at CMM and the Department of Medicine, Solna, Karolinska Institutet as well as the Stockholm Center for Bioelectronic Medicine at MedTechLabs. “A better understanding of these mechanisms will allow for more precise applications that harness the nervous system to regulate inflammation.”
The researchers showed that electrical stimulation of the vagus nerve in inflammation shifts the balance between inflammatory and specialised anti-inflammatory molecules, which promotes healing.
“Inflammation and its resolution plays a key role in a wide range of common diseases, including autoimmune diseases and cardiovascular diseases,” says Peder Olofsson. “Our findings provide insights on how the nervous system can accelerate resolution of inflammation by activating defined signalling pathways.”
The researchers will continue to study how nerves regulate the healing of inflammation in more detail.
“The vagus nerve is only one of many nerves that regulate the immune system. We will continue to map the networks of nerves that regulate inflammation at the molecular level and study how these signals are involved in disease development,” says Dr Olofsson. “We hope that this research will provide a better understanding of how pathological inflammation can resolve, and contribute to more effective treatments of the many inflammatory diseases, such as atherosclerosis and rheumatism.”
The study was supported by grants from the Knut and Alice Wallenberg Foundation, the Swedish Research Council, the Swedish Heart-Lung Foundation, MedTechLabs and Novo Nordisk. Peder Olofsson holds shares in Emune AB. Co-author Jesmond Dalli is the founder of and head of research at Resolomics Ltd.
This article is based on a press release from Karolinska Institutet.
Publication
”Vagus nerve stimulation promotes resolution of inflammation by a mechanism that involves Alox15 and requires the α7nAChR subunit”. April S. Caravaca, Alessandro L. Gallina, Laura Tarnawski, Vladimir Shavva, Romain A. Colas, Jesmond Dalli, Stephen G. Malin, Henrik Hult, Hildur Arnardottir, Peder S. Olofsson. PNAS (Proceedings of the National Academy of Sciences), online 27 May 2022, doi: 10.1073/pnas.2023285119.
Published: 2022-06-01
.jpg)
Lars Klareskog (photo: Gustav Mårtensson).
EIT Health Scandinavia International has awarded SEK 23 million in research grants to support international collaboration in precision prevention in rheumatology, with almost half of the grant distributed to Karolinska Institutet and Region Stockholm. The project is led by researchers based at CMM and the scope includes creating the conditions for the early diagnosis of risk factors for developing RA (rheumatoid arthritis) and enabling patients at risk of developing RA to manage the risk through lifestyle changes.
Today, it can be difficult to identify individuals who are at high risk of developing RA, as these individuals often seek out care for diffuse symptoms (e.g.muscle pain or joint pain) that are also common in individuals who are not at risk of developing RA. Researchers at KI have now been awarded a grant from EIT Health Scandinavia.
The research project is being led by the CMMers Lars Klareskog, professor at the Department of Medicine, Solna and CMM Group Leader, as well as Martina Johannesson, project coordinator, in collaboration with the rheumatologists at Karolinska University Hospital and the Center for Rheumatology at the Academic Specialist Center, SLSO. The study includes an international partnership with the University of Erlangen-Nuremberg, Germany and Leiden University in the Netherlands, institutions known for their work with the prevention of RA.
“What makes this project unique is that the patients themselves are a driving force in the effort to prevent RA, which we can follow up on thanks to the international collaboration”, says Lars Klareskog, professor at KI. “For starters, the group in Erlangen has recently shown that a six-month course of treatment with a particular antirheumatic drug (DMARD) reduced the risk of developing RA by about 75%. Our project will now provide an opportunity to conduct additional clinical trials for the prevention of RA in a collaboration between KI, Region Stockholm and our international partners”, says Klareskog.
About the research project
The research project aims, in part, to make it easier to identify at-risk patients. This includes a digital questionnaire where individuals with joint and muscle problems respond to a series of questions. The answers are then analysed using an algorithm that identifies patients who are at risk of developing RA. Individuals who are found to be "positive", i.e. at-risk, using this algorithm are offered a blood test and an antibody analysis (developed by one of the project partners, ThermoFisher) to determine if their individual risk of developing RA is actually elevated. High risk patients are referred to rheumatology units for further follow-up.
In the next step, at-risk patients will use an app (which will be developed by the project partner, Elsa Science) to report several lifestyle factors that are associated with the risk of developing RA. This step also aims to provide an opportunity to reduce the risk of developing the disease by enabling patients to take preventative measures on their own. Another benefit of the study is that when patients are not in pain, the number of days a patient takes sick leave will also decrease. This is, of course, a tremendous benefit for the individual, but it benefits society as well.
The project builds on a previous Vinnova-funded project that was based in Stockholm on the prevention and early treatment of RA.
Several partners are included in the research project.
Karolinska Institutet
Elsa Science
Region Stockholm
ThermoFisher Scientific (Uppsala)
Friedrich Alexander, the University of Erlangen-Nuremberg, Germany
Leiden University Medical Center (the Netherlands) with consultation and participation from the Swedish Rheumatism Association and the Dutch Arthritis Foundation.
In total, the grant amounts to SEK 23 million, of which approximately SEK 10 million will go to Stockholm.
Period: May 2022 – December 2024
This article is based on a press release from Karolinska Institutet.
Published: 2022-05-13
Six predictors could help determine the amount of lithium needed to treat patients with bipolar disorder, according to a large study led by researchers at CMM and Karolinska Institutet. The study, published in the journal Lancet Psychiatry, also pinpoints for the first time genetic markers that seem to influence how quickly the body eliminates lithium from its system.
“Our model could already now be used to predict how much lithium a patient with bipolar disorder will need. This could cut valuable time spent on finding the right dose for each patient, potentially with life-saving impact,” says the study’s senior author Martin Schalling, professor at the Department of Molecular Medicine and Surgery, Karolinska Institutet and shared CMM Group Leader of the C. Lavebratt and M. Schalling Group.
Lithium is one of the most important treatments for patients with bipolar disorder, a condition that has been linked to an increased risk of suicide. The chemical substance works as a mood stabilizer and reduces episodes of depression and mania. How much is needed varies greatly between individuals and finding the right dose for each patient is key as too much can be toxic while too little is ineffective. To minimize the risk of side-effects, clinicians tend to initiate treatment at low doses that increase over time, meaning it could take months before the treatment has an effect.

Vincent Millischer (photo: Irene Gutierrez Perez), Martin Schalling (photo: Ulf Sirborn).
Studied more than 2,300 patients
To overcome this, researchers have long sought to find a model that could predict dose response in individual patients. Previous studies have identified markers such as age, sex and kidney function as possible predictors of how quickly the body eliminates lithium from its system (lithium clearance), which can be used to determine the amount needed on a daily basis. However, most studies have been limited by small sample sizes.
In the current study, the researchers examined electronic health records and registry data from 2,357 patients with bipolar disorder, which may represent the largest sample size for this kind of study to date. Both men and women in ages ranging from 17 to 89 were included, mainly of European ancestry.
Help in treatment decisions
The study found associations between the speed of lithium clearance and age, sex, kidney function (measured as eGFR), serum lithium concentrations and medication with diuretics and substances targeting the renin-angiotensin-aldosterone system (RAAS), which could be used to treat hypertension and other conditions.
“Our findings suggest that older patients, women, patients with reduced kidney function, and those taking certain medications require lower doses of lithium. Interestingly, we also discovered that the amount of lithium taken and lithium concentrations in the blood do not seem to be completely proportional, which goes somewhat against current thinking. Our model based on these predictors explained around 50-60 percent of the variance in lithium clearance, which is better than previous models and could be used to inform treatment decision,” says first author Vincent Millischer, a postdoctoral researcher at the Department for Molecular Medicine and Surgery, affiliated researcher at CMM and resident in psychiatry at the Medical University of Vienna.
Genetic links
The study also found associations between a lower lithium clearance and one genetic locus on chromosome 11 and could also show that genetic variants affecting BMI and kidney function were associated with lithium clearance. Even though adding the genetic markers only marginally improved the model’s predictive capability, the researchers say it opens the opportunity of personalized medicine in lithium treatment in the future.
“Next we will test our model in a clinical trial to see if it can reduce the time it takes to find the right amount of lithium for each patient,” Martin Schalling says. “If the outcome is positive, we will develop a digital app that could be used by psychiatrists in the future to help assess lithium dosage for patients with bipolar disorder.”
The study was funded by the Stanley Medical Research Institute, the Swedish Research Council, the Swedish Foundation for Strategic Research, the Swedish Brain Foundation, the Söderström Königska Foundation, the Bror Gadelius Minnesfond, the Swedish Mental Health Fund, Karolinska Institutet and Karolinska University Hospital. Some of the authors have declared receiving consulting and lecture fees from various pharmaceutical companies while others have declared no competing interests.
Publication: ”Improving lithium dose prediction using population pharmacokinetics and pharmacogenomics: a cohort genome-wide association study in Sweden,” Vincent Millischer, Granville J Matheson, Sarah E Bergen, Brandon J Coombes, Katja Ponzer, Fredrik Wikström, Karolina Jagiello, Martin Lundberg, Peter Stenvinkel, Joanna M Biernacka, Olof Breuer, Lina Martinsson, Mikael Landén, Lena Backlund, Catharina Lavebratt, Martin Schalling, The Lancet Psychiatry, online May 12, 2022, doi: 10.1016/PIIS2215-0366(22)00100-6
This press release was also published on Karolinska Institutet's webpage.
Published: 2022-04-28

Mattias Bronge (photo: Erik Holmgren), Hans Grönlund (photo:Jens Sølvberg).
A new publication by researchers from CMM sheds light on new target molecules of potential significance for personalised treatment of multiple sclerosis (MS). The study was published in Science Advances.
MS, a chronic inflammatory disease of the central nervous system, is one of the most common causes of neurological disability among young adults. Immune cells mistakenly attack the so called myelin sheaths, a fatty layer surrounding nerve fibers. Consequently, the electric signal cannot be transmitted properly in the nerve and eventually the nerve fibers are lost. MS causes neurological symptoms such as sensory disorders, difficulties with walking and balance and impaired vision. Current treatments reduce the number of disease relapses and alleviate symptoms, but no cure is available yet.
“Existing MS treatments are quite indiscriminate in their effect on the immune system, which risks eventually causing complications, such as infections,” says the first author Mattias Bronge, PhD student in Hans Grönlund’s CMM Group and at the Department of Clinical Neuroscience, Karolinska Institutet. “Guiding future treatments more accurately towards the immune cells driving the disease can therefore lead to greater efficacy and fewer side effects.”
The researchers analysed 63 proteins in blood samples from MS patients and healthy controls and identified four new molecules, called autoantigens that are targeted by the autoimmune T cell attack in MS. The tested proteins were selected in collaboration with the Human Protein Atlas and Professor Torbjörn Gräslund at KTH Royal Institute of Technology, and the study was conducted by KI, KTH and Region Stockholm.
Hans Grönlund’s and Tomas Olsson’s Groups at CMM have previously developed a method that makes it possible to identify the T cells that react to specific autoantigens. Thanks to this method, four new molecules can be added to the few already known autoantigens, thus making a significant contribution to future developments in diagnosis and treatment.
“Our method makes it possible to present these autoantigens in a way that enables us to identify and subsequently disable the T cells that react to them,” says Hans Grönlund.
Not all MS patients react to the same autoantigens but by applying precision medicine methods such as the identification of each patient’s autoantigen-specific and disease-driving immune cells, personalized treatments can be created.
“Once a patient’s individual autoantigen profile is identified, a treatment can be adapted accordingly,” explains Dr Grönlund the last author of the study. “Most autoimmune diseases are driven by T cells and, if we can find a way to target them in diseases like MS, we can pave the way for more precise treatments with fewer side effects for other autoimmune diseases. Thanks to our long-standing collaboration with Professor Roland Martin at the University of Zürich, our method will be included in a phase 2 clinical study that aims to ‘switch off’ the aggressive T cells which drive MS development and progression.”
The study was financed by Vinnova, the Swedish Research Council, the Swedish Brain Fund, Neuro, the Margareta af Uggla Foundation, Stratneuro and Region Stockholm. Hans Grönlund is founder of NEOGAP Therapeutics AB which has patented the method used and jointly holds the patent for the autoantigens featured in the study with Mattias Bronge. Co-authors Claudia Carvalho-Queiroz, Ola B. Nilsson, Andreas Kaiser and Guro Gafvelin are employed by NEOGAP Therapeutics AB.
This article is based on a press release from Karolinska Institutet.
Publication
”Identification of four novel T cell autoantigens and personal autoreactive profiles in multiple sclerosis”. Mattias Bronge, Klara Asplund Högelin, Olivia G. Thomas, Sabrina Ruhrmann, Claudia Carvalho-Queiroz, Ola B. Nilsson, Andreas Kaiser, Manuel Zeitelhofer, Erik Holmgren, Mathias Linnerbauer, Milena Z. Adzemovic, Cecilia Hellström, Ivan Jelcic, Hao Liu, Peter Nilsson, Jan Hillert, Lou Brundin, Katharina Fink, Ingrid Kockum, Katarina Tengvall, Roland Martin, Hanna Tegel, Torbjörn Gräslund, Faiez Al Nimer, André Ortlieb Guerreiro-Cacais, Mohsen Khademi, Guro Gafvelin, Tomas Olsson och Hans Grönlund. Science Advances, online 27 April 2022, doi: 10.1126/sciadv.abn1823.
Published: 2022-04-21

In a unique cohort of patients who have been treated for malaria in Sweden, CMM researchers have studied the stages of the immune response during the course of malaria infection. Their findings provide an important contribution to the question of how disease tolerance in malaria develops. The results are now published in the journal Cell Reports.
Malaria is caused by parasites that are spread to humans through the bites of infected mosquitoes. Once the parasites enter the body, they follow the bloodstream to enter the liver were they grow, divide and enter the bloodstream again, where they infect red blood cells. People who have malaria usually feel very sick with a high fever and shaking chills. The disease itself is partly a result of the immune response to the blood-stage of the infection, which

induces a strong inflammatory response. According to the WHO, the disease affected more than 241 million people and caused more than 600,000 deaths in 2020, mainly among young children in sub-Saharan Africa.
Natural immunity to malaria develops over time with repeated malaria episodes but protection against severe malaria develops more rapidly through so-called tolerance. It is important to gain a better understanding of how humans fight this serious disease in order to develop more efficient vaccines or treatments.
Investigated immune cells in different disease stages.
PhD student Julius Lautenbach is the first author of the Cell Reports publication. Together with his colleagues, he investigated the immune cells and proteins in blood samples from patients who had been treated for acute malaria infection at Karolinska University Hospital in Solna, Sweden and had recovered.
The patients were tested on six occasions during one year following the onset of the disease. A total of 53 patients were included, 17 of whom had contracted malaria for the first time, while 36 had grown up in malaria endemic areas, had had malaria many times before and now contracted the disease again after travel.
“Since we have followed the patients here in Sweden, we can study the natural course of the immune response after a malaria infection, without the risk of a new infection interfering with the results. This cohort has proved to be very valuable for studying the immunology of malaria," says co-author Professor Anna Färnert, Group Leader at CMM and Senior infectious diseases physician at Karolinska University Hospital.
In patients who had been infected for the first time, there was a strong inflammation produced by the innate immune system, while people who were re-infected had an ability to suppress the inflammation.
“In those who have had malaria before, we saw that the early presence of parasite-specific antibodies interrupt the first stages of the inflammation and prevent a certain type of inflammatory T-cells, called ɣ𝛿 T cells, from expanding," says Christopher Sundling, CMM Team Leader and last author of the study.
Possibility to improve vaccines
Currently, there is only one vaccine against malaria, called Mosquirix, and it was recommended for use by the WHO in October 2021. It targets the stage when the parasite first moves from the mosquito into the liver. Once it enters the bloodstream and gives symptoms, the parasite has transformed and at that stage, the vaccine does not work.
“This is a weakness of the current vaccine. Understanding how tolerance develops and what happens in the blood stage can help us develop other types of vaccines, which may not fully protect against infection but will lessen the chances of becoming seriously ill. If such a vaccine can enable people to survive the first infections that kill so many, we could save many lives," Christopher Sundling explains.
Even though efforts such as distribution of mosquito nets, improved diagnostics and treatments have led to a global decrease of malaria incidence over the past decades, the covid pandemic broke that positive trend.
“We now need to continue to ensure that people are protected from being bitten by infected mosquitoes and have access to rapid and effective treatment. But to further reduce the burden of disease and eventually eradicate malaria, new tools are required. An effective vaccine is really needed; that is how we have been able to manage other infections, also in poor countries," says Anna Färnert.
The research has been funded by the Swedish Research Council, Magnus Bergvall Foundation, the Åke Wiberg Foundation, Region Stockholm, and the Marianne and Marcus Wallenberg Foundation, as well as doctoral grants from Karolinska Institutet. The researchers declare that there are no conflicts of interest.
This article is based on a press release from Karolinska Institutet.
Publication
”Systems analysis shows a role of cytophilic antibodies in shaping innate tolerance to malaria.” Maximilian Julius Lautenbach, Victor Yman, Carolina Sousa Silva, Nadir Kadri, Ioanna Broumou, Sherwin Chan, Sina Angenendt, Klara Sondén, David Fernando Plaza, Anna Färnert, Christopher Sundling. Cell Reports, online 19 April 2022, doi: 10.1016/j.celrep.2022.110709.
Julius Lautenbach (photo: private), Anna Färnert (photo: Ulf Sirborn) and Christopher Sundling (photo: private).

Published: 2022-04-08

Michael Sundström. Photo. Anne-Li Engström
The Board of the Center for Molecular Medicine (CMM) has appointed Michael Sundström as the new Director of CMM. An experienced scientist, business leader and manager, Michael assumed his position on April 1, 2022.
Michael Sundström received his PhD from Uppsala University, followed by postdoctoral studies at Karolinska Institutet. He has more than 30 years of international experience in leading pharmaceutical and biotechnology organisations. He has held positions as Director for structure-based drug design and oncology Research & Development portfolio management at Pharmacia, as well as senior positions at Actar and Biovitrum. In 2003, he was one of the co-founders of the Structural Genomics Consortium (SGC) working at the University of Oxford as Chief Scientist. After moving to the positions of Managing Director for the Novo Nordisk Foundation Center for Protein Research in Copenhagen, and later Vice President of Discovery Research at Karolinska Development in Stockholm, he rejoined the SGC in mid-2014. As Scientific Director for the SGC Karolinska laboratory based at CMM, he mainly focuses on leading large European projects in partnership with the pharmaceutical industry. The projects aim to generate high-quality research tools, such as antibodies and chemical probes, for use in translational medicine research studies.
As CMM Director Michael Sundström will work in close cooperation with the Karolinska University Hospital, Karolinska Institutet and Region Stockholm in order to continue strengthening the connection between clinical and molecular research. In addition, together with the scientists, the CMM Board, CMM Steering Group and support personnel, he will further develop the Center as a leading and attractive cross-disciplinary research environment.
“I am very glad to have Michael Sundström on board as the new CMM Director. With his profile and experience, he is the right candidate to shoulder the responsibility of further developing our internationally recognised research center, with excellent foundations laid by previous Directors Professor Lars Terenius, Professor Lars Klareskog and Professor Helena Erlandsson Harris. With Michael Sundström leading the way, we are looking forward to continuing our close collaboration with the Karolinska University Hospital, Karolinska Institutet and Region Stockholm. I strongly believe Michael Sundström will lead CMM into the next phase. The future is bright.”, says Liselotte Jansson, Chair of the Board of the Center for Molecular Medicine.”
“It is truly exciting to take on this new role and set out to further develop CMM together with all the professionals who are the basis for its creative research environment. I believe that CMM has an excellent model for working at the lead of translational research, connecting the real health-related needs with intellectual capital, forefront scientific knowledge and technological possibilities. I am looking forward to further strengthening the organisation and our collective research outputs.”
About CMM
CMM is a foundation instituted by the Stockholm County Council (SLL), currently Region Stockholm and works in close collaboration between Karolinska University Hospital and Karolinska Institutet.
Published: 2022-03-08

Fredrik Wermeling and Gustavo Monasterio
Photos: Erik Holmgren and private, respectively.
Two CMM researchers, Fredrik Wermeling and Gustavo Monasterio, have received funding from the Swedish Cancer Society (Cancerfonden).
Associate Professor and CMM Team Leader Fredrik Wermeling received the Senior Investigator award, which will cover his salary during six years. The title of Fredrik’s research project is ”Identification of new drug targets and innovative therapeutic modalities to overcome drug resistance”. The purpose of the Senior Investigator Award is to give established and highly qualified researchers who have not yet reached a final academic position, the opportunity to do full time cancer research during six years.
Gustavo Monasterio, postdoctoral researcher in Eduardo Villablanca’s CMM Group, was awarded a Postdoctoral Position funded by the Swedish Cancer Society. The grant will cover Gustavo's salary for 3 years and the awarded postdoctoral project is titled ”Defining the role of B cells during intestinal cancer”. The purpose of the Swedish Cancer Society’s funding of Postdoctoral Positions is to benefit cancer research by recruiting successful young researchers to the field.
Published: 2022-03-08

Nikolaos Taxiarchis Skenteris and Ljubica Matic.
Photos: Private and Tintin Vidhammer, respectively.
CMM Researchers from the Vascular surgery group, at the Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, have for a number of years studied vascular calcification as a prominent pathophysiological process characterised by the deposit-ion of calcium crystals in the media or intima of vessels. In the new study led by CMMer PhD candidate Nikolaos Taxiarchis Skenteris and Associate Professor Ljubica Matic (CMM Team Leader), published in the Clinical and Translational Medicine journal, they utilised several large patient cohorts to investigate common mechanisms of vascular calcification in different vascular territories. Osteomodulin (OMD), a proteoglycan previously involved in bone mineralisation, was identi-fied as a novel biomarker in this context. The study reports a consistent and broad association of both circulating and tissue OMD levels with various forms of cardiovascular calcification, highlighting its’ potential for further biomarker evaluation in larger clinical trials.
Calcification is a key feature of late-stage diabetes, renal and cardiovascular disease, linked to major adverse events such as myocardial infarction and stroke. Other pathological vascular changes encompass inflammation, lipid accumulation, extracellular matrix remodeling and alterations in smooth muscle cell function. Understanding mechanisms of these processes as well as their interplay in disease progression, is of key importance for finding new clinical biomarkers or therapeutic targets. This study shows that OMD is an important early modulator of cardiovascular calcification processes, enriched in association with vessel wall inflammation and osteoblastic transition of vascular smooth muscle cells, with the capacity to attenuate matrix calcification once it is secreted in the extracellular tissue.
This study was organized as an international collaboration utilizing clinical material and data from several large cohorts of atherosclerosis (Biobank of Karolinska Endarterectomy, KI), chronic kidney disease (Kärl Tx biobank, Karolinska Institutet in collaboration with Prof Peter Stenvinkel) and calcific aortic valve disease material obtained by Prof Leon Schurgers’ group, Maastricht University. Through these collaborations we had access to unique patient material, but were also able to study advanced murine models of vascular calcification in order to understand the mechanisms regulated by OMD. Our findings imply that OMD has the potential both as a plasma bio-marker and as a therapeutic target for assessing cardiovascular calcification”, says Ljubica Matic, Team Leader in the Vascular Surgery group, MMK, Karolinska Institutet and corresponding author of the study.

Illustration of deposited calcium in the arteries.
Image: Wikimedia Commons, author: www.scientificanimations.com
The study was conducted in the framework of the EU Horizon 2020 International Training Network INTRICARE which was initiated in 2017 as collaboration among Maastricht University, The Netherlands, Aachen University Clinic, Germany and Karolinska Institute. The aim of INTRICARE, that gathered totally 15 PhD students across three Universities, was to study risks of vascular Intimal calcification and roads to regression of cardiovascular disease.
This research was funded by a research grant from the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement No 722609 (INTRICARE); Dutch Thrombosis Society, Netherlands Organization for Scientific Research, grants from the Swedish Research Council (VR), Swedish Heart-Lung Foundation (HLF), Swedish Society for Medical Research (SSMF), CIMED, Stockholm County Council (ALF), and research funding from the Mats Kleberg, Sven and Ebba Hagberg, Tore Nilsson, Magnus Bergvall and Karolinska Institutet Foundations, Sweden.
Publication:
Osteomodulin attenuates smooth muscle cell osteogenic transition in vascular calcification.
Skenteris NT, Seime T, Witasp A, Karlöf E, Wasilewski GB, Heuschkel MA, Jaminon AMG, Oduor L, Dzhanaev R, Kronqvist M, Lengquist M, Peeters FECM, Söderberg M, Hultgren R, Roy J, Maegdefessel L, Arnardottir H, Bengtsson E, Goncalves I, Quertermous T, Goettsch C, Stenvinkel P, Schurgers LJ, Matic L. Clin Transl Med. 2022 Feb;12(2):e682. doi: 10.1002/ctm2.682.
Read the Karolinska Institutet press release here.
Published: 2022-03-02

Eduardo Villablanca (photo: Knut och Alice Wallenberg Foundation, Magnus Bergström).
Which genes are expressed in the large intestine during tissue repair and where exactly? Using a technique called spatial transcriptomics, Eduardo Villablanca and members of his Group at CMM, together with other researchers, created a unique expression map of individual genes in mouse intestinal tissue and compared it to expression data from cell types in the human colon. This model can be used as a tool for understanding how different diseases affect the colon. The study is published in the journal Nature Communications.
Spatial transcriptomics is a technique that allows the visualization of the gene expression landscape in a tissue. It was developed at SciLifeLab by scientists from KTH Royal Institute of Technology and Karolinska Institutet. In the study performed by Eduardo Villablanca and co-workers, the technique was applied in
a novel way. To visualize a long tubular organ like the colon, they rolled up the tissue like a Swiss roll, and thus managed to fit and map the entire gene expression landscape of a long organ.
“Our spatial transcriptomics-driven visualisation enabled us to discover several previously unknown aspects, such as that the colon is divided into more segments than once thought,” says the study’s corresponding author Eduardo J. Villablanca, CMM Group Leader and docent at the Department of Medicine, Solna at Karolinska Institutet.

Based on combined transcription data human colon tissue and the spatial transcriptomics from the whole mouse colon, the researchers found that the location of certain intestinal and immune cells was the same in both mice and humans. This model is thus suitable for translational studies of human diseases. Using the novel gene expression map of the colon in the context of recovery after injury, the research group could also show that the genes for the more difficult to treat forms of the ulcerative colitis were found in tissue that was more damaged.
Image showing different gene expression programs in specific compartments of the murine colon. Credit: Ludvig Larsson.
Eduardo Villablanca’s Group worked on this study together with other researchers from Karolinska Institutet, Science for Life Laboratory, University Medical Center Hamburg-Eppendorf and Heidelberg University.
The researchers now aim at creating a reference map for the gene expression of all tissues of the digestive organs, from the mouth to the rectum.

Pipeline for the processing and analysis of the whole murine colon using spatial transcriptomics. Credit: Ludvig Larsson.
The Swedish Research Council, the Swedish Research Council for Environment, Agricultural Sciences and Spatial Planning (Formas), the Swedish Cancer Society and the Knut and Alice Wallenberg Foundation, among others, provided funding for this study, conducted at Karolinska Institutet.
Declarations of interest: Eduardo J. Villablanca has received research funding from the pharmaceutical company F. Hoffmann-La Roche; and Camilla Engblom, Ludvig Larsson and Joakim Lundeberg are scientific advisors to 10X Genomics, which acquired the company Spatial Transcriptomics in 2018.
This article is based on a press release from Karolinska Institutet:
Publication
”The spatial transcriptomic landscape of the healing mouse intestine following damage”, Sara M. Parigi, Ludvig Larsson, Srustidhar Das, Ricardo O. Ramirez Flores, Annika Frede, Kumar P. Tripathi, Oscar E. Diaz, Katja Selin, Rodrigo A. Morales, Xinxin Luo, Gustavo Monasterio, Camilla Engblom, Nicola Gagliani, Julio Saez-Rodriguez, Joakim Lundeberg och Eduardo J. Villablanca, Nature Communications, online Feb. 11, 2022, doi: 10.1038/s41467-022-28497-0
Published: 2022-02-16

Marie Wahren-Herlenius (photo: Hjärt-Lungfonden).
CMM Group Leader Professor Marie Wahren-Herlenius receives a total of SEK 5,100,000 from the Swedish Heart-Lung Foundation for researching the rare and serious children's syndrome Kawasaki's disease. Typically, the patients initially present with a high and persistent fever. The syndrome is a form of general blood vessel inflammation, vasculitis. It mainly affects children under 5 years of age and the causes are unknown. Without treatment, around 20 percent of the patients develop abnormal local swellings of the coronary arteries – the blood vessels that supply blood to the heart muscle. These artery swellings are called aneurysms and lead to death in approximately one percent of the affected children.
"We want to identify the prevalence of Kawasaki's disease in Sweden and examine how genetic and environmental factors contribute to the development of the disease" says Marie Wahren-Herlenius in Karolinska Institutet’s press release.
Marie Wahren-Herlenius and her research group will search for yet unknown disease mechanisms that are suitable for the development of new treatment methods with the aim of more efficient prevention of coronary artery aneurysms.
Published: 2022-02-16

Onur Parlak (photo: Magdalena Lindén).
In March 2021 the European Innovation Council (EIC) was launched with the aim of identifying, developing and scaling up breakthrough technologies and game-changing innovations. This funding program is split into four different but linked funding schemes to support innovations throughout their lifecycle; from early-stage research, to proof of concept, technology transfer, and the financing and scale up of start-ups and small-medium enterprises.
The CMMer Assistant Professor Onur Parlak is the responsible investigator for the Karolinska Institutet part of a multicenter collaboration that is one of 56 selected projects to receive the first financing from the Pathfinder program (EIC’s fund for early-stage development of future technologies). The project aims at developing a bioelectronic skin patch that can predict epileptic seizures before they happen. The project received in total EUR 3.7 million from EIC and involves four academic partners and two
companies: University of Portugal (coordinates the project), Karolinska Institutet, Danish Epilepsy Center, Centre National de la Recherche Scientifique, Biostrike and Kinetikos Health.
Epilepsy is a neurological disease that is characterized by epileptic seizures, which are bursts of abnormal activity in the brain. The symptoms of a seizure vary, but can include uncontrollable jerking and shaking, collapsing or losing awareness and staring blankly into space. Epilepsy is one of the most common serious central nervous system disorders worldwide. Prediction of epileptic seizures is particularly important for prevention of the fatal complication, called sudden unexpected death in epilepsy (SUDEP). The epidermal biosensor that will be developed in this EIC-awarded project is a patch that measures cortisol secreted through the skin through built-in microelectronic technology. The patch also has medication-containing capsules. The registered cortisol measurements are continuously processed by an algorithm that will signal if a risk level is reached, which in turn will lead to immediate drug administration. Simultaneously the device will send an alarm signal to healthcare workers and family members, alerting them to a potential epileptic seizure.
Karolinska Institutet receives ~EUR 1 million for the sensor design and prototyping of the epidermal electronic device, a work headed by Onur Parlak in the lab at CMM.

Illustration of the project, edited by Onur Parlak
Published: 2022-02-16

Richard Rosenquist Brandell (photo: Pär Olsson).
Professor Richard Rosenquist Brandell is a Group Leader at CMM, Senior Physician at the Karolinska University Hospital and Director of Genomic Medicine Sweden (GMS). He has been named Networker of the Year by the Swedish Network Against Cancer (Nätverket mot cancer).
The aim of the network is to raise common issues for the cancer-profiled patient associations through opinion formation and knowledge-raising activities. Networker of the Year is a honorary award given to a person or business that, through its commitment, supports the Network against the cancer's goal of strengthening the patient's influence and opportunities for optimal care.
The motivation of the jury:
“Richard is a true enthusiast who is passionate about better and equal cancer care in Sweden and for patient collaboration in research and care. He is well liked among patients, colleagues and in all networks. Richard has supported the Network Against Cancer's goal of strengthening the patient's opportunities for optimal care. His research and involvement in precision medicine inspires the profession, academia, patients and business as well as political decision-makers.”
“This is a great honor for me and I am really moved to receive this award from the Network Against Cancer. We have worked closely with each other to achieve equal access to precision medicine in healthcare. The Network Against Cancer has been part of the whole journey with GMS”, says Richard Rosenquist Brandell in a press release from Karolinska Institutet.
The award was presented at World Cancer Day on February 4 and it consists of a diploma and the honor.
Published: 2021-12-21

CMMers Hildur Arnardottir (photo: Stefan Zimmermann) and Magnus Bäck (photo: Leonard Gren).
CMM researchers publish a study in Journal of Clinical Investigation, showing that a receptor activated by substances formed from omega-3 fatty acids plays a vital role in preventing inflammation in blood vessels and reducing atherosclerosis.
Cardiovascular diseases are the major cause of deaths worldwide. Atherosclerosis is a cardiovascular disease that is characterized by chronic inflammation in the blood vessels, which leads to the formation of lesions and subsequent thickening of the inner lining of the artery walls.
Both risk factors and protecting factors for atherosclerosis have been identified. Intake of food that is rich in omega-3 fatty acids has been associated with lower risk for atherosclerosis and now a study published in the Journal of Clinical Investigation sheds light on an important molecular player behind this protection. The CMMers Hildur Arnardottir and Magnus Bäck are the first and last author of the study, respectively.
When omega-3 fatty acids are metabolized, a type of anti-inflammatory compounds called resolvins, are formed. Resolvins switch off the inflammation and stimulate tissue healing. The authors of the study found that the resolvin receptor GPR32 is dysregulated in patients with atherosclerosis. This led them to create a mouse model for atherosclerosis where the GPR32 receptor is overexpressed. They found that signalling via the GPR32 receptor counteracted atherosclerosis and inflammation in the blood vessels and stimulated healing.
“This discovery can pave the way for completely new strategies for treating and preventing atherosclerosis by arresting inflammation in the blood vessels, while also turning on the body’s healing processes with the help of omega-3 fatty acids, for example,” says Hildur Arnardottir.
The researchers are planning to continue investigating the implications of dysregulated signaling through GPR32 receptors.
“We’ll now be studying the mechanisms behind the failed management of inflammation in the blood vessels and how omega-3 mediated stop signals can be used to treat atherosclerosis,” says professor Magnus Bäck, Group Leader at CMM and principal investigator behind the research project.
The study was mainly financed by the Swedish Research Council, the Swedish Heart-Lung Foundation, King Gustaf V and Queen Victoria's Foundation of Freemasons and Region Stockholm. There are no reported conflicts of interest.
This article is based on a press release from Karolinska Institutet.
Publication
Hildur Arnardottir, Silke Thul, Sven-Christian Pawelzik,Glykeria Karadimou, Gonzalo Artiach, Alessandro L. Gallina, Victoria Mysdotter, Miguel Carracedo, Laura Tarnawski, April S. Caravaca, Roland Baumgartner, Daniel F.J. Ketelhuth, Peder S. Olofsson, Gabrielle Paulsson-Berne, Göran K. Hansson, Magnus Bäck. The resolvin D1 receptor GPR32 transduces inflammation-resolution and atheroprotection. The Journal of Clinical Investigation, 15 December 2021, doi: 10.1172/JCI142883.
Published: 2022-02-01

Olle Kämpe. Photo: Private
Professor Olle Kämpe is a Group Leader at CMM and his passion is to understand what autoimmunity is. In November, he was awarded the Minerva Foundation’s Bror-Axel Lamberg Prize in Endocrinology during the Annual Meeting of the Finnish Endocrine Society. The Finnish Endocrine Society nominates three distinguished Finnish or Nordic scientist in the field of endocrinology from whom the Minerva Foundation selects the winner. The prize is €10 000 and this was the third time the prize was awarded.
The motivation of the Prize Committee:
“Olle Kämpe has a record of accomplishment in endocrine research spanning three decades and covering studies from basic genetic studies to registry and epidemiology research. The red thread in his research has been autoimmune endocrine diseases.
Kämpe has described several autoantigens and autoantibodies in organ-specific autoimmune diseases. His research is characterized by broad and complementary methodological approaches including the use of non- traditional disease models to understand the pathogenesis of autoimmune diseases and his work has significantly improved the diagnosis, treatment and follow-up of patients.”
Olle Kämpe has received numerous awards for his work. In 2010 he was elected member of The Swedish Royal Academy of Science and in 2016 member of the Nobel Assembly. He is currently member of the Nobel Committee for Medicine or Physiology. He received the Swedish Medical Society's Jubilee Prize in 2016 and the European Hormone Medal in 2020.
Published: 2021-12-14

CMMers Maribel Aranda-Guillén and Nils Landgren, authors of the PNAS publication. Photo: Olle Kämpe
The most common approach to finding what causes a disease is to study many affected patients in order to find a common denominator. In this new study published in Proceedings of the National Academy of Sciences (PNAS), however, the researchers started from the opposite end by testing whether a suspected autoantigen can be matched with a disease.
Autoantigens are the body’s own proteins that the immune system reacts against in autoimmune diseases. Transglutaminase 1 (TGM1), which is found in the skin, belongs to a protein family with many known autoantigens. TGM1 has been previously linked to a hereditary skin disease and now the researchers wanted to know if TGM1 was also involved as an autoantigen in acquired skin diseases. They searched for it in more than 500 patients with a wide range of autoimmune and unexplained skin diseases. Eventually, they found a match.
“We saw that patients with the cancer-associated and severe blister-causing skin disease paraneoplastic pemphigus presented antibodies against TGM1. We confirmed the findings in a larger group of patients and
could conclude that TGM1 antibodies were completely specific for paraneoplastic pemphigus,” says Nils Landegren, medical doctor and researcher specialising in autoimmune diseases at Uppsala University and affiliated to Olle Kämpe’s group at Karolinska Institutet and CMM. Nils lead this study, which was conducted together with other CMM researchers, Karolinska Institutet, Uppsala University as well as Osaka City University, Japan.
Knowing which autoantigens are associated with a specific disease is important, both to understand the disease mechanisms and to be able to diagnose autoimmune diseases and to give patients the correct treatment.
“We believe that TGM1 antibodies can be valuable as a diagnostic marker in investigations of blister-forming diseases. Patients that test positive should be quickly investigated to eliminate the possibility of undiscovered cancers,” says Nils Landegren.
The researchers believe that this new approach to identifying biomarkers can be used in studies of all sorts of autoimmune diseases, and that this could be a way to leverage the growing amount of openly available data on gene expression and function.
This study was funded by the Swedish Research Council, the Knut and Alice Wallenberg Foundation, the Swedish Society for Medical Research, the Göran Gustafsson Foundation and the Novo Nordisk foundation, among others. The contributing researchers have applied for a patent for the diagnostic use of TGM1 antibodies.
This text is based on the press releases from Uppsala University and Karolinska Institutet.
Publication
Nils Landegren, Norito Ishii, Maribel Aranda Guillen, Horður Ingi Gunnarsson, Fabian Sardh, Åsa Hallgren, Mona Ståhle, Eva Hagforsen, Maria Bradley, Per-Henrik Edqvist, Fredrik Ponten, Outi Mäkitie, Liv Eidsmo, Lars Norlén, Adnane Achour, Ingrid Dahlbom, Ilma Korponay-Szabo, Daniel Agardh, Mohammad Alimohammadi, Daniel Eriksson, Takashi Hashimoto, Olle Kämpe. A gene-centric approach to biomarker discovery identifies transglutaminase 1 as an epidermal autoantigen. PNAS 2021 vol. 118, DOI: 10.1073/pnas.2100687118, http://doi.org/10.1073/pnas.2100687118
Published: 2021-12-14

Anna Färnert (photo: Ulf Sirborn), Kristina Broliden (photo: Stefan Zimmermann), Christopher Sundling (photo: private)
Published: 2021-12-14
On December 2, the Swedish Research Council published the decisions on applications to be awarded project grants within Development research this year.
Three researchers from CMM obtained grants within this category:
Anna Färnert was awarded SEK 4 499 000 for the project titled ”Malaria immunitet: studier av immunsvar mot variabla parasitantigen”.
Kristina Broliden was awarded SEK 4 451 370 for the project titled ”Identifiering av molekylära signaler i genitalslemhinnan hos kvinnor som avgör känslighet för sexuellt överförbara infektioner”.
Christopher Sundling was awarded SEK 4 439 000 for the project titled ”Utvärdering av nya diagnostiska tester för detektion av progression från latent till aktiv tuberkulos”.
Karolinska Institutet press release (in Swedish)

Top to bottom, left to right: Magdalena Paolino (photo: Ragnar Söderberg Foundation), Karin Loré (photo: Ulf Sirborn), Anna Lindstrand (photo: Ola Lindström), Annika Lindblom (photo: Karolinska Institutet), Taras Kreslavisky (photo: Ulf Sirborn)
The Swedish Cancer Society (Cancerfonden) has awarded five researchers from CMM with grants for the following projects:
Taras Kreslavisky received a total of SEK 3 000 000 during 3 years for the project titled "Do tumor –infiltrating gamma-delta T lymphocytes recognize tumor antigens?"
Magdalena Paolino received a total of SEK 2 400 000 during 3 years for the project titled "A new ubiquitin-related gene in colorectal cancer: preclinical studies to test the effects of its inhibition in tumor growth and metastasis".
Annika Lindblom received a total of SEK 2 000 000 during 2 years for the project titled "Cancer predisposition and prevention".
Anna Lindstrand received a total of SEK 1 800 000 kr during 3 years for the project titled "Using zebrafish to develop combination therapies to overcome drug resistance in AML".
Karin Loré received a total of SEK 1 600 000 kr during 2 years for the project titled "SARS-CoV-2 vaccination in highly immunocompromised recipients of CAR T cell therapy and allogeneic stem cell transplantation".
The list of awarded projects can be found here.
Published: 2021-12-01

Susanna Brauner. Photo: Ulf Sirborn
Published: 2021-11-18
The Swedish Society for Medical Research (SSMF) annually awards SSMF’s Large Grant to young and promising medical researchers. This year the CMMer Susanna Brauner is one of the three recipients from Karolinska Institutet. In total, the foundation received 158 applications out of which eight were granted. Susanna’s grant consists of 5 256 000 SEK for four years research part time for the project “Myasthenia gravis- towards improved prognostication and treatment.”
Myasthenia gravis is a serious, potentially life-threatening neuromuscular disease. It is caused by an autoimmune reaction directed against the nerve connection on the skeletal muscles, resulting in fluctuating muscular fatigability (decrease of the muscle’s ability to perform over time).
Susanna's research has three “arms” that span from pathogenesis to treatment in myasthenia gravis (MG).
"It is nice that there are people who believe in you as a researcher and want invest money in a relatively uncommon disease such as MG. It makes you feel that you are on the right track. This establishment grant allows me to combine research time with my clinical work and hire more people to the research team", says Susanna Brauner.
Read the Karolinska Institutet press release here.

Long Jiang and Fredrik Wermeling. Photos: Erik Holmgren
CRISPR/Cas9 gene editing is a promising technique in the rapidly developing field of precision medicine. In a recent publication, the CMMers Long Jiang (PhD Student), Fredrik Wermeling (Team Leader), and co-authors show that the editing technique can lead to the enrichment of cells with mutations in a broad p53-linked network of cancer-related genes. p53 is a protein that is central to the cellular response following DNA damage. Notably, inactivating p53 mutations are the most common genetic alterations found in cancer, and several viruses have evolved mechanisms to interfere with the activity of p53. In the study recently published in Cancer Research, CRISPR gene editing was found to give survival advantages to cells with inactivating mutations in the p53 gene, as well as in genes part of a CRISPR-p53 interactome. The enrichment of the potentially cancerous cells was due to the survival advantage compared to cells with a fully functional p53 pathway. By studying the biology of p53 in the context of CRISPR-induced DNA damage, the researchers additionally identified several strategies to enable safer CRISPR use. In follow-up studies Fredrik Wermeling and his research team plan to explore those strategies in more clinic-centered tests.
The study was conducted in collaboration with David P. Lane’s group (MTC) and financed by the Swedish Research Council, the Swedish Cancer Society, Karolinska Institutet, the Magnus Bergvall Foundation, the China Scholarship Council, and the Nanyang Technological University-Karolinska Institutet Joint PhD Program.
Read more in Karolinska Institutet’s press release.
“CRISPR/Cas9-induced DNA damage enriches for mutations in a p53-linked interactome: implications for CRISPR-based therapies”, Long Jiang, Katrine Ingelshed, Yunbing Shen, Sanjaykumar V. Boddul, Vaishnavi Srinivasan Iyer, Zsolt Kasza, Saikiran Sedimbi, David P. Lane, and Fredrik Wermeling. Cancer Research, 18 November 2021, doi:10.1158/0008-5472.CAN-21-1692.
Published: 2021-11-17

Åsa Wheelock
Åsa Wheelock, Team Leader at CMM, has received SEK 3 million from the Swedish Research Council within the category of postcovid research. The awarded project led by Åsa, aims at developing a home monitoring system to facilitate continuous follow-up of long-COVID patients.
During the pandemic, a large group of severely affected individuals has emerged, with late onset persistent disease. While those requiring hospital care are largely elderly or multi-morbid individuals, the majority of long-COVID patients are formerly young, healthy individuals who due to mild initial symptoms generally did not seek hospital care. It is important to follow-up and understand the disease of this group of patients.
The project, titled ”Home monitoring and molecular phenotyping of patients with long-COVID for sub-grouping and follow-up of efficacy of interventions” aims at home monitoring the daily fluctuations in symptoms, activity and physiological parameters of the patients. This will help to identify sub-groups of the disease, with specific emphasis on those with lung involvement. The molecular phenotyping of the patients will be done in lung samples collected with bronchoscopy, and from particles in exhaled air. To evaluate the molecular biomarkers, the researchers will use a systems medicine workflow. With the help of the fully developed system, Åsa Wheelock and her colleagues plan to evaluate the effectiveness of various forms of treatment of long-COVID.
Published: 2021-11-10
Low Levels of MicroRNA-210 in Red Blood Cells Causes
Blood Vessel Injury in Type 2 Diabetes Patients

Zhichao Zhou and John Pernow. Private photos.
Published: 2021-10-29, updated 2021-11-25
Patients with type 2 diabetes develop damage to the blood vessels over time, which in turn increases the risk for blood cloths and complications such as heart attack and stroke. The mechanisms behind the vascular injury are however unknown. Associate Professor Zhichao Zhou and Professor John Pernow (CMM Group Leader) have, together with colleagues from CMM and Karolinska Institutet, published a paper in the journal Diabetes, showing that the effect is caused by low levels of the molecule microRNA-210 in red blood cells of type 2 diabetes patients and mice.
The researchers analyzed molecular changes in red blood cells from 36 patients with type 2 diabetes and from 32 healthy subjects, as well as in red blood cells from mice. They found that levels of the small molecule microRNA-210 were significantly reduced in blood cells from type 2 diabetes patients and diabetic mice. The reduction in microRNA-210 caused alterations in specific vascular protein levels, and impaired blood vessel endothelial cell function. Using a laboratory experimental setup, the researchers were able to restore dysfunctional blood vessel walls to normal by increasing the microRNA-210 levels in diabetic human red blood cells.
The results of this study may open up for new therapies targeted at increasing red blood cell microRNA-210 levels in type 2 diabetes patients.
Read more: Karolinska Institutet's press release
CMM Researchers Awarded Swedish Research Council Grants

Researchers from CMM have been awarded this years’ grants from the Swedish Research Council. The grant decision was announced on Thursday the 28th of October.
The following CMM researchers were awarded the 2021 project grants:
Robert Harris receives SEK 9 800 000 in project grant for the project “Nya immunterapier för neurodegenerativa sjukdomar”.
Marie Wahren-Herlenius receives SEK 9 800 000 in project grant for the project "Molekylär patogenes vid kongenitalt hjärtblock".
Taras Kreslavskiy receives SEK 5 200 000 in project grant for the project “Kartläggning av dynamiken i minnes B-cell- och plasmacellsdifferentiering”.
Ann Nordgren receives SEK 5 200 000 in project grant for the project “Medfödda genetiska faktorers betydelse för barncancer”.
Fredrik Wermeling receives SEK 5 200 000 in project grant for the project “Studier av neutrofila granulocyter vid autoimmun ledinflammation”.
Eduardo Villablanca receives SEK 5 200 000 in project grant for the project “Bestämning av den cellulära och molekylära arkitekturen av tarmregeneration efter akut inflammation.”
Helena Erlandsson Harris receives SEK 4 800 000 in project grant for the project “Inflammation, destruktion, smärta och neuroinflammation vid juvenil artrit, med ett särskilt fokus på alarminet HMGB1”.
Alexander Espinosa receives SEK 4 800 000 in project grant for the project “En ny humaniserad musmodell för att kartlägga IL8:s roll i cancer och immunterapi”.
Anna Färnert receives SEK 4 800 000 in project grant for the project “Hur blir och förblir man immun mot malaria”.
Ulf Hedin receives SEK 4 800 000 in project grant for the project “Instabil ateroskleros: från patient till molekyl till patient”.
Lara Kular receives SEK 4 800 000 in project grant for the project “Epigenetik av lung-hjärnaxeln vid multipel skleros: att reda ut effekterna av rökning på sjukdomspatogenes och progression”.
Kristina Broliden receives SEK 3 600 000 in project grant for the project “Identifiering av molekylära mekanismer för känslighet mot mukosala virusinfektioner”.
Sebastian Lewandowski receives SEK 2 400 000 in project grant for the project “Terapeutisk hämning av perivaskulär fibroblastaktivitet i ALS-modeller”.
Pontus Naucler receives SEK 2 400 000 in project grant for the project “Datadriven precisionsmedicin för ökad patientsäkerhet”.
Ola Nilsson receives SEK 2 400 000 in project grant for the project “Oklara tillväxt och skelettsjukdomar hos barn – från klinik till molekylära mekanismer”.
Read Swedish Research Council decision here.
Read Karolinska Institutet press release here.
Published: 2021-10-29
Per Svenningsson receives 1.86 million US dollar for studies on Parkinson’s disease

Per Svenningsson, Group Leader at CMM, has received a grant from About the Aligning Science Across Parkinson’s (ASAP) initiative, consisting of approximately 1.86 million US dollar for three years.
The purpose is to study how abnormal protein aggregates may spread from the gut to the brain to drive the early stages of Parkinson’s disease.
The grant is part of a project led by Project Director, Dr. Kaplitt, professor of neurological surgery, at Weill Cornell Medicine (US).
Read more in the Karolinska Institutet press release here.
Per Svenningsson. Photo: Ulf Sirborn.
Published: 2021-10-28
New Clues Linking Obesity and Type 2 Diabetes

Carolina Hagberg
Photo: Karolinska Institutet
The CMM Team Leader Carolina Hagberg, from the Department of Medicine, Solna, is the joint first author of a recent publication in Nature Medicine titled “Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce”. Qian Li is the other joint first author and the study was led by Kirsty Spaldling from the Department of Cell and Molecular Biology, Karolinska Institutet.
Obesity is considered an important risk factor for many chronic diseases, including diabetes, cardiovascular disease and cancer. The expansion of fat tissue (adipose tissue) in obesity is due to an increase in both fat cell progenitor maturation (differentiation) and mature fat cell size, with the latter dominating in human obesity. Fat cells, also called adipocytes, however, were previously thought to be unable to divide or enter cell cycle, and many conclusions about their behaviour have been based on this belief.
This is normal for many types of fully differentiated cells like adipocytes. In contrast, cells that retain their capacity to proliferate can upon the right ques enter the cell cycle, and following a series of events it grows and eventually divides. Before division, cells must first double their genetic material via replication in preparation for cell division.
“In the current study, we demonstrate that freshly isolated, mature human adipocytes unexpectedly displayed a gene and protein signature indicative of an active cell cycle program, expressing cell cycle markers such as KI67 and cyclins”, Carolina Hagberg explains.
“Adipocytes, however, do not seem to divide, rather they undergo an endoreplicative cell cycle, passing through the interphase without dividing, and thereby becoming polyploid, i.e. having more than the normal two copies of DNA.” This is in fact a more common practice then commonly known, and among other hepatocytes, megakaryocytes and trophoblasts (a cell type in the placenta) also undergo endoreplication without cell division.
For this study, the authors analyzed adipose tissue from 63 non-obese individuals (BMI under 30) who underwent surgery for umbilical hernia or cholecysectomy for gallstone disease, as well as 196 overweight individuals who underwent bariatric surgery for obesity.
It is well known that obesity is associated with a highly increased risk for insulin resistance, and insulin is necessary to regulate energy, glucose, for the body's cells. A persistent failure of the body’s cells to respond to insulin leads to the development of type 2 diabetes. High insulin levels have also been proposed to have direct pathological effects on tissues. In the study presented in Nature Medicine the researchers investigate the relationship between high insulin levels, obesity and the active cell cycle in adipocytes.
“Adipocyte cell cycle progression associates with patient obesity and hyperinsulinemia, and we could identify a concomitant increase in cell size, nuclear size and nuclear DNA content in freshly isolated fat cells,” says Carolina Hagberg. “In vitro, insulin stimulation was able to induce adipocyte DNA replication, measured via EdU incorporation and the doubling of adipocyte DNA content by FACS. We also show that insulin can act as a mitogen and stimulate AKT-phosphorylation in adipocytes independent of patient insulin resistance.”
In obese individuals, the adipose tissue shows signs of inflammation, but the molecular mechanism has been debated. “We found that, in contrast to normal fluctuations in insulin levels, chronic hyperinsulinemia in vitro or in patients, was associated with a subsequent cell cycle exit, leading to a premature senescent transcriptomic and pro-inflammatory secretory profile in adipocytes. This is important as premature senescence is rapidly becoming recognized as an important mediator of stress-induced tissue dysfunction, and has been implicated in the pathogenesis of obesity and type 2 diabetes but never shown to occur in mature adipocytes. By demonstrating that adipocytes can activate a cell cycle program we define the mechanism whereby mature human adipocytes senesce and demonstrate that by targeting the adipocyte cell cycle program using metformin, a common drug for treatment of type-2 diabetes, it is possible to impact adipocyte senescence and obesity-associated adipose tissue inflammation.”
"The results are important for understanding basal fat cell biology and the pathological changes to fat tissue function that occur during obesity and insulin resistance", says Carolina Hagberg, shared first author on the publication.
Publication:
Qian Li*, Carolina E. Hagberg*, Helena Silva Cascales, Shuai Lang, Mervi T. Hyvönen,
Firoozeh Salehzadeh, Ping Chen, Ida Alexandersson, Eleni Terezaki, Matthew J. Harms,
Maria Kutschke2,7, Nahida Arifen2, Niels Krämer1, Myriam Aouadi 2,5, Carole Knibbe8,
Jeremie Boucher, Anders Thorell and Kirsty L. Spalding. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nature Medicine, (2021) Oct 4, online ahead of print.
*Shared first authors
Link to Karolinska Institutet press release here.
Published: 2021-10-20
Vinnova Funding to Precision Medicine in Rheumatoid Arthritis

Per-Johan Jakobsson
Photo: Karolinska Institutet
Vinnova, Sweden’s innovation agency, recently funded eleven innovation environments in
precision medicine across the country. Professor Per-Johan Jakobsson, Group Leader at CMM, is the project leader and contact person for one of the three funded environments at Karolinska Institutet.
The project is called “Prevention and early therapy of rheumatoid arthritis using precision medicine”. The partners are Karolinska Institutet, Region Stockholm (where Karolinska Universitetssjukhuset and Centrum för Reumatologi are involved), Thermo Fisher Scientific, Elsa Science and Uppsala University. At Karolinska Institutet Per-Johan Jakobsson is the principal investigator for the project, with support from Lars Klareskog and Dr Martina Johannesson who is the project coordinator. They will receive SEK 20 million for 2.5 years that will be distributed between the different partners. CMM News met Per-Johan through Zoom for a short interview.
Where are we at today when it comes to the use of precision medicine in diagnostics, treatment and prevention of rheumatoid arthritis (RA)?
“We are at the beginning of the new era. We can identify individuals at high risk of developing rheumatoid arthritis (RA), using information such as serology (mainly identification of antibodies through blood samples), environmental risks like smoking, certain musculoskeletal symptoms and genetics. In our risk cohort (a group of individuals who are at very high risk of developing disease) close to 40% of the included individuals develop RA within 2 years. Now, with this project, we are going one step further and we actually like to use the term “precision prevention”. Recent data show that treatment of high-risk individuals with the T-cell inhibiting antibody Abatacept (commonly used treatment for moderate to severe RA) delayed and potentially prevented the development of RA in those individuals (Rech J et al., 2021, ACRabstracts.org). With data and knowledge gathered so far, there is thus a promising future for precision prevention medicine in RA.”
What is the plan for “Prevention and early therapy of rheumatoid arthritis using precision medicine” with the funding from Vinnova?
“Our RA research is already an innovation environment where we integrate quality registers, clinical as well as molecular data. The project that Vinnova has invested in is a natural continuation for us. We will use a web-based screening test “Rheumatic?” and an application for self-monitoring under development by one of the project partners, the e-health company Elsa Science. If the answers in “Rheumatic?” by a study participant matches with inflammation-like musculoskeletal symptoms, the persons will be asked to leave samples for genetic and serological analysis using diagnostic tests developed by another company-partner, Thermo Fisher Scientific. Then the person gets to see a doctor. Through the app for self-monitoring, the study participants will be able to track their symptoms and eventually share them with health-care. If the subsequent results identify the person to be at high risk for developing RA, the person will then be followed by the rheumatologists involved in the study and may also be included in clinical trials for prevention of RA. These tools will provide a coherent system that supports not only precision prevention of RA, but also enables rapid diagnosis and very early therapy for those who develop arthritis despite our prevention efforts. Today the average time from getting arthritis to receiving first treatment is 6 months. By applying the methods in this project, our goal, in addition to preventing the disease, is to be able to give the patients targeted treatment very soon after the first episode of arthritis. Our biggest goal of the project, however, is prevention.”
Published: 2021-09-28
Large Investment in Precision Medicine

Richard Rosenquist Brandell
Photo: Rick Guidotti
Genomic Medicine Sweden (GMS) has received 220 million SEK for continued introduction of precision medicine into Swedish healthcare. Through Sweden’s innovation agency Vinnova, the Swedish government has invested SEK 96 million in GMS and Swedish regional health authorities and universities have added another SEK 124 million.
Richard Rosenquist Brandell, Group Leader at CMM, is the director of the Swedish national infrastructure Genomic Medicine Sweden (GMS), launched in 2018. The aim of GMS is to translate innovation in genomics into clinical practice and to implement a sustainable infrastructure for precision medicine in Sweden. Precision medicine a more personalised approach to healthcare, seeking to take into account individual variability in genes, environment, and lifestyle for each person. The individual patient can thus be more effectively diagnosed and treated with a reduced risk of short and long-term side-effects.
GMS has coordinated development and implementation of genomic technologies for clinical use in healthcare throughout Sweden. By performing broad gene panel and whole-genome sequencing these tests detect genetic variations in cancer and rare diseases, that in turn improve diagnostics, treatment and follow-up of the patients. In all Swedish regions with university healthcare, there are now established regional Genomic Medicine Centers (GMC). A national genomics platform has been developed to enable the analysis and sharing of huge amounts of data in a harmonized and secure way. The genomic platform is also an important resource for research and development of new, targeted drugs. During the upcoming phases, GMS will also focus the work on linking the refined genetic diagnostics to clinical studies.
The aim of the recent funding of GMS is to contribute to the ongoing introduction of precision medicine into healthcare, strengthening Swedish research and promoting new research and innovation collaborations between industry, healthcare and academia. The long-term goal is that eventually every individual in Sweden should have access to personalised and more precise diagnostics and treatment.
Read more:
Published: 2021-09-28
Albin Björk is Awarded the Swedish Rheumatological Association's Prize for Best Dissertation


Albin Björk
Photo: Saga Rebecka Herlenius
The CMMer Albin Björk won the Swedish Rheumatological Association's (SRF) prize for best dissertation in 2021. His thesis, "Immunopathogenic mechanisms in primary Sjögren's syndrome", was focused on the characterization of exogenous and endogenous factors contributing to the immunopathology of the autoimmune disease primary Sjögren’s syndrome.
In the studies included in the thesis, Albin Björk and colleagues found that cigarette smoking does not appear to increase the risk of the disease, but that it may instead be a protective factor. A history of infections, however, was associated with a higher risk of development of Sjögren’s syndrome.
When studying lymphocytes from patients with Sjögren’s syndrome, they found aberrances in B cell gene expression patterns and dysregulation of the CXCR5/CXCL13 axis, which is important for lymphocyte migration.
In the thesis, Albin and his co-authors also found additional support for the hypothesis of infections as an environmental risk factor for systemic autoimmune disease. They did so by testing serological and cellular responses against viral antigens in patients with systemic autoimmune disease on no or mild treatment and found that the patients developed higher levels of virus-specific antibodies compared to healthy controls.
Albin Björk defended his thesis on April 23rd 2021 with Marie Wahren-Herlenius and Alexander Espinosa as supervisor and co-supervisor, respectively. Currently he is working as a physician at the Center for Rheumatology, which is a part of the Academic Specialist Center, and will continue his research on rheumatic diseases in parallel.
Read more:
Published: 2021-07-07
38 million SEK to Per Svenningsson’s Research on Parkinson’s Disease

Per Svenningsson, Group Leader at CMM, has received funding from Nordstjernan Holding AB and Axel Johnson Group consisting of 38 millions SEK for five years.
The purpose of the funding is to support Per Svenningsson’s research on basic mechanisms in the pathogenesis of Parkinson’s disease as well as the search for a future tailored treatment slow down disease progression.
Read more: Karolinska Institutet press release
Per Svenningsson. Photo: Ulf Sirborn.
Published: 2021-07-08
CMM Researchers Find That Autoantibodies Are Possible Contributors in Causing Fibromyalgia

Emerson Krock. Photo: Private
Camilla Svensson. Photo: Ulf Sirborn
Emerson Krock, Postdoc, and Camilla Svensson, Professor and Group Leader, are co-first and last authors, respectively, of a recent publication in the Journal of Clinical Investigation, showing that autoantibodies are possible contributors to fibromyalgia, a disorder characterized by chronic widespread pain in muscles and bones.
Researchers at Karolinska Institutet, King’s College London and the University of Liverpool, UK conducted the study as a collaboration.
The results pave the way for new approaches to the treatment of fibromyalgia.
Read more: Karolinska Institutet press release
Publication
“Passive transfer of fibromyalgia symptoms from patients to mice,” *Andreas Goebel, *Emerson Krock, Clive Gentry, Mathilde R. Israel, Alexandra Jurczak, Carlos Morado Urbina, Katalin Sandor,Nisha Vastani, Margot Maurer, Ulku Cuhadar, Serena Sensi, Yuki Nomura, Joana Menezes, Azar Baharpoor, Louisa Brieskorn, Angelica Sandström, Jeanette Tour, Diana Kadetoff, Lisbet Haglund, Eva Kosek, Stuart Bevan, *Camilla I. Svenssonand *David A. Andersson, Journal of Clinical Investigation, online 1 July, 2021, doi: 10.1172/JCI144201 (*co-first/last authors)
Published: 2021-09-01
Nine CMM Researchers Receive Funding from the Swedish Brain Foundation (Hjärnfonden) 2021

Photo: Unsplash, Robina Weermeijer
The Swedish Brain Foundation awards scholarships and grants to researchers and research groups to enable or enhance important basic and clinical research regarding the central nervous system.
This year the following researchers from CMM received grants:
(Name, Project Title, Research Area)
-
Faiez al Nimer, Detailed characterization of immune cells in multiple sclerosis, with focus on EBV, MS and other neuroinflammatory diseases
-
Eric Herlenius, Inflammation, neural networks, breathing difficulties and inspiration!, The brain of children and adolescents
-
Jan Hillert, Individualized treatment of multiple sclerosis for optimal long term outcome, MS and other neuroinflammatory diseases
-
Maja Jagodic, Epigenetics: New functional and therapeutic applications in multiple sclerosis, MS and other neuroinflammatory diseases
-
Ingrid Kockum, Genetic and protein biomarkers for risk and severity of multiple sclerosis, MS and other neuroinflammatory diseases
-
Ann Nordgren, Genetic mapping of children with neurological disabilities without diagnosis, The brain of children and adolescents
-
Tomas Olsson, Risk genes and pathogenesis in multiple sclerosis, MS and other neuroinflammatory diseases
-
Fredrik Piehl, Is myelin repair a realistic possibility in multiple sclerosis?, MS and other neuroinflammatory diseases
-
Per Svenningsson, GBA-Parkinson disease as a target for precision medicine, ALS, Parkinson’s disease, Huntington and other neurodegenerative diseases
Full list of awardees on the Swedish Brain Foundation website
Published: 2021-06-29
CMMers Receive StratNeuro Funding

Maja Jagodic. Photo: Karolinska Institutet

Bob Harris. Photo: Karolinska Institutet
The strategic research area neuroscience at Karolinska Institutet (StratNeuro) has awarded a total of 18 MSEK to six Collaborative Neuroscience Research Projects. Each project is funded with 3 MSEK.
Maja Jagodic and Bob Harris, both CMM Group Leaders and Professors at the Department of Clinical Neuroscience, have received funding for one collaborative project each. The purpose of the funding is to establish new collaborations.
Maja Jagodic was the main applicant for a project titled: "Targeting neuroinflammation and neurodegeneration using artificial ligands". The co-applicant was John Löfblom, Department of Protein Science, The Royal Institute of Technology (KTH).
Bob Harris, applied for funding of the project titled: "Novel immunotherapy targeting myeloid cells for neurological diseases: Drug repurposing and nanoengineering" and his co-applicant was Björn Högberg, Department of Medical Biochemistry and Biophysics, Karolinska Institutet.
The strategic research areas are efforts by the Swedish government in certain selected areas that has been ongoing for more than ten years. Karolinska Institutet receives governmental strategic funding for six fields: stem cells, diabetes, neuroscience, cancer, epidemiology and health research.
StratNeuro has the mission to integrate clinical and basic research and to foster a new generation of leaders and scientists in translational neuroscience.
The external evaluator panel for the collaborative StratNeuro funding 2021 consisted of the following researchers:
David Engblom, Linköping University
Cecilia Lindberg, Lund University
Paolo Medini, Umeå University
Håkan Olausson, Linköping University
Åsa Petersén, Lund University
Mart Saarma, Helsinki University, Finland
Published: 2021-06-24
Pontus Naucler Leads COVID-19 Vaccine Research Platform Funded by the Swedish Research Council

Pontus Naucler. Photo: Private
Pontus Naucler, researcher in Anna Färnert’s Group at CMM, will lead FASTER, a national COVID-19 vaccine research platform, one of six research environments awarded with a total of SEK 100 million from the Swedish Research Council.
FASTER will include register-based research and clinical trials as a basis for studying the immune response to the vaccines, as well as their safety and effectiveness.
This national platform will be a part of the large EU network called “VACCELERATE” which has the aim of evaluating ongoing and future vaccinations as well as making the process of clinical testing of vaccines faster in the future.
Published: 2021-06-23
Strategic Research Funding for Germline Development Studies
Qiaolin Deng, Associate Professor and CMM Team Leader, is one of the six 2021 Junior Grant awardees in the Karolinska Institutet Strategic Research Area in Stem Cells and Regenerative Medicine. Qiaolin’s research team is interested in the developmental principles of the germline (the cells that pass on genetic information to the progeny, i.e. the egg and sperm) in health and disease and the title of the awarded project is ‘Revealing deleterious gene dosage effects on germline specification and testicular stem cell niche in Klinefelter syndrome by single-cell technology’. The project will be supported with 4 MSEK for 2 years.


Project picture: Genetic mouse models and in vitro hPG-CLCs differentiation of KS-iPSCs as tools to study global gene dosage effects.
Qiaolin Deng. Photo: Sanjiv Risal
Why did you choose germline development as your research niche and why is this research relevant?
“As a researcher, I have always been very inter-ested in the early development of the embryos, and the germline is the basis for this development. The development of the germline in an individual is in itself a process of continuous maturation that goes beyond puberty. There is intricate regulation at the molecular level involved in this process. Many cases of non-heritable variation involve germline epigenetics, and these are questions that have not been extensively studied before. The epigenetic regulation influences the development at all levels, also processes such as clonality and migration of the germ cells. Over the past few years new technological advances have made it possible to study these questions with a deeper resolution. When I chose my research focus, I also caught the moment of technology advancement.“
Klinefelter syndrome is caused by the presence of at least one additional X-chromosome in the male. There are few distinguishing features of the syndrome before puberty which makes early diagnosis difficult. However, after puberty, the testicular environment starts to degenerate and sperm can usually not be produced as a result. Thus, one of the main features of the syndrome is male infertility.
Can you briefly explain the aim of the project for which you were awarded the Junior Grant?
“In the present project we will use Klinefelter- derived iPS cells*. We want to study how the extra chromosome influences germline devel-opment. Persons with Klinefelter syndrome suffer from infertility but it is not because they lack the germ cells from the beginning. At the start of puberty, the germ cell pool is still there however, there is a “toxic” process in the testicular environment that starts at puberty in individuals with Klinefelter syndrome. We ask the question why the extra X chromosome does not do much harm initially. What is the impact of gene dosage and allelic gene expression on germ cell maturation? I always think about allelic expression since my postdoc projects were focused on that. Our preferred method in this project is single cell sequencing and at CMM we have a great core facility for that!”
With whom do you collaborate?
“Our main collaborators are Jan-Bernd Stukenborg at KBH, KI (his team works on fertility preservation) and at CMM we collaborate with Ning Xu Landén in the single sequencing studies of wound healing.”
What is the relevance of these studies from a patient/public health perspective?
“Klinefelter syndrome is estimated to occur as frequently as 1 in 600 births, thus contributing significantly to male infertility on a population level. Even though our research is very basic, I would say that one ambitious goal is to be able to find ways to save their fertility earlier. We strive to do that by understanding the molecular mechanisms of their germline development better.”
*The iPS cells (induced pluripotent stem cells) originate from skin biopsies from patients with Klinefelter syndrome. The fibroblasts are then re-programmed back to an embryonic state. This helps us to study human disease in a better way.