1
|
Ma X, Zhang J, Jiang Q, Li YX, Yang G. Human microbiome-derived peptide affects the development of experimental autoimmune encephalomyelitis via molecular mimicry. EBioMedicine 2024; 111:105516. [PMID: 39724786 DOI: 10.1016/j.ebiom.2024.105516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Gut commensal microbiota has been identified as a potential environmental risk factor for multiple sclerosis (MS), and numerous studies have linked the commensal microorganism with the onset of MS. However, little is known about the mechanisms underlying the gut microbiome and host-immune system interaction. METHODS We employed bioinformatics methodologies to identify human microbial-derived peptides by analyzing their similarity to the MHC II-TCR binding patterns of self-antigens. Subsequently, we conducted a range of in vitro and in vivo assays to assess the encephalitogenic potential of these microbial-derived peptides. FINDINGS We analyzed 304,246 human microbiome genomes and 103 metagenomes collected from the MS cohort and identified 731 nonredundant analogs of myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). Of note, half of these analogs could bind to MHC II and interact with TCR through structural modeling of the interaction using fine-tuned AlphaFold. Among the 8 selected peptides, the peptide (P3) shows the ability to activate MOG35-55-specific CD4+ T cells in vitro. Furthermore, P3 shows encephalitogenic capacity and has the potential to induce EAE in some animals. Notably, mice immunized with a combination of P3 and MOG35-55 develop severe EAE. Additionally, dendritic cells could process and present P3 to MOG35-55-specific CD4+ T cells and activate these cells. INTERPRETATION Our data suggests the potential involvement of a MOG35-55-mimic peptide derived from the gut microbiota as a molecular trigger of EAE pathogenesis. Our findings offer direct evidence of how microbes can initiate the development of EAE, suggesting a potential explanation for the correlation between certain gut microorganisms and MS prevalence. FUNDING National Natural Science Foundation of China (82371350 to GY).
Collapse
Affiliation(s)
- Xin Ma
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Jian Zhang
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Qianling Jiang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yong-Xin Li
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
2
|
Jatczak-Pawlik I, Jurewicz A, Domowicz M, Ewiak-Paszyńska A, Stasiołek M. CHI3L1 in Multiple Sclerosis-From Bench to Clinic. Cells 2024; 13:2086. [PMID: 39768177 PMCID: PMC11674340 DOI: 10.3390/cells13242086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with a complex and not fully understood etiopathological background involving inflammatory and neurodegenerative processes. CHI3L1 has been implicated in pathological conditions such as inflammation, injury, and neurodegeneration, and is likely to play a role in the physiological development of the CNS. CHI3L1 is primarily produced by CNS macrophages, microglia, and activated astrocytes. The CHI3L1 expression pattern in MS lesions might support the important role of astrocytes in modulating inflammatory processes in this disease. The potential applications of CHI3L1 as a biomarker in MS are multifactorial. The measurement of CHI3L1 in body fluids might find its role in the early diagnosis of MS. In further stages, the monitoring of CHI3L1 levels might provide information on disease severity and progression, enabling a better adjustment of therapeutic strategies. Importantly, CHI3L1 might potentially serve as a marker of ongoing glial activation, reflecting the dynamic response of the CNS cells to the inflammatory processes in MS. Although preliminary findings have been promising, further research is needed to validate the utility of CHI3L1 measurements in the diagnosis and prediction of the progression of MS. Additionally, comparisons with other biomarkers might be useful in clinical practice.
Collapse
Affiliation(s)
- Izabela Jatczak-Pawlik
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland
| | - Anna Jurewicz
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland
| | - Małgorzata Domowicz
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland
| | - Alicja Ewiak-Paszyńska
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland
| | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland
| |
Collapse
|
3
|
Kolanek A, Cemaga R, Maciejczyk M. Role and Diagnostic Significance of Apolipoprotein D in Selected Neurodegenerative Disorders. Diagnostics (Basel) 2024; 14:2814. [PMID: 39767175 PMCID: PMC11675071 DOI: 10.3390/diagnostics14242814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
The World Health Organization in 2021 ranked Alzheimer's disease and other dementias as the seventh leading cause of death globally. Neurodegenerative disorders are progressive, intractable, and often fatal diseases. Early diagnosis may allow patients to enjoy prolonged survival with attenuated symptomatology because of early intervention. Hence, further research on finding non-invasive biomarkers of neurodegenerative diseases is warranted. Apolipoprotein D (ApoD) is a glycoprotein involved in lipid metabolism, oxidative stress regulation, and inflammation. It is expressed in various body fluids and regions of the central nervous system. ApoD's roles in neuroprotection, lipid transport, and anti-inflammatory processes are crucial as far as the prevention of neurodegenerative pathologies is concerned. This review aims to summarize the background knowledge on ApoD, and it covers studies indexed in the PubMed, Scopus, and Web of Science databases. It discusses the evidence for the multifaceted roles of ApoD in the mechanisms and pathogenesis of multiple sclerosis, Alzheimer's disease, and Parkinson's disease. ApoD may be a specific, sensitive, easily obtained, cost-effective biomarker for neurodegenerative diseases and its applications in diagnostic practices, treatment strategies, and advancing neurodegenerative disorders' management.
Collapse
Affiliation(s)
- Agata Kolanek
- Students’ Scientific Club “Biochemistry of Civilization Diseases” at the Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 15-233 Bialystok, Poland; (A.K.); (R.C.)
| | - Roman Cemaga
- Students’ Scientific Club “Biochemistry of Civilization Diseases” at the Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 15-233 Bialystok, Poland; (A.K.); (R.C.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 2c Mickiewicza Street, 15-233 Bialystok, Poland
| |
Collapse
|
4
|
Pressley KR, Schwegman L, De Oca Arena MM, Huizar CC, Zamvil SS, Forsthuber TG. HLA-transgenic mouse models to study autoimmune central nervous system diseases. Autoimmunity 2024; 57:2387414. [PMID: 39167553 PMCID: PMC11470778 DOI: 10.1080/08916934.2024.2387414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/23/2024]
Abstract
It is known that certain human leukocyte antigen (HLA) genes are associated with autoimmune central nervous system (CNS) diseases, such as multiple sclerosis (MS), but their exact role in disease susceptibility and etiopathogenesis remains unclear. The best studied HLA-associated autoimmune CNS disease is MS, and thus will be the primary focus of this review. Other HLA-associated autoimmune CNS diseases, such as autoimmune encephalitis and neuromyelitis optica will be discussed. The lack of animal models to accurately capture the complex human autoimmune response remains a major challenge. HLA transgenic (tg) mice provide researchers with powerful tools to investigate the underlying mechanisms promoting susceptibility and progression of HLA-associated autoimmune CNS diseases, as well as for elucidating the myelin epitopes potentially targeted by T cells in autoimmune disease patients. We will discuss the potential role(s) of autoimmune disease-associated HLA alleles in autoimmune CNS diseases and highlight information provided by studies using HLA tg mice to investigate the underlying pathological mechanisms and opportunities to use these models for development of novel therapies.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | | | - Carol Chase Huizar
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Scott S. Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
5
|
Munger KC, Rotstein DL. Racial and Ethnic Disease Phenotype Differences Are Driven by Genetics: Commentary. Mult Scler 2024; 30:11-12. [PMID: 39658903 PMCID: PMC11633066 DOI: 10.1177/13524585241301304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Affiliation(s)
| | - Dalia L Rotstein
- University of Toronto, Department of Medicine, Toronto, ON, Canada; St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
6
|
Abouelmagd ME, AbdelMeseh M, Hassan AA, Ali MA, Mohamed RG, Mady A, Hindawi MD, Meshref M. History of head trauma and the risk of multiple sclerosis: A systematic review and meta-analysis. Mult Scler Relat Disord 2024; 92:106183. [PMID: 39603062 DOI: 10.1016/j.msard.2024.106183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/31/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic disease of the central nervous system with unclear etiology involving genetic, environmental, and immunological factors. The potential link between head trauma and MS is controversial, with conflicting evidence. This systematic review and meta-analysis aim to assess the risk of developing MS following head trauma. METHODS A systematic search of electronic databases was conducted, including studies that investigated the risk of MS in individuals with a history of head trauma compared to those without. Observational studies, including cohort and case-control designs, were included. Data synthesis was conducted using RevMan software. GRADE was used to assess the certainty of evidence. RESULTS Fifteen studies comprising 1,619,640 participants were included in the meta-analysis. The overall odds of developing MS were significantly higher in the head trauma group compared to the control group (OR = 1.41;95 % CI = [1.23, 1.61]; P < 0.00001; I2 = 62 %). Sensitivity analyses based on the number of participants and quality further supported our results. Subgroup analysis showed that results remained consistent across different head trauma identification methods (P = 0.92), early age head trauma and head trauma defined as TBI, or concussion were also significant predictors of MS (P < 0.0001). Analysis of the number of hits suggested a dose-response relationship between the number of head injuries and the risk of MS. According to the GRADE, all outcomes were classified as low or very low certainty of the evidence. CONCLUSION This meta-analysis suggests that a history of head trauma may be associated with an increased risk of developing MS. Further research is warranted to support our findings and explore the mechanisms linking head trauma to MS.
Collapse
Affiliation(s)
| | | | - Atef A Hassan
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt; Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA.
| | | | - Rashad G Mohamed
- Manchester Program for Medical Education, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Abdelrahman Mady
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt; Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA.
| | - Mahmoud Diaa Hindawi
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt; Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
7
|
Briggs FBS. Racial and Ethnic Disease Phenotype Differences Are Driven by Genetics - Yes. Mult Scler 2024; 30:5-8. [PMID: 39658897 DOI: 10.1177/13524585241293683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Affiliation(s)
- Farren B S Briggs
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
8
|
Zoledziewska M, Lorefice L. Gut microbiota and multiple sclerosis: a potential diagnostic and prognostic marker? Neurodegener Dis Manag 2024; 14:189-192. [PMID: 39600266 PMCID: PMC11703474 DOI: 10.1080/17582024.2024.2435249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Magdalena Zoledziewska
- Institute of Genetic and Biomedical Research (IRGB), Italian National Research Council (CNR), Monserrato, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Center, ASL Cagliari, Department of Medical Sciences and Public Health, Binaghi Hospital, University of Cagliari, Cagliari, Italy
| |
Collapse
|
9
|
Pisa M, Watson JL, Spencer JI, Niblett G, Mahjoub Y, Lockhart A, Yates RL, Yee SA, Hadley G, Ruiz J, Esiri MM, Kessler B, Fischer R, DeLuca GC. A role for vessel-associated extracellular matrix proteins in multiple sclerosis pathology. Brain Pathol 2024; 34:e13263. [PMID: 38659387 PMCID: PMC11483522 DOI: 10.1111/bpa.13263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple sclerosis (MS) is unsurpassed for its clinical and pathological hetherogeneity, but the biological determinants of this variability are unknown. HLA-DRB1*15, the main genetic risk factor for MS, influences the severity and distribution of MS pathology. This study set out to unravel the molecular determinants of the heterogeneity of MS pathology in relation to HLA-DRB1*15 status. Shotgun proteomics from a discovery cohort of MS spinal cord samples segregated by HLA-DRB*15 status revealed overexpression of the extracellular matrix (ECM) proteins, biglycan, decorin, and prolargin in HLA-DRB*15-positive cases, adding to established literature on a role of ECM proteins in MS pathology that has heretofore lacked systematic pathological validation. These findings informed a neuropathological characterisation of these proteins in a large autopsy cohort of 41 MS cases (18 HLA-DRB1*15-positive and 23 HLA-DRB1*15-negative), and seven non-neurological controls on motor cortical, cervical and lumbar spinal cord tissue. Biglycan and decorin demonstrate a striking perivascular expression pattern in controls that is reduced in MS (-36.5%, p = 0.036 and - 24.7%, p = 0.039; respectively) in lesional and non-lesional areas. A concomitant increase in diffuse parenchymal accumulation of biglycan and decorin is seen in MS (p = 0.015 and p = 0.001, respectively), particularly in HLA-DRB1*15-positive cases (p = 0.007 and p = 0.046, respectively). Prolargin shows a faint parenchymal pattern in controls that is markedly increased in MS cases where a perivascular deposition pattern is observed (motor cortex +97.5%, p = 0.001; cervical cord +49.1%, p = 0.016). Our findings point to ECM proteins and the vascular interface playing a central role in MS pathology within and outside the plaque area. As ECM proteins are known potent pro-inflammatory molecules, their parenchymal accumulation may contribute to disease severity. This study brings to light novel factors that may contribute to the heterogeneity of the topographical variation of MS pathology.
Collapse
Affiliation(s)
- Marco Pisa
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | | - Jonathan I. Spencer
- Centre for Cardiovascular Medicine and DevicesWilliam Harvey Research Institute, Queen Mary University of LondonLondonUK
| | - Guy Niblett
- Oxford Medical SchoolUniversity of OxfordOxfordUK
| | - Yasamin Mahjoub
- Faculty of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Andrew Lockhart
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Richard L. Yates
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Sydney A. Yee
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Gina Hadley
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Jennifer Ruiz
- Mandell MS CenterTrinity Health of New EnglandHartfordConnecticutUSA
| | - Margaret M. Esiri
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Benedict Kessler
- Mass Spectrometry LaboratoryTarget Discovery Institute, University of OxfordOxfordUK
| | - Roman Fischer
- Mass Spectrometry LaboratoryTarget Discovery Institute, University of OxfordOxfordUK
| | - Gabriele C. DeLuca
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| |
Collapse
|
10
|
Robinson WH, Younis S, Love ZZ, Steinman L, Lanz TV. Epstein-Barr virus as a potentiator of autoimmune diseases. Nat Rev Rheumatol 2024; 20:729-740. [PMID: 39390260 DOI: 10.1038/s41584-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/12/2024]
Abstract
The Epstein-Barr virus (EBV) is epidemiologically associated with development of autoimmune diseases, including systemic lupus erythematosus, Sjögren syndrome, rheumatoid arthritis and multiple sclerosis. Although there is well-established evidence for this association, the underlying mechanistic basis remains incompletely defined. In this Review, we discuss the role of EBV infection as a potentiator of autoimmune rheumatic diseases. We review the EBV life cycle, viral transcription programmes, serological profiles and lytic reactivation. We discuss the epidemiological and mechanistic associations of EBV with systemic lupus erythematosus, Sjögren syndrome, rheumatoid arthritis and multiple sclerosis. We describe the potential mechanisms by which EBV might promote autoimmunity, including EBV nuclear antigen 1-mediated molecular mimicry of human autoantigens; EBV-mediated B cell reprogramming, including EBV nuclear antigen 2-mediated dysregulation of autoimmune susceptibility genes; EBV and host genetic factors, including the potential for autoimmunity-promoting strains of EBV; EBV immune evasion and insufficient host responses to control infection; lytic reactivation; and other mechanisms. Finally, we discuss the therapeutic implications and potential therapeutic approaches to targeting EBV for the treatment of autoimmune disease.
Collapse
Affiliation(s)
- William H Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA.
- VA Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Shady Younis
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Zelda Z Love
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences and Paediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tobias V Lanz
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Drosu N, Anderson M, Bilodeau PA, Nishiyama S, Mikami T, Bobrowski-Khoury N, Cabot J, Housman D, Levy M. CD4 T cells restricted to DRB1*15:01 recognize two Epstein-Barr virus glycoproteins capable of intracellular antigen presentation. Proc Natl Acad Sci U S A 2024; 121:e2416097121. [PMID: 39432795 PMCID: PMC11536159 DOI: 10.1073/pnas.2416097121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Both genetic and environmental factors contribute to multiple sclerosis (MS) risk. Infection with the Epstein-Barr virus (EBV) is the strongest environmental risk factor, and HLA-DR15 is the strongest genetic risk factor for MS. We employed computational methods and in vitro assays for CD4 T cell activation to investigate the DR15-restricted response to EBV. Using a machine learning-based HLA ligand predictor, the EBV glycoprotein B (gB) was predicted to be enriched in epitopes restricted to presentation by DRB1*15:01. In DR15-positive individuals, two epitopes comprised the major CD4 T cell response to gB. Surprisingly, the expression of recombinant gB in a DR15-homozygous B cell line or primary autologous B cells elicited a CD4 T cell response, indicating that intracellular gB was loaded onto HLA class II molecules. By deleting the signal sequence of gB, we determined that this pathway for direct activation of CD4 T cells was dependent on trafficking to the endoplasmic reticulum (ER) within the B cell. We screened seven recombinant EBV antigens from the ER compartment for immune responses in DR15-negative vs. DR15-homozygous individuals. In addition to gB, gH was a key CD4 T cell target in individuals homozygous for DR15. Compared to non-DR15 controls, DR15-homozygotes had significantly higher T cell responses to both gB and gH but not to EBV latent or lytic antigens overall. Responses to gB and gH were slightly elevated in DR15 homozygotes with MS. Our results link MS environmental and genetic risk factors by demonstrating that HLA-DR15 dictates CD4 T cell immunity to EBV antigens.
Collapse
Affiliation(s)
- Natalia Drosu
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Monique Anderson
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Philippe A. Bilodeau
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Shuhei Nishiyama
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Takahisa Mikami
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Natasha Bobrowski-Khoury
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Jackson Cabot
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - David Housman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Michael Levy
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| |
Collapse
|
12
|
Mittl K, Hayashi F, Dandekar R, Schubert RD, Gerdts J, Oshiro L, Loudermilk R, Greenfield A, Augusto DG, Ramesh A, Tran E, Koshal K, Kizer K, Dreux J, Cagalingan A, Schustek F, Flood L, Moore T, Kirkemo LL, Cooper T, Harms M, Gomez R, Sibener L, Cree BAC, Hauser SL, Hollenbach JA, Gee M, Wilson MR, Zamvil SS, Sabatino JJ. Antigen specificity of clonally-enriched CD8+ T cells in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611010. [PMID: 39282370 PMCID: PMC11398516 DOI: 10.1101/2024.09.07.611010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
CD8+ T cells are the dominant lymphocyte population in multiple sclerosis (MS) lesions where they are highly clonally expanded. The clonal identity, function, and antigen specificity of CD8+ T cells in MS are not well understood. Here we report a comprehensive single-cell RNA-seq and T cell receptor (TCR)-seq analysis of the cerebrospinal fluid (CSF) and blood from a cohort of treatment-naïve MS patients and control participants. A small subset of highly expanded and activated CD8+ T cells were enriched in the CSF in MS that displayed high activation, cytotoxicity and tissue-homing transcriptional profiles. Using a combination of unbiased and targeted antigen discovery approaches, MS-derived CD8+ T cell clonotypes recognizing Epstein-Barr virus (EBV) antigens and multiple novel mimotopes were identified. These findings shed vital insight into the role of CD8+ T cells in MS and pave the way towards disease biomarkers and therapeutic targets.
Collapse
|
13
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
14
|
Golikova EA, Alshevskaya AA, Alrhmoun S, Sivitskaya NA, Sennikov SV. TCR-T cell therapy: current development approaches, preclinical evaluation, and perspectives on regulatory challenges. J Transl Med 2024; 22:897. [PMID: 39367419 PMCID: PMC11451006 DOI: 10.1186/s12967-024-05703-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
TCR-T cell therapy represents a promising advancement in adoptive immunotherapy for cancer treatment. Despite its potential, the development and preclinical testing of TCR-T cells face significant challenges. This review provides a structured overview of the key stages in preclinical testing, including in silico, in vitro, and in vivo methods, within the context of the sequential development of novel therapies. This review aimed to systematically outline the processes for evaluating TCR-T cells at each stage: from in silico approaches used to predict target antigens, assess cross-reactivity, and minimize off-target effects, to in vitro assays designed to measure cell functionality, cytotoxicity, and activation. Additionally, the review discusses the limitations of in vivo testing in animal models, particularly in accurately reflecting the human tumor microenvironment and immune responses. Performed analysis emphasizes the importance of these preclinical stages in the safe and effective development of TCR-T cell therapies. While current models provide valuable insights, we identify critical gaps, particularly in in vivo biodistribution and toxicity assessments, and propose the need for enhanced standardization and the development of more representative models. This structured approach aims to improve the predictability and safety of TCR-T cell therapy as it advances towards clinical application.
Collapse
Affiliation(s)
- Elena A Golikova
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
| | - Alina A Alshevskaya
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia.
| | - Saleh Alrhmoun
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
- Federal State Budgetary Scientific Institution, "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099, Novosibirsk, Russia
| | - Natalia A Sivitskaya
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
| | - Sergey V Sennikov
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
- Federal State Budgetary Scientific Institution, "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099, Novosibirsk, Russia
| |
Collapse
|
15
|
Xu P, Li Y, Zhuang X, Yue L, Ma Y, Xue W, Ji L, Zhan Y, Ou Y, Qiao T, Wu D, Liu P, Chen H, Cheng Y. Changes in immune subsets during chemotherapy as prognosis biomarkers for multiple myeloma patients by longitudinal monitoring. Immunol Res 2024; 72:1185-1197. [PMID: 39254909 DOI: 10.1007/s12026-024-09521-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
Multiple myeloma (MM) is a malignancy of plasma cells accompanied by immune dysfunction. This study aimed to provide a comprehensive and dynamic characterization of the peripheral immune environment in MM patients and find its diagnostic and prognostic values for therapy. The peripheral immune profiles of MM inpatients and healthy controls were assessed by flow cytometry. A longitudinal study of immune subsets was observed during cycles of chemotherapy. The diagnostic and prognostic models were established based on immune subsets by the absolute shrinkage and selection operator (LASSO) and multivariate regression. MM patients possessed an impeded immune landscape, including reduced activation of B cells, increased effective T cells and regulatory T cells (Tregs), augmented CD16 expression on monocytes and dendritic cell percentages, decreased CD56dimCD16+ natural killer cells (NKs), and amplified CD56bright and HLA-DR+ natural killer T cells (NKTs). Chemotherapy has different dynamic effects on specific cells, of which 2 cycles is the key turning point. NKT, dendritic cells, naïve Tc and Th cells, HLA-DR+ Tc cells, CD56dim NKTs, CD16++ monocytes, and CD25+ B cells could have the diagnostic value, and a prognostic model including neutrophils, naïve Tc cells, CD56brightCD16dim NKs, and CD16+ dendritic cells was established with acceptable accuracy. Our data showed dynamic and abnormal peripheral immune profiles in MM patients, which had prognostic values and could provide the basis for clinical therapy.
Collapse
Affiliation(s)
- Pengcheng Xu
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital QingPu Branch, Fudan University, Shanghai, China
| | - Ying Li
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xibing Zhuang
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Yue
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanna Ma
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Wenjin Xue
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lili Ji
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanxia Zhan
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Ou
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tiankui Qiao
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Duojiao Wu
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hao Chen
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Thoracic Surgery, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Yunfeng Cheng
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Guan D, Li Y, Zhao X, Wang K, Guo Y, Dong N, Cui Y, Gao Y, Wang M, Wang J, Ren Y, Shang P, Liu Y. Hederagenol improves multiple sclerosis by modulating Th17 cell differentiation. IUBMB Life 2024; 76:845-857. [PMID: 38838376 DOI: 10.1002/iub.2863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Abstract
Multiple sclerosis (MS) is a common autoimmune illness that is difficult to treat. The upregulation of Th17 cells is critical in the pathological process of MS. Hederagenol (Hed) has been shown to lower IL-17 levels, although its role in MS pathophysiology is uncertain. In this study, we explore whether Hed could ameliorate MS by modulating Th17 cell differentiation, with the goal of identifying new treatment targets for MS. The experimental autoimmune encephalomyelitis (EAE) mouse model was conducted and Hed was intraperitoneally injected into mice. The weight was recorded and the clinical symptom grade was assessed. Hematoxylin-eosin staining was carried out to determine the extent of inflammation in the spinal cord and liver. The luxol Fast Blue staining was performed to detect the pathological changes in the myelin sheath. Nerve damage was detected using NeuN immunofluorescence staining and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. Immunohistology approaches were used to study alterations in immune cells in the spinal cord. The proportions of T cell subsets in the spleens were analyzed by flow cytometry. RORγt levels were measured using quantitative real-time PCR or Western blot. The activity of the RORγt promoter was analyzed by Chromatin immunoprecipitation. Hed administration reduced the clinical symptom grade of EAE mice, as well as the inflammatory infiltration, demyelination, and cell disorder of the spinal cord, while having no discernible effect on the mouse weight. In addition, Hed treatment significantly reduced the number of T cells, particularly Th17 cells in the spinal cord and spleen-isolated CD4+ T cells. Hed lowered the RORγt levels in spleens and CD4+ T cells and overexpression of RORγt reversed the inhibitory effect of Hed on Th17 differentiation. Hed decreased nerve injury by modulating Th17 differentiation through the RORγt promoter. Hed regulates Th17 differentiation by reducing RORγt promoter activity, which reduces nerve injury and alleviates EAE.
Collapse
MESH Headings
- Animals
- Th17 Cells/immunology
- Th17 Cells/drug effects
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Cell Differentiation/drug effects
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/pathology
- Multiple Sclerosis/immunology
- Mice
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Female
- Oleanolic Acid/analogs & derivatives
- Oleanolic Acid/pharmacology
- Mice, Inbred C57BL
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Spinal Cord/immunology
- Interleukin-17/metabolism
- Interleukin-17/genetics
Collapse
Affiliation(s)
- Dongsheng Guan
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yingxia Li
- The College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xu Zhao
- Department of Pharmacy, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Kun Wang
- Department of Pharmacy, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yanke Guo
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ning Dong
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinglin Cui
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yinghe Gao
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Mengmeng Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jing Wang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yihan Ren
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Penghui Shang
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yuxuan Liu
- Department of Neurology, the Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
17
|
Faust MA, Gibbs L, Oviedo JM, Cornwall DH, Fairfax KC, Zhou Z, Lamb TJ, Evavold BD. B Cells Influence Encephalitogenic T Cell Frequency to Myelin Oligodendrocyte Glycoprotein (MOG)38-49 during Full-length MOG Protein-Induced Demyelinating Disease. Immunohorizons 2024; 8:729-739. [PMID: 39330967 PMCID: PMC11447661 DOI: 10.4049/immunohorizons.2400069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Although T cells are encephalitogenic during demyelinating disease, B cell-depleting therapies are a successful treatment for patients with multiple sclerosis. Murine models of demyelinating disease utilizing myelin epitopes, such as myelin oligodendrocyte glycoprotein (MOG)35-55, induce a robust CD4 T cell response but mitigate the contribution of pathological B cells. This limits their efficacy for investigating how B cell depletion affects T cells. Furthermore, induction of experimental autoimmune encephalomyelitis with a single CD4 T cell epitope does not reflect the breadth of epitopes observed in the clinic. To better model the adaptive immune response, mice were immunized with the full-length MOG protein or the MOG1-125 extracellular domain (ECD) and compared with MOG35-55. Mature MOG-reactive B cells were generated only by full-length MOG or ECD. The CNS-localized T cell response induced by full-length MOG is characterized by a reduction in frequency and the percentage of low-affinity T cells with reactivity toward the core epitope of MOG35-55. B cell depletion with anti-CD20 before full-length MOG-induced, but not ECD-induced, demyelinating disease restored T cell reactivity toward the immunodominant epitope of MOG35-55, suggesting the B cell-mediated control of encephalitogenic epitopes. Ultimately, this study reveals that anti-CD20 treatment can influence T cell epitopes found in the CNS during demyelinating disease.
Collapse
Affiliation(s)
- Michael A. Faust
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Lisa Gibbs
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Juan M. Oviedo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Douglas H. Cornwall
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Keke C. Fairfax
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Zemin Zhou
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Tracey J. Lamb
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Brian D. Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| |
Collapse
|
18
|
Steimle A, Neumann M, Grant ET, Willieme S, De Sciscio A, Parrish A, Ollert M, Miyauchi E, Soga T, Fukuda S, Ohno H, Desai MS. Gut microbial factors predict disease severity in a mouse model of multiple sclerosis. Nat Microbiol 2024; 9:2244-2261. [PMID: 39009690 PMCID: PMC11371644 DOI: 10.1038/s41564-024-01761-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 06/14/2024] [Indexed: 07/17/2024]
Abstract
Gut bacteria are linked to neurodegenerative diseases but the risk factors beyond microbiota composition are limited. Here we used a pre-clinical model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), to identify microbial risk factors. Mice with different genotypes and complex microbiotas or six combinations of a synthetic human microbiota were analysed, resulting in varying probabilities of severe neuroinflammation. However, the presence or relative abundances of suspected microbial risk factors failed to predict disease severity. Akkermansia muciniphila, often associated with MS, exhibited variable associations with EAE severity depending on the background microbiota. Significant inter-individual disease course variations were observed among mice harbouring the same microbiota. Evaluation of microbial functional characteristics and host immune responses demonstrated that the immunoglobulin A coating index of certain bacteria before disease onset is a robust individualized predictor of disease development. Our study highlights the need to consider microbial community networks and host-specific bidirectional interactions when aiming to predict severity of neuroinflammation.
Collapse
Affiliation(s)
- Alex Steimle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mareike Neumann
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stéphanie Willieme
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alessandro De Sciscio
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Eiji Miyauchi
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
19
|
Marti Z, Ruder J, Thomas OG, Bronge M, De La Parra Soto L, Grönlund H, Olsson T, Martin R. Enhanced and cross-reactive in vitro memory B cell response against Epstein-Barr virus nuclear antigen 1 in multiple sclerosis. Front Immunol 2024; 15:1334720. [PMID: 39257578 PMCID: PMC11385009 DOI: 10.3389/fimmu.2024.1334720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/04/2024] [Indexed: 09/12/2024] Open
Abstract
Multiple sclerosis (MS) is a prototypical autoimmune disease of the central nervous system (CNS). In addition to CD4+ T cells, memory B cells are now recognized as a critical cell type in the disease. This is underlined by the fact that the best-characterized environmental risk factor for MS is the Epstein-Barr virus (EBV), which can infect and persist in memory B cells throughout life. Several studies have identified changes in anti-EBV immunity in patients with MS. Examples include elevated titers of anti-EBV nuclear antigen 1 (EBNA1) antibodies, interactions of these with the MS-associated HLA-DR15 haplotype, and molecular mimicry with MS autoantigens like myelin basic protein (MBP), anoctamin-2 (ANO2), glial cell adhesion molecule (GlialCAM), and alpha-crystallin B (CRYAB). In this study, we employ a simple in vitro assay to examine the memory B cell antibody repertoire in MS patients and healthy controls. We replicate previous serological data from MS patients demonstrating an increased secretion of anti-EBNA1380-641 IgG in cell culture supernatants, as well as a positive correlation of these levels with autoantibodies against GlialCAM262-416 and ANO21-275. For EBNA1380-641 and ANO21-275, we provide additional evidence suggesting antibody cross-reactivity between the two targets. Further, we show that two efficacious MS treatments - natalizumab (NAT) and autologous hematopoietic stem cell transplantation (aHSCT) - are associated with distinct changes in the EBNA1-directed B cell response and that these alterations can be attributed to the unique mechanisms of action of these therapies. Using an in vitro system, our study confirms MS-associated changes in the anti-EBNA1 memory B cell response, EBNA1380-641 antibody cross-reactivity with ANO21-275, and reveals treatment-associated changes in the immunoglobulin repertoire in MS.
Collapse
Affiliation(s)
- Zoe Marti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Josefine Ruder
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Olivia G Thomas
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Bronge
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo De La Parra Soto
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hans Grönlund
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neurocience, Karolinska Institutet, Stockholm, Sweden
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Zancan V, Nasello M, Bigi R, Reniè R, Buscarinu MC, Mechelli R, Ristori G, Salvetti M, Bellucci G. Gut Microbiota Composition Is Causally Linked to Multiple Sclerosis: A Mendelian Randomization Analysis. Microorganisms 2024; 12:1476. [PMID: 39065244 PMCID: PMC11278727 DOI: 10.3390/microorganisms12071476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Accumulating evidence links the microbial communities inhabiting the gut to the pathophysiological processes underlying multiple sclerosis (MS). However, most studies on the microbiome in MS are correlative in nature, thus being at risk of confounding and reverse causality. Mendelian randomization (MR) analyses allow the estimation of the causal relationship between a risk factor and an outcome of interest using genetic variants as proxies for environmental exposures. Here, we performed a two-sample MR to assess the causality between the gut microbiome and MS. We extracted genetic instruments from summary statistics from three large genome-wide association studies (GWASs) on the gut microbiome (18,340, 8959, and 7738 subjects). The exposure data were derived from the latest GWAS on MS susceptibility (47,429 patients and 68,374 controls). We pinpointed several microbial strains whose abundance is linked with enhanced MS risk (Actinobacteria class, Bifidobacteriaceae family, Lactobacillus genus) or protection (Prevotella spp., Lachnospiranaceae genus, Negativibacillus genus). The largest risk effect was seen for Ruminococcus Torques (OR, 2.89, 95% C.I. 1.67-5, p = 1.51 × 10-4), while Akkermansia municiphila emerged as strongly protective (OR, 0.43, 95% C.I. 0.32-0.57, p = 1.37 × 10-8). Our findings support a causal relationship between the gut microbiome and MS susceptibility, reinforcing the relevance of the microbiome-gut-brain axis in disease etiology, opening wider perspectives on host-environmental interactions for MS prevention.
Collapse
Affiliation(s)
- Valeria Zancan
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Martina Nasello
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Rachele Bigi
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Roberta Reniè
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Chiara Buscarinu
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Rosella Mechelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00163 Rome, Italy
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giovanni Ristori
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, 00179 Rome, Italy
| | - Marco Salvetti
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy
| | - Gianmarco Bellucci
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
21
|
Kohlgruber AC, Dezfulian MH, Sie BM, Wang CI, Kula T, Laserson U, Larman HB, Elledge SJ. High-throughput discovery of MHC class I- and II-restricted T cell epitopes using synthetic cellular circuits. Nat Biotechnol 2024:10.1038/s41587-024-02248-6. [PMID: 38956325 DOI: 10.1038/s41587-024-02248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/16/2024] [Indexed: 07/04/2024]
Abstract
Antigen discovery technologies have largely focused on major histocompatibility complex (MHC) class I-restricted human T cell receptors (TCRs), leaving methods for MHC class II-restricted and mouse TCR reactivities relatively undeveloped. Here we present TCR mapping of antigenic peptides (TCR-MAP), an antigen discovery method that uses a synthetic TCR-stimulated circuit in immortalized T cells to activate sortase-mediated tagging of engineered antigen-presenting cells (APCs) expressing processed peptides on MHCs. Live, tagged APCs can be directly purified for deconvolution by sequencing, enabling TCRs with unknown specificity to be queried against barcoded peptide libraries in a pooled screening context. TCR-MAP accurately captures self-reactivities or viral reactivities with high throughput and sensitivity for both MHC class I-restricted and class II-restricted TCRs. We elucidate problematic cross-reactivities of clinical TCRs targeting the cancer/testis melanoma-associated antigen A3 and discover targets of myocarditis-inciting autoreactive T cells in mice. TCR-MAP has the potential to accelerate T cell antigen discovery efforts in the context of cancer, infectious disease and autoimmunity.
Collapse
Affiliation(s)
- Ayano C Kohlgruber
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Genetics, Harvard University Medical School, Boston, MA, USA
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Mohammad H Dezfulian
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Genetics, Harvard University Medical School, Boston, MA, USA
| | - Brandon M Sie
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Genetics, Harvard University Medical School, Boston, MA, USA
| | - Charlotte I Wang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Genetics, Harvard University Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Tomasz Kula
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Genetics, Harvard University Medical School, Boston, MA, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Uri Laserson
- Department of Genetics and Genomic Sciences and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - H Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephen J Elledge
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Genetics, Harvard University Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
22
|
Abbadessa G, Lepore MT, Bruzzaniti S, Piemonte E, Miele G, Signoriello E, Perna F, De Falco C, Lus G, Matarese G, Bonavita S, Galgani M. Ocrelizumab Alters Cytotoxic Lymphocyte Function While Reducing EBV-Specific CD8 + T-Cell Proliferation in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200250. [PMID: 38662990 PMCID: PMC11087045 DOI: 10.1212/nxi.0000000000200250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND AND OBJECTIVES The role of B cells in the pathogenic events leading to relapsing multiple sclerosis (R-MS) has only been recently elucidated. A pivotal step in defining this role has been provided by therapeutic efficacy of anti-CD20 monoclonal antibodies. Indeed, treatment with anti-CD20 can also alter number and function of other immune cells not directly expressing CD20 on their cell surface, whose activities can contribute to unknown aspects influencing therapeutic efficacy. We examined the phenotype and function of cytotoxic lymphocytes and Epstein-Barr virus (EBV)-specific immune responses in people with R-MS before and after ocrelizumab treatment. METHODS In this prospective study, we collected blood samples from people with R-MS (n = 41) before and 6 and 12 months after initiating ocrelizumab to assess the immune phenotype and the indirect impact on cytotoxic functions of CD8+ T and NK cells. In addition, we evaluated the specific anti-EBV proliferative responses of both CD8+ T and NK lymphocytes as surrogate markers of anti-EBV activity. RESULTS We observed that while ocrelizumab depleted circulating B cells, it also reduced the expression of activation and migratory markers on both CD8+ T and NK cells as well as their in vitro cytotoxic activity. A comparable pattern in the modulation of immune molecules by ocrelizumab was observed in cytotoxic cells even when patients with R-MS were divided into groups based on their prior disease-modifying treatment. These effects were accompanied by a significant and selective reduction of CD8+ T-cell proliferation in response to EBV antigenic peptides. DISCUSSION Taken together, our findings suggest that ocrelizumab-while depleting B cells-affects the cytotoxic function of CD8+ and NK cells, whose reduced cross-activity against myelin antigens might also contribute to its therapeutic efficacy during MS.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Female
- Adult
- Male
- Herpesvirus 4, Human/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Middle Aged
- Immunologic Factors/pharmacology
- Multiple Sclerosis, Relapsing-Remitting/drug therapy
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/blood
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Prospective Studies
- Cell Proliferation/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lymphocyte Activation/drug effects
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Maria Teresa Lepore
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Sara Bruzzaniti
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Erica Piemonte
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppina Miele
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Elisabetta Signoriello
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Francesco Perna
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Chiara De Falco
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - G Lus
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppe Matarese
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Simona Bonavita
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Mario Galgani
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| |
Collapse
|
23
|
Jin H, Chen Y, Zhang D, Lin J, Huang S, Wu X, Deng W, Huang J, Yao Y. YTHDF2 favors protumoral macrophage polarization and implies poor survival outcomes in triple negative breast cancer. iScience 2024; 27:109902. [PMID: 38812540 PMCID: PMC11134561 DOI: 10.1016/j.isci.2024.109902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Patients with triple-negative breast cancer (TNBC) frequently experience resistance to chemotherapy, leading to recurrence. The approach of optimizing anti-tumoral immunological effect is promising in overcoming such resistance, given the heterogeneity and lack of biomarkers in TNBC. In this study, we focused on YTHDF2, an N6-methyladenosine (m6A) RNA-reader protein, in macrophages, one of the most abundant intra-tumoral immune cells. Using single-cell sequencing and ex vivo experiments, we discovered that YTHDF2 significantly promotes pro-tumoral phenotype polarization of macrophages and is closely associated with down-regulated antigen-presentation signaling to other immune cells in TNBC. The in vitro deprivation of YTHDF2 favors anti-tumoral effect. Expressions of multiple transcription factors, especially SPI1, were consistently observed in YTHDF2-high macrophages, providing potential therapeutic targets for new strategies. In conclusion, YTHDF2 in macrophages appears to promote pro-tumoral effects while suppressing immune activity, indicating the treatment targeting YTHDF2 or its transcription factors could be a promising strategy for chemoresistant TNBC.
Collapse
Affiliation(s)
- Hao Jin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Yue Chen
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Dongbo Zhang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Junfan Lin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Songyin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Xiaohua Wu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Wen Deng
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Jiandong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province 518055, China
- Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province 516621, China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province 516621, China
| |
Collapse
|
24
|
Van Der Byl W, Nüssing S, Peters TJ, Ahn A, Li H, Ledergor G, David E, Koh AS, Wagle MV, Deguit CDT, de Menezes MN, Travers A, Sampurno S, Ramsbottom KM, Li R, Kallies A, Beavis PA, Jungmann R, Bastings MMC, Belz GT, Goel S, Trapani JA, Crabtree GR, Chang HY, Amit I, Goodnow CC, Luciani F, Parish IA. The CD8 + T cell tolerance checkpoint triggers a distinct differentiation state defined by protein translation defects. Immunity 2024; 57:1324-1344.e8. [PMID: 38776918 DOI: 10.1016/j.immuni.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/01/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Peripheral CD8+ T cell tolerance is a checkpoint in both autoimmune disease and anti-cancer immunity. Despite its importance, the relationship between tolerance-induced states and other CD8+ T cell differentiation states remains unclear. Using flow cytometric phenotyping, single-cell RNA sequencing (scRNA-seq), and chromatin accessibility profiling, we demonstrated that in vivo peripheral tolerance to a self-antigen triggered a fundamentally distinct differentiation state separate from exhaustion, memory, and functional effector cells but analogous to cells defectively primed against tumors. Tolerant cells diverged early and progressively from effector cells, adopting a transcriptionally and epigenetically distinct state within 60 h of antigen encounter. Breaching tolerance required the synergistic actions of strong T cell receptor (TCR) signaling and inflammation, which cooperatively induced gene modules that enhanced protein translation. Weak TCR signaling during bystander infection failed to breach tolerance due to the uncoupling of effector gene expression from protein translation. Thus, tolerance engages a distinct differentiation trajectory enforced by protein translation defects.
Collapse
Affiliation(s)
- Willem Van Der Byl
- The Kirby Institute for Infection and Immunity, UNSW, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia
| | - Simone Nüssing
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Timothy J Peters
- Garvan Institute of Medical Research, Sydney, NSW, Australia; University of New South Wales Sydney, Sydney, NSW, Australia
| | - Antonio Ahn
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Hanjie Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Guy Ledergor
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Andrew S Koh
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Mayura V Wagle
- Garvan Institute of Medical Research, Sydney, NSW, Australia; John Curtin School of Medical Research, ANU, Canberra, ACT, Australia
| | | | - Maria N de Menezes
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Avraham Travers
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shienny Sampurno
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kelly M Ramsbottom
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany; Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Maartje M C Bastings
- Institute of Materials, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Interfaculty Bioengineering Institute, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gabrielle T Belz
- The Frazer Institute, The University of Queensland, Brisbane, QLD, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Shom Goel
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Gerald R Crabtree
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA; Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chris C Goodnow
- School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Fabio Luciani
- The Kirby Institute for Infection and Immunity, UNSW, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, UNSW, Sydney, NSW, Australia.
| | - Ian A Parish
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; John Curtin School of Medical Research, ANU, Canberra, ACT, Australia.
| |
Collapse
|
25
|
Thomas OG, Haigh TA, Croom-Carter D, Leese A, Van Wijck Y, Douglas MR, Rickinson A, Brooks JM, Taylor GS. Heightened Epstein-Barr virus immunity and potential cross-reactivities in multiple sclerosis. PLoS Pathog 2024; 20:e1012177. [PMID: 38843296 PMCID: PMC11156336 DOI: 10.1371/journal.ppat.1012177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/08/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is a likely prerequisite for multiple sclerosis (MS) but the underlying mechanisms are unknown. We investigated antibody and T cell responses to EBV in persons with MS (pwMS), healthy EBV-seropositive controls (HC) and post-infectious mononucleosis (POST-IM) individuals up to 6 months after disease resolution. The ability of EBV-specific T cell responses to target antigens from the central nervous system (CNS) was also investigated. METHODS Untreated persons with relapsing-remitting MS, POST-IM individuals and HC were, as far as possible, matched for gender, age and HLA-DRB1*15:01. EBV load was determined by qPCR, and IgG responses to key EBV antigens were determined by ELISA, immunofluorescence and Western blot, and tetanus toxoid antibody responses by multiplex bead array. EBV-specific T cell responses were determined ex vivo by intracellular cytokine staining (ICS) and cross-reactivity of in vitro-expanded responses probed against 9 novel Modified Vaccinia Ankara (MVA) viruses expressing candidate CNS autoantigens. RESULTS EBV load in peripheral blood mononuclear cells (PBMC) was unchanged in pwMS compared to HC. Serologically, while tetanus toxoid responses were unchanged between groups, IgG responses to EBNA1 and virus capsid antigen (VCA) were significantly elevated (EBNA1 p = 0.0079, VCA p = 0.0298) but, importantly, IgG responses to EBNA2 and the EBNA3 family antigens were also more frequently detected in pwMS (EBNA2 p = 0.042 and EBNA3 p = 0.005). In ex vivo assays, T cell responses to autologous EBV-transformed B cells and to EBNA1 were largely unchanged numerically, but significantly increased IL-2 production was observed in response to certain stimuli in pwMS. EBV-specific polyclonal T cell lines from both MS and HC showed high levels of autoantigen recognition by ICS, and several neuronal proteins emerged as common targets including MOG, MBP, PLP and MOBP. DISCUSSION Elevated serum EBV-specific antibody responses in the MS group were found to extend beyond EBNA1, suggesting a larger dysregulation of EBV-specific antibody responses than previously recognised. Differences in T cell responses to EBV were more difficult to discern, however stimulating EBV-expanded polyclonal T cell lines with 9 candidate CNS autoantigens revealed a high level of autoreactivity and indicate a far-reaching ability of the virus-induced T cell compartment to damage the CNS.
Collapse
Affiliation(s)
- Olivia G. Thomas
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Tracey A. Haigh
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Deborah Croom-Carter
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Alison Leese
- School of Biological Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Yolanda Van Wijck
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Michael R. Douglas
- Dudley Group of Hospitals NHS Foundation Trust, Dudley, United Kingdom
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alan Rickinson
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Jill M. Brooks
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Graham S. Taylor
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| |
Collapse
|
26
|
Neidhart S, Vlad B, Hilty M, Högelin KA, Ziegler M, Berenjeno-Correa E, Reichen I, Stridh P, Jelcic I, Khademi M, Kockum I, Sospedra M, Al Nimer F, Martin R, Jelcic I. HLA Associations of Intrathecal IgG Production against Specific Viruses in Multiple Sclerosis. Ann Neurol 2024; 95:1112-1126. [PMID: 38551149 DOI: 10.1002/ana.26921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/05/2024] [Accepted: 03/04/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE Specific human leucocyte antigen (HLA) alleles are not only associated with higher risk to develop multiple sclerosis (MS) and other autoimmune diseases, but also with the severity of various viral and bacterial infections. Here, we analyzed the most specific biomarker for MS, that is, the polyspecific intrathecal IgG antibody production against measles, rubella, and varicella zoster virus (MRZ reaction), for possible HLA associations in MS. METHODS We assessed MRZ reaction from 184 Swiss patients with MS and clinically isolated syndrome (CIS) and 89 Swiss non-MS/non-CIS control patients, and performed HLA sequence-based typing, to check for associations of positive MRZ reaction with the most prevalent HLA alleles. We used a cohort of 176 Swedish MS/CIS patients to replicate significant findings. RESULTS Whereas positive MRZ reaction showed a prevalence of 38.0% in MS/CIS patients, it was highly specific (97.7%) for MS/CIS. We identified HLA-DRB1*15:01 and other tightly linked alleles of the HLA-DR15 haplotype as the strongest HLA-encoded risk factors for a positive MRZ reaction in Swiss MS/CIS (odds ratio [OR], 3.90, 95% confidence interval [CI] 2.05-7.46, padjusted = 0.0004) and replicated these findings in Swedish MS/CIS patients (OR 2.18, 95%-CI 1.16-4.02, padjusted = 0.028). In addition, female MS/CIS patients had a significantly higher probability for a positive MRZ reaction than male patients in both cohorts combined (padjusted <0.005). INTERPRETATION HLA-DRB1*15:01, the strongest genetic risk factor for MS, and female sex, 1 of the most prominent demographic risk factors for developing MS, predispose in MS/CIS patients for a positive MRZ reaction, the most specific CSF biomarker for MS. ANN NEUROL 2024;95:1112-1126.
Collapse
Affiliation(s)
- Stephan Neidhart
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Benjamin Vlad
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Marc Hilty
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Klara Asplund Högelin
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mario Ziegler
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Ernesto Berenjeno-Correa
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Ina Reichen
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Pernilla Stridh
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Mohsen Khademi
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP) PrecisionMS of the University of Zurich, Zurich, Switzerland
| | - Faiez Al Nimer
- Center for Molecular Medicine, Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP) PrecisionMS of the University of Zurich, Zurich, Switzerland
- Center for Molecular Medicine, Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich and University Hospital, Zurich, Switzerland
- Clinical Research Priority Program MS (CRPP) PrecisionMS of the University of Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Johansson E, Alfredsson L, Strid P, Kockum I, Olsson T, Hedström AK. Head trauma results in manyfold increased risk of multiple sclerosis in genetically susceptible individuals. J Neurol Neurosurg Psychiatry 2024; 95:554-560. [PMID: 38212058 PMCID: PMC11103305 DOI: 10.1136/jnnp-2023-332643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Large register-based studies have reported an association between head trauma and increased risk of multiple sclerosis (MS). We aimed to investigate possible interactions between head trauma and MS-associated HLA genes in relation to MS risk. METHODS We used a Swedish population-based case-control study (2807 incident cases, 5950 matched controls with HLA genotypes available for 2057 cases, 2887 controls). Subjects with and without a history of self-reported head trauma were compared regarding MS risk, by calculating ORs with 95% CIs using logistic regression models. Additive interaction between head trauma, HLA-DRB1*1501 and absence of HLA-A*0201, was assessed by calculating the attributable proportion (AP) due to interaction. RESULTS A history of head trauma was associated with a 30% increased risk of subsequently developing MS (OR 1.34, 95% CI 1.17 to 1.53), with a trend showing increased risk of MS with increasing number of head impacts (p=0.03). We observed synergistic effects between recent head trauma and HLA-DRB1*15:01 as well as absence of HLA*02:01 in relation to MS risk (each AP 0.40, 95% CI 0.1 to 0.7). Recent head trauma in individuals with both genetic risk factors rendered an 18-fold increased risk of MS, compared with those with neither the genetic risk factors nor a history of head trauma (OR 17.7, 95% CI 7.13 to 44.1). CONCLUSIONS Our findings align with previous observations of a dose-dependent association between head trauma and increased risk of MS and add a novel aspect of this association by revealing synergistic effects between recent head trauma and MS-associated HLA genes.
Collapse
Affiliation(s)
- Eva Johansson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Alfredsson
- Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Pernilla Strid
- Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Cortese M, Leng Y, Bjornevik K, Mitchell M, Healy BC, Mina MJ, Mancuso JD, Niebuhr DW, Munger KL, Elledge SJ, Ascherio A. Serologic Response to the Epstein-Barr Virus Peptidome and the Risk for Multiple Sclerosis. JAMA Neurol 2024; 81:515-524. [PMID: 38497939 PMCID: PMC10949154 DOI: 10.1001/jamaneurol.2024.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/24/2023] [Indexed: 03/19/2024]
Abstract
Importance It remains unclear why only a small proportion of individuals infected with the Epstein-Barr virus (EBV) develop multiple sclerosis (MS) and what the underlying mechanisms are. Objective To assess the serologic response to all EBV peptides before the first symptoms of MS occur, determine whether the disease is associated with a distinct immune response to EBV, and evaluate whether specific EBV epitopes drive this response. Design, Setting, and Participants In this prospective, nested case-control study, individuals were selected among US military personnel with serum samples stored in the US Department of Defense Serum Repository. Individuals with MS had serum collected at a median 1 year before onset (reported to the military in 2000-2011) and were matched to controls for age, sex, race and ethnicity, blood collection, and military branch. No individuals were excluded. The data were analyzed between September 1, 2022, and August 31, 2023. Exposure Antibodies (enrichment z scores) to the human virome measured using VirScan (phage-displayed immunoprecipitation and sequencing). Main Outcome and Measure Rate ratios (RRs) for MS for antibodies to 2263 EBV peptides (the EBV peptidome) were estimated using conditional logistic regression, adjusting for total anti-EBV nuclear antigen 1 (EBNA-1) antibodies, which have consistently been associated with a higher MS risk. The role of antibodies against other viral peptides was also explored. Results A total of 30 individuals with MS were matched with 30 controls. Mean (SD) age at sample collection was 27.8 (6.5) years; 46 of 60 participants (76.7%) were male. The antibody response to the EBV peptidome was stronger in individuals with MS, but without a discernible pattern. The antibody responses to 66 EBV peptides, the majority mapping to EBNA antigens, were significantly higher in preonset sera from individuals with MS (RR of highest vs lowest tertile of antibody enrichment, 33.4; 95% CI, 2.5-448.4; P for trend = .008). Higher total anti-EBNA-1 antibodies were also associated with an elevated MS risk (top vs bottom tertile: RR, 27.6; 95% CI, 2.3-327.6; P for trend = .008). After adjusting for total anti-EBNA-1 antibodies, risk estimates from most EBV peptides analyses were attenuated, with 4 remaining significantly associated with MS, the strongest within EBNA-6/EBNA-3C, while the association between total anti-EBNA-1 antibodies and MS persisted. Conclusion and Relevance These findings suggest that antibody response to EBNA-1 may be the strongest serologic risk factor for MS. No single EBV peptide stood out as being selectively targeted in individuals with MS but not controls. Larger investigations are needed to explore possible heterogeneity of anti-EBV humoral immunity in MS.
Collapse
Affiliation(s)
- Marianna Cortese
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Yumei Leng
- Division of Genetics, Department of Medicine, Howard Hughes Medical Institute, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Kjetil Bjornevik
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Moriah Mitchell
- Division of Genetics, Department of Medicine, Howard Hughes Medical Institute, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Program in Systems, Synthetic, and Quantitative Biology, Harvard University, Boston, Massachusetts
| | - Brian C. Healy
- Brigham Multiple Sclerosis Center, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts
| | | | - James D. Mancuso
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - David W. Niebuhr
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Kassandra L. Munger
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Epidemiology, Biogen, Cambridge, Massachusetts
| | - Stephen J. Elledge
- Division of Genetics, Department of Medicine, Howard Hughes Medical Institute, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Alberto Ascherio
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Rheum Dis Clin North Am 2024; 50:201-227. [PMID: 38670721 PMCID: PMC11232920 DOI: 10.1016/j.rdc.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
30
|
‘t Hart BA, van Luijn MM. EBV infection drives MS pathology: No. Mult Scler 2024; 30:485-487. [PMID: 38602258 PMCID: PMC11010563 DOI: 10.1177/13524585241235833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Bert A ‘t Hart
- Department of Anatomy and Neurosciences, Amsterdam University Medical Center, VUMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Nataf S, Guillen M, Pays L. The Immunometabolic Gene N-Acetylglucosamine Kinase Is Uniquely Involved in the Heritability of Multiple Sclerosis Severity. Int J Mol Sci 2024; 25:3803. [PMID: 38612613 PMCID: PMC11011344 DOI: 10.3390/ijms25073803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
The clinical severity of multiple sclerosis (MS), an autoimmune disorder of the central nervous system, is thought to be determined by environmental and genetic factors that have not yet been identified. In a recent genome-wide association study (GWAS), a single nucleotide polymorphism (SNP), rs10191329, has been associated with MS severity in two large independent cohorts of patients. Different approaches were followed by the authors to prioritize the genes that are transcriptionally regulated by such an SNP. It was concluded that the identified SNP regulates a group of proximal genes involved in brain resilience and cognitive abilities rather than immunity. Here, by conducting an alternative strategy for gene prioritization, we reached the opposite conclusion. According to our re-analysis, the main target of rs10191329 is N-Acetylglucosamine Kinase (NAGK), a metabolic gene recently shown to exert major immune functions via the regulation of the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) pathway. To gain more insights into the immunometabolic functions of NAGK, we analyzed the currently known list of NAGK protein partners. We observed that NAGK integrates a dense network of human proteins that are involved in glucose metabolism and are highly expressed by classical monocytes. Our findings hold potentially major implications for the understanding of MS pathophysiology.
Collapse
Affiliation(s)
- Serge Nataf
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue du Doyen Lépine, F-69500 Bron, France
- Lyon-Est School of Medicine, University Claude Bernard Lyon 1, 43 Bd du 11 Novembre 1918, F-69100 Villeurbanne, France
| | - Marine Guillen
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue du Doyen Lépine, F-69500 Bron, France
| | - Laurent Pays
- Bank of Tissues and Cells, Hospices Civils de Lyon, Hôpital Edouard Herriot, Place d’Arsonval, F-69003 Lyon, France
- Stem-Cell and Brain Research Institute, 18 Avenue du Doyen Lépine, F-69500 Bron, France
- Lyon-Est School of Medicine, University Claude Bernard Lyon 1, 43 Bd du 11 Novembre 1918, F-69100 Villeurbanne, France
| |
Collapse
|
32
|
Jons D, Grut V, Bergström T, Zetterberg H, Biström M, Gunnarsson M, Vrethem M, Brenner N, Butt J, Blennow K, Nilsson S, Kockum I, Olsson T, Waterboer T, Sundström P, Andersen O. Seroreactivity against lytic, latent and possible cross-reactive EBV antigens appears on average 10 years before MS induced preclinical neuroaxonal damage. J Neurol Neurosurg Psychiatry 2024; 95:325-332. [PMID: 37802637 PMCID: PMC10958269 DOI: 10.1136/jnnp-2023-331868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) and presymptomatic axonal injury appear to develop only after an Epstein-Barr virus (EBV) infection. This association remains to be confirmed across a broad preclinical time range, for lytic and latent EBV seroreactivity, and for potential cross-reacting antigens. METHODS We performed a case-control study with 669 individual serum samples obtained before clinical MS onset, identified through cross-linkage with the Swedish MS register. We assayed antibodies against EBV nuclear antigen 1 (EBNA1), viral capsid antigen p18, glycoprotein 350 (gp350), the potential cross-reacting protein anoctamin 2 (ANO2) and the level of sNfL, a marker of axonal injury. RESULTS EBNA1 (latency) seroreactivity increased in the pre-MS group, at 15-20 years before clinical MS onset, followed by gp350 (lytic) seroreactivity (p=0.001-0.009), ANO2 seropositivity appeared shortly after EBNA1-seropositivity in 16.7% of pre-MS cases and 10.0% of controls (p=0.001).With an average lag of almost a decade after EBV, sNfL gradually increased, mainly in the increasing subgroup of seropositive pre-MS cases (p=8.10-5 compared with non-MS controls). Seropositive pre-MS cases reached higher sNfL levels than seronegative pre-MS (p=0.038). In the EBNA1-seropositive pre-MS group, ANO2 seropositive cases had 26% higher sNfL level (p=0.0026). CONCLUSIONS Seroreactivity against latent and lytic EBV antigens, and in a subset ANO2, was detectable on average a decade before the appearance of a gradually increasing axonal injury occurring in the last decade before the onset of clinical MS. These findings strengthen the hypothesis of latent EBV involvement in the pathogenesis of MS.
Collapse
Affiliation(s)
- Daniel Jons
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Viktor Grut
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Martin Biström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Martin Gunnarsson
- Department of Neurology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Magnus Vrethem
- Department of Neurology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Nicole Brenner
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Julia Butt
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Staffan Nilsson
- Mathematical Sciences, Chalmers University of Technology, Göteborg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Goteborg, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, The Karolinska Neuroimmunology & Multiple Sclerosis Center, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, The Karolinska Neuroimmunology & Multiple Sclerosis Center, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | - Peter Sundström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Oluf Andersen
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
33
|
Holm Hansen R, von Essen MR, Reith Mahler M, Cobanovic S, Sellebjerg F. Sustained effects on immune cell subsets and autoreactivity in multiple sclerosis patients treated with oral cladribine. Front Immunol 2024; 15:1327672. [PMID: 38433828 PMCID: PMC10904620 DOI: 10.3389/fimmu.2024.1327672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Cladribine tablet therapy is an efficacious treatment for multiple sclerosis (MS). Recently, we showed that one year after the initiation of cladribine treatment, T and B cell crosstalk was impaired, reducing potentially pathogenic effector functions along with a specific reduction of autoreactivity to RAS guanyl releasing protein 2 (RASGRP2). In the present study we conducted a longitudinal analysis of the effect of cladribine treatment in patients with RRMS, focusing on the extent to which the effects observed on T and B cell subsets and autoreactivity after one year of treatment are maintained, modulated, or amplified during the second year of treatment. Methods In this case-control exploratory study, frequencies and absolute counts of peripheral T and B cell subsets and B cell cytokine production from untreated patients with relapsing-remitting MS (RRMS) and patients treated with cladribine for 52 (W52), 60 (W60), 72 (W72) and 96 (W96) weeks, were measured using flow cytometry. Autoreactivity was assessed using a FluoroSpot assay. Results We found a substantial reduction in circulating memory B cells and proinflammatory B cell responses. Furthermore, we observed reduced T cell responses to autoantigens possibly presented by B cells (RASGRP2 and a-B crystallin (CRYAB)) at W52 and W96 and a further reduction in responses to the myelin antigens myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) after 96 weeks. Conclusion We conclude that the effects of cladribine observed after year one are maintained and, for some effects, even increased two years after the initiation of a full course of treatment with cladribine tablets.
Collapse
Affiliation(s)
- Rikke Holm Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Marina Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Mie Reith Mahler
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Stefan Cobanovic
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Hauser SL. Silencing Immune Dialogue in Multiple Sclerosis. N Engl J Med 2024; 390:662-663. [PMID: 38354146 PMCID: PMC10959047 DOI: 10.1056/nejme2314434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Affiliation(s)
- Stephen L Hauser
- From the UCSF Weill Institute for Neurosciences and the Department of Neurology, University of California, San Francisco, San Francisco
| |
Collapse
|
35
|
Tieck MP, Vasilenko N, Ruschil C, Kowarik MC. Peripheral memory B cells in multiple sclerosis vs. double negative B cells in neuromyelitis optica spectrum disorder: disease driving B cell subsets during CNS inflammation. Front Cell Neurosci 2024; 18:1337339. [PMID: 38385147 PMCID: PMC10879280 DOI: 10.3389/fncel.2024.1337339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
B cells are fundamental players in the pathophysiology of autoimmune diseases of the central nervous system, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). A deeper understanding of disease-specific B cell functions has led to the differentiation of both diseases and the development of different treatment strategies. While NMOSD is strongly associated with pathogenic anti-AQP4 IgG antibodies and proinflammatory cytokine pathways, no valid autoantibodies have been identified in MS yet, apart from certain antigen targets that require further evaluation. Although both diseases can be effectively treated with B cell depleting therapies, there are distinct differences in the peripheral B cell subsets that influence CNS inflammation. An increased peripheral blood double negative B cells (DN B cells) and plasmablast populations has been demonstrated in NMOSD, but not consistently in MS patients. Furthermore, DN B cells are also elevated in rheumatic diseases and other autoimmune entities such as myasthenia gravis and Guillain-Barré syndrome, providing indirect evidence for a possible involvement of DN B cells in other autoantibody-mediated diseases. In MS, the peripheral memory B cell pool is affected by many treatments, providing indirect evidence for the involvement of memory B cells in MS pathophysiology. Moreover, it must be considered that an important effector function of B cells in MS may be the presentation of antigens to peripheral immune cells, including T cells, since B cells have been shown to be able to recirculate in the periphery after encountering CNS antigens. In conclusion, there are clear differences in the composition of B cell populations in MS and NMOSD and treatment strategies differ, with the exception of broad B cell depletion. This review provides a detailed overview of the role of different B cell subsets in MS and NMOSD and their implications for treatment options. Specifically targeting DN B cells and plasmablasts in NMOSD as opposed to memory B cells in MS may result in more precise B cell therapies for both diseases.
Collapse
Affiliation(s)
| | | | | | - M. C. Kowarik
- Department of Neurology and Stroke, Center for Neurology, and Hertie-Institute for Clinical Brain Research Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
36
|
Gong X, Ma Y, Deng X, Li A, Li X, Kong X, Liu Y, Liu X, Guo K, Yang Y, Li Z, Wei H, Zhou D, Hong Z. Intestinal dysbiosis exacerbates susceptibility to the anti-NMDA receptor encephalitis-like phenotype by changing blood brain barrier permeability and immune homeostasis. Brain Behav Immun 2024; 116:34-51. [PMID: 38030048 DOI: 10.1016/j.bbi.2023.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023] Open
Abstract
Changes in the intestinal microbiota have been observed in patients with anti-N-methyl-D-aspartate receptor encephalitis (NMDARE). However, whether and how the intestinal microbiota is involved in the pathogenesis of NMDARE susceptibility needs to be demonstrated. Here, we first showed that germ-free (GF) mice that underwent fecal microbiota transplantation (FMT) from NMDARE patients, whose fecal microbiota exhibited low short-chain fatty acid content, decreased abundance of Lachnospiraceae, and increased abundance of Verrucomicrobiota, Akkermansia, Parabacteroides, Oscillospirales, showed significant behavioral deficits. Then, these FMT mice were actively immunized with an amino terminal domain peptide from the GluN1 subunit (GluN1356-385) to mimic the pathogenic process of NMDARE. We found that FMT mice showed an increased susceptibility to an encephalitis-like phenotype characterized by more clinical symptoms, greater pentazole (PTZ)-induced susceptibility to seizures, and higher levels of T2 weighted image (T2WI) hyperintensities following immunization. Furthermore, mice with dysbiotic microbiota had impaired blood-brain barrier integrity and a proinflammatory condition. In NMDARE-microbiota recipient mice, the levels of Evan's blue (EB) dye extravasation increased, ZO-1 and claudin-5 expression decreased, and the levels of proinflammatory cytokines (IL-1, IL-6, IL-17, TNF-α and LPS) increased. Finally, significant brain inflammation, mainly in hippocampal and cortical regions, with modest neuroinflammation, immune cell infiltration, and reduced expression of NMDA receptors were observed in NMDARE microbiota recipient mice following immunization. Overall, our findings demonstrated that intestinal dysbiosis increased NMDARE susceptibility, suggesting a new target for limiting the occurrence of the severe phenotype of NMDARE.
Collapse
Affiliation(s)
- Xue Gong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yaru Ma
- Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaolin Deng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Aiqing Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xingjie Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xueying Kong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yue Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xu Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Kundian Guo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuting Yang
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhongxin Li
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
37
|
Abstract
Recent evidence suggests that infection with the Epstein Barr virus (EBV) initiates a prodromal phase of multiple sclerosis (MS) in individuals with genetic and environmental predispositions for this autoimmune disease. In the context of the main genetic risk factor, the major histocompatibility complex (MHC) class II molecule HLA-DRB1*1501, EBV infection is less well controlled in a preclinical mouse model. CD4+ T cells that are primed during EBV infection and recognize EBV transformed B cells in an HLA-DRB1*1501 restricted fashion, cross-react more frequently with myelin autoantigens that are thought to mediate MS. While EBV emerges as an important, possibly essential trigger of MS, more mechanistic insights into this connection are required to understand if targeting of EBV infection itself or of cross-reactive immune responses that recognize both viral and autoantigens might prevent or even allow to treat MS.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland.
| |
Collapse
|
38
|
Lee CY, Chan KH. Personalized Use of Disease-Modifying Therapies in Multiple Sclerosis. Pharmaceutics 2024; 16:120. [PMID: 38258130 PMCID: PMC10820407 DOI: 10.3390/pharmaceutics16010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Multiple sclerosis is an important neurological disease affecting millions of young patients globally. It is encouraging that more than ten disease-modifying drugs became available for use in the past two decades. These disease-modifying therapies (DMTs) have different levels of efficacy, routes of administration, adverse effect profiles and concerns for pregnancy. Much knowledge and caution are needed for their appropriate use in MS patients who are heterogeneous in clinical features and severity, lesion load on magnetic resonance imaging and response to DMT. We aim for an updated review of the concept of personalization in the use of DMT for relapsing MS patients. Shared decision making with consideration for the preference and expectation of patients who understand the potential efficacy/benefits and risks of DMT is advocated.
Collapse
Affiliation(s)
- Chi-Yan Lee
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 405B, 4/F, Professorial Block, 102 Pokfulam Road, Hong Kong
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Koon-Ho Chan
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 405B, 4/F, Professorial Block, 102 Pokfulam Road, Hong Kong
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
- Research Center of Heart, Brain, Hormone and Healthy Aging, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
39
|
Fonseca A, Szysz M, Ly HT, Cordeiro C, Sepúlveda N. IgG Antibody Responses to Epstein-Barr Virus in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Their Effective Potential for Disease Diagnosis and Pathological Antigenic Mimicry. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:161. [PMID: 38256421 PMCID: PMC10820613 DOI: 10.3390/medicina60010161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024]
Abstract
Background and Objectives: The diagnosis and pathology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) remain under debate. However, there is a growing body of evidence for an autoimmune component in ME/CFS caused by the Epstein-Barr virus (EBV) and other viral infections. Materials and Methods: In this work, we analyzed a large public dataset on the IgG antibodies to 3054 EBV peptides to understand whether these immune responses could help diagnose patients and trigger pathological autoimmunity; we used healthy controls (HCs) as a comparator cohort. Subsequently, we aimed at predicting the disease status of the study participants using a super learner algorithm targeting an accuracy of 85% when splitting data into train and test datasets. Results: When we compared the data of all ME/CFS patients or the data of a subgroup of those patients with non-infectious or unknown disease triggers to the data of the HC, we could not find an antibody-based classifier that would meet the desired accuracy in the test dataset. However, we could identify a 26-antibody classifier that could distinguish ME/CFS patients with an infectious disease trigger from the HCs with 100% and 90% accuracies in the train and test sets, respectively. We finally performed a bioinformatic analysis of the EBV peptides associated with these 26 antibodies. We found no correlation between the importance metric of the selected antibodies in the classifier and the maximal sequence homology between human proteins and each EBV peptide recognized by these antibodies. Conclusions: In conclusion, these 26 antibodies against EBV have an effective potential for disease diagnosis in a subset of patients. However, the peptides associated with these antibodies are less likely to induce autoimmune B-cell responses that could explain the pathogenesis of ME/CFS.
Collapse
Affiliation(s)
- André Fonseca
- Faculty of Sciences and Technology, University of Algarve, 8005-139 Faro, Portugal; (A.F.); (C.C.)
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Mateusz Szysz
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| | - Hoang Thien Ly
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| | - Clara Cordeiro
- Faculty of Sciences and Technology, University of Algarve, 8005-139 Faro, Portugal; (A.F.); (C.C.)
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Nuno Sepúlveda
- CEAUL—Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Faculty of Mathematics & Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland; (M.S.); (H.T.L.)
| |
Collapse
|
40
|
Kapoor S, Maréchal L, Sirois I, Caron É. Scaling up robust immunopeptidomics technologies for a global T cell surveillance digital network. J Exp Med 2024; 221:e20231739. [PMID: 38032361 PMCID: PMC10689202 DOI: 10.1084/jem.20231739] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
The human immunopeptidome plays a central role in disease susceptibility and resistance. In our opinion, the development of immunopeptidomics and other peptide sequencing technologies should be prioritized during the next decade, particularly within the framework of the Human Immunopeptidome Project initiative. In this context, we present bold ideas, fresh arguments, and call upon industrial partners and funding organizations to support and champion this important initiative that we believe has the potential to save countless lives in the future.
Collapse
Affiliation(s)
- Saketh Kapoor
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Loïze Maréchal
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Canada
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Canada
| | - Étienne Caron
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Canada
- Yale Center for Immuno-Oncology, Yale Center for Systems and Engineering Immunology, Yale Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
41
|
Stern LJ, Clement C, Galluzzi L, Santambrogio L. Non-mutational neoantigens in disease. Nat Immunol 2024; 25:29-40. [PMID: 38168954 PMCID: PMC11075006 DOI: 10.1038/s41590-023-01664-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/29/2023] [Indexed: 01/05/2024]
Abstract
The ability of mammals to mount adaptive immune responses culminating with the establishment of immunological memory is predicated on the ability of the mature T cell repertoire to recognize antigenic peptides presented by syngeneic MHC class I and II molecules. Although it is widely believed that mature T cells are highly skewed towards the recognition of antigenic peptides originating from genetically diverse (for example, foreign or mutated) protein-coding regions, preclinical and clinical data rather demonstrate that novel antigenic determinants efficiently recognized by mature T cells can emerge from a variety of non-mutational mechanisms. In this Review, we describe various mechanisms that underlie the formation of bona fide non-mutational neoantigens, such as epitope mimicry, upregulation of cryptic epitopes, usage of non-canonical initiation codons, alternative RNA splicing, and defective ribosomal RNA processing, as well as both enzymatic and non-enzymatic post-translational protein modifications. Moreover, we discuss the implications of the immune recognition of non-mutational neoantigens for human disease.
Collapse
Affiliation(s)
- Lawrence J Stern
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbiology Program, UMass Chan Medical School, Worcester, MA, USA
| | - Cristina Clement
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
42
|
Ghosh S, Bhatti GK, Sharma PK, Kandimalla R, Mastana SS, Bhatti JS. Potential of Nano-Engineered Stem Cells in the Treatment of Multiple Sclerosis: A Comprehensive Review. Cell Mol Neurobiol 2023; 44:6. [PMID: 38104307 DOI: 10.1007/s10571-023-01434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023]
Abstract
Multiple sclerosis (MS) is a chronic and degrading autoimmune disorder mainly targeting the central nervous system, leading to progressive neurodegeneration, demyelination, and axonal damage. Current treatment options for MS are limited in efficacy, generally linked to adverse side effects, and do not offer a cure. Stem cell therapies have emerged as a promising therapeutic strategy for MS, potentially promoting remyelination, exerting immunomodulatory effects and protecting against neurodegeneration. Therefore, this review article focussed on the potential of nano-engineering in stem cells as a therapeutic approach for MS, focusing on the synergistic effects of combining stem cell biology with nanotechnology to stimulate the proliferation of oligodendrocytes (OLs) from neural stem cells and OL precursor cells, by manipulating neural signalling pathways-PDGF, BMP, Wnt, Notch and their essential genes such as Sox, bHLH, Nkx. Here we discuss the pathophysiology of MS, the use of various types of stem cells in MS treatment and their mechanisms of action. In the context of nanotechnology, we present an overview of its applications in the medical and research field and discuss different methods and materials used to nano-engineer stem cells, including surface modification, biomaterials and scaffolds, and nanoparticle-based delivery systems. We further elaborate on nano-engineered stem cell techniques, such as nano script, nano-exosome hybrid, nano-topography and their potentials in MS. The article also highlights enhanced homing, engraftment, and survival of nano-engineered stem cells, targeted and controlled release of therapeutic agents, and immunomodulatory and tissue repair effects with their challenges and limitations. This visual illustration depicts the process of utilizing nano-engineering in stem cells and exosomes for the purpose of delivering more accurate and improved treatments for Multiple Sclerosis (MS). This approach targets specifically the creation of oligodendrocytes, the breakdown of which is the primary pathological factor in MS.
Collapse
Affiliation(s)
- Sushruta Ghosh
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University, Rajasthan, India
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University, Rajasthan, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
- Department of Applied Biology, CSIR-Indian Institute of Technology, Hyderabad, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India.
| |
Collapse
|
43
|
Gericke C, Kirabali T, Flury R, Mallone A, Rickenbach C, Kulic L, Tosevski V, Hock C, Nitsch RM, Treyer V, Ferretti MT, Gietl A. Early β-amyloid accumulation in the brain is associated with peripheral T cell alterations. Alzheimers Dement 2023; 19:5642-5662. [PMID: 37314431 DOI: 10.1002/alz.13136] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Fast and minimally invasive approaches for early diagnosis of Alzheimer's disease (AD) are highly anticipated. Evidence of adaptive immune cells responding to cerebral β-amyloidosis has raised the question of whether immune markers could be used as proxies for β-amyloid accumulation in the brain. METHODS Here, we apply multidimensional mass-cytometry combined with unbiased machine-learning techniques to immunophenotype peripheral blood mononuclear cells from a total of 251 participants in cross-sectional and longitudinal studies. RESULTS We show that increases in antigen-experienced adaptive immune cells in the blood, particularly CD45RA-reactivated T effector memory (TEMRA) cells, are associated with early accumulation of brain β-amyloid and with changes in plasma AD biomarkers in still cognitively healthy subjects. DISCUSSION Our results suggest that preclinical AD pathology is linked to systemic alterations of the adaptive immune system. These immunophenotype changes may help identify and develop novel diagnostic tools for early AD assessment and better understand clinical outcomes.
Collapse
Affiliation(s)
- Christoph Gericke
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
| | - Tunahan Kirabali
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
| | - Roman Flury
- Institute of Mathematics, University of Zurich, Zurich, Switzerland
| | - Anna Mallone
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Institute of Microbiology, ETHZ, Zurich, Switzerland
| | - Chiara Rickenbach
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
| | - Luka Kulic
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Roche Pharma Research and Early Development, Roche, Basel, Switzerland
| | - Vinko Tosevski
- Mass Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Center for Prevention and Dementia Therapy, University of Zurich, Schlieren, Switzerland
- Neurimmune AG, Schlieren, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Neurimmune AG, Schlieren, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Center for Prevention and Dementia Therapy, University of Zurich, Schlieren, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Maria Teresa Ferretti
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Women's Brain Project, Guntershausen, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine - IREM, University of Zurich, Schlieren, Switzerland
- Center for Prevention and Dementia Therapy, University of Zurich, Schlieren, Switzerland
- Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland
| |
Collapse
|
44
|
Thomas OG, Olsson T. Mimicking the brain: Epstein-Barr virus and foreign agents as drivers of neuroimmune attack in multiple sclerosis. Front Immunol 2023; 14:1304281. [PMID: 38022632 PMCID: PMC10655090 DOI: 10.3389/fimmu.2023.1304281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
T cells have an essential role in adaptive immunity against pathogens and cancer, but failure of thymic tolerance mechanisms can instead lead to escape of T cells with the ability to attack host tissues. Multiple sclerosis (MS) occurs when structures such as myelin and neurons in the central nervous system (CNS) are the target of autoreactive immune responses, resulting in lesions in the brain and spinal cord which cause varied and episodic neurological deficits. A role for autoreactive T cell and antibody responses in MS is likely, and mounting evidence implicates Epstein-Barr virus (EBV) in disease mechanisms. In this review we discuss antigen specificity of T cells involved in development and progression of MS. We examine the current evidence that these T cells can target multiple antigens such as those from pathogens including EBV and briefly describe other mechanisms through which viruses could affect disease. Unravelling the complexity of the autoantigen T cell repertoire is essential for understanding key events in the development and progression of MS, with wider implications for development of future therapies.
Collapse
Affiliation(s)
- Olivia G. Thomas
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Neuroimmunology Unit, Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
45
|
Lerner A, Benzvi C, Vojdani A. Cross-reactivity and sequence similarity between microbial transglutaminase and human tissue antigens. Sci Rep 2023; 13:17526. [PMID: 37845267 PMCID: PMC10579360 DOI: 10.1038/s41598-023-44452-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023] Open
Abstract
Microbial transglutaminase (mTG) is a bacterial survival factor, frequently used as a food additive to glue processed nutrients. As a result, new immunogenic epitopes are generated that might drive autoimmunity. Presently, its contribution to autoimmunity through epitope similarity and cross-reactivity was investigated. Emboss Matcher was used to perform sequence alignment between mTG and various antigens implicated in many autoimmune diseases. Monoclonal and polyclonal antibodies made specifically against mTG were applied to 77 different human tissue antigens using ELISA. Six antigens were detected to share significant homology with mTG immunogenic sequences, representing major targets of common autoimmune conditions. Polyclonal antibody to mTG reacted significantly with 17 out of 77 tissue antigens. This reaction was most pronounced with mitochondrial M2, ANA, and extractable nuclear antigens. The results indicate that sequence similarity and cross-reactivity between mTG and various tissue antigens are possible, supporting the relationship between mTG and the development of autoimmune disorders 150W.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel.
- Ariel University, Ariel, Israel.
| | - Carina Benzvi
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| | | |
Collapse
|
46
|
Carlberg C, Mycko MP. Linking Mechanisms of Vitamin D Signaling with Multiple Sclerosis. Cells 2023; 12:2391. [PMID: 37830605 PMCID: PMC10571821 DOI: 10.3390/cells12192391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
Environmental triggers often work via signal transduction cascades that modulate the epigenome and transcriptome of cell types involved in the disease process. Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system being characterized by a combination of recurring inflammation, demyelination and progressive loss of axons. The mechanisms of MS onset are not fully understood and genetic variants may explain only some 20% of the disease susceptibility. From the environmental factors being involved in disease development low vitamin D levels have been shown to significantly contribute to MS susceptibility. The pro-hormone vitamin D3 acts via its metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) as a high affinity ligand to the transcription factor VDR (vitamin D receptor) and is a potent modulator of the epigenome at thousands of genomic regions and the transcriptome of hundreds of genes. A major target tissue of the effects of 1,25(OH)2D3 and VDR are cells of innate and adaptive immunity, such as monocytes, dendritic cells as well as B and T cells. Vitamin D induces immunological tolerance in T cells and reduces inflammatory reactions of various types of immune cells, all of which are implicated in MS pathogenesis. The immunomodulatory effects of 1,25(OH)2D3 contribute to the prevention of MS. However, the strength of the responses to vitamin D3 supplementation is highly variegated between individuals. This review will relate mechanisms of individual's vitamin D responsiveness to MS susceptibility and discuss the prospect of vitamin D3 supplementation as a way to extinguish the autoimmunity in MS.
Collapse
Affiliation(s)
- Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Marcin P. Mycko
- Department of Neurology, Laboratory of Neuroimmunology, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
| |
Collapse
|
47
|
Holm Hansen R, von Essen MR, Mahler MR, Cobanovic S, Binko TS, Sellebjerg F. Cladribine Effects on T and B Cells and T Cell Reactivity in Multiple Sclerosis. Ann Neurol 2023; 94:518-530. [PMID: 37191113 DOI: 10.1002/ana.26684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Cladribine tablet therapy is an efficacious treatment for multiple sclerosis (MS), however, its mechanism of action on T and B cell subsets remains unclear. The purpose of this study was to investigate the treatment effects of cladribine on the peripheral pool of T and B cells subsets and reactivity toward central nervous system (CNS) antigens. METHODS In this cross-sectional exploratory study, frequencies and absolute counts of peripheral T and B cell subsets and B cell cytokine production from untreated patients with relapsing-remitting MS (RRMS) and patients treated with cladribine for 1 year were measured using flow cytometry. Autoreactivity was assessed using a FluoroSpot assay. RESULTS We found that 1 year after initiation of cladribine treatment, a lower number of CD4+ T cells was persisting whereas CD19+ B cell counts were normalized compared to untreated patients with RRMS. Follicular helper T cells and their effecter subsets producing cytokines exerting distinct B cell helper activity were lower and, additionally, the peripheral B cell pool was skewed toward a naïve and anti-inflammatory phenotype. Finally, reactivity to the recently identified CNS-enriched autoantigen RAS guanyl-releasing protein 2 (RASGRP2), but not to myelin basic protein and myelin oligodendrocyte glycoprotein, was lower in cladribine-treated patients. INTERPRETATION Together, these investigations on T and B cell subsets suggest that cladribine treatment impairs the B-T cell crosstalk and reduces their ability to mediate pathogenic effector functions. This may result in specific reduction of autoreactivity to RASGRP2 which is expressed in B cells and brain tissue. ANN NEUROL 2023;94:518-530.
Collapse
Affiliation(s)
- Rikke Holm Hansen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Marina Rode von Essen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Mie Reith Mahler
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Stefan Cobanovic
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Tomas Sorm Binko
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Zong B, Yu F, Zhang X, Pang Y, Zhao W, Sun P, Li L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res Rev 2023; 90:102026. [PMID: 37532007 DOI: 10.1016/j.arr.2023.102026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Since the discovery of the mechanosensitive Piezo1 channel in 2010, there has been a significant amount of research conducted to explore its regulatory role in the physiology and pathology of various organ systems. Recently, a growing body of compelling evidence has emerged linking the activity of the mechanosensitive Piezo1 channel to health and disease of the central nervous system. However, the exact mechanisms underlying these associations remain inadequately comprehended. This review systematically summarizes the current research on the mechanosensitive Piezo1 channel and its implications for central nervous system mechanobiology, retrospects the results demonstrating the regulatory role of the mechanosensitive Piezo1 channel on various cell types within the central nervous system, including neural stem cells, neurons, oligodendrocytes, microglia, astrocytes, and brain endothelial cells. Furthermore, the review discusses the current understanding of the involvement of the Piezo1 channel in central nervous system disorders, such as Alzheimer's disease, multiple sclerosis, glaucoma, stroke, and glioma.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Fengzhi Yu
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyou Zhang
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Yige Pang
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, Zhejiang, China
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
49
|
Shah A, Panchal V, Patel K, Alimohamed Z, Kaka N, Sethi Y, Patel N. Pathogenesis and management of multiple sclerosis revisited. Dis Mon 2023; 69:101497. [PMID: 36280474 DOI: 10.1016/j.disamonth.2022.101497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Multiple sclerosis is an autoimmune chronic inflammatory disease characterized by selective destruction of myelin in the CNS neurons (including optic nerve). It was first described in the 19th century and remained elusive owing to the disease's unique relapsing and remitting course. The widespread and debilitating prevalence of multiple sclerosis (MS) has prompted the development of various treatment modalities for its effective management. METHODS AND OBJECTIVES A literature review was conducted using the electronic databases PubMed and Google Scholar. The main objective of the review was to compile the advances in pathogenesis, classifications, and evolving treatment modalities for MS. RESULTS The understanding of the pathogenesis of MS and the potential drug targets for its precise treatment has evolved significantly over the past decade. The experimental developments are also motivating and present a big change coming up in the next 5 years. Numerous disease-modifying therapies (DMTs) have revolutionized the management of MS: interferon (IFN) preparations, monoclonal antibodies-natalizumab and ocrelizumab, immunomodulatory agents-glatiramer acetate, sphingosine 1-phosphate receptor 1 (S1PR1) modulators (Siponimod) and teriflunomide. The traditional parenteral drugs are now available as oral formulations improving patient acceptability. Repurposing various agents used for related diseases may reinforce the drug reserve to manage MS and are under trials. Although at a nascent phase, strategies to enhance re-myelination by stimulating oligodendrocytes are fascinating and hold promise for better outcomes in patients with MS. CONCLUSIONS The recent past has seen staggering inclusions to the management of multiple sclerosis catalyzing a significant turnabout in our approach to diagnosis, treatment, and prognosis. Since the advent of DMTs various other oral and injectable agents have been approved. The advances in MS therapeutics and diagnostics have laid the ground for further research and development to enhance the quality of life of afflicted patients.
Collapse
Affiliation(s)
- Abhi Shah
- Smt NHL MMC, Ahmedabad, Gujarat, 380006, India; PearResearch, India
| | - Viraj Panchal
- Smt NHL MMC, Ahmedabad, Gujarat, 380006, India; PearResearch, India
| | - Kashyap Patel
- Baroda Medical College, Vadodara, India; PearResearch, India
| | - Zainab Alimohamed
- Muhimbili University of Health and Allied Sciences (MUHAS), Tanzania; PearResearch, India
| | - Nirja Kaka
- PearResearch, India; GMERS Medical College, Himmatnagar, India
| | - Yashendra Sethi
- PearResearch, India; Government Doon Medical College, Dehradun, Uttarakhand, India
| | - Neil Patel
- PearResearch, India; GMERS Medical College, Himmatnagar, India.
| |
Collapse
|
50
|
Sahlolbei M, Azangou-Khyavy M, Khanali J, Khorsand B, Shiralipour A, Ahmadbeigi N, Madjd Z, Ghanbarian H, Ardjmand A, Hashemi SM, Kiani J. Engineering chimeric autoantibody receptor T cells for targeted B cell depletion in multiple sclerosis model: An in-vitro study. Heliyon 2023; 9:e19763. [PMID: 37809446 PMCID: PMC10559048 DOI: 10.1016/j.heliyon.2023.e19763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Recent evidence suggests that B cells and autoantibodies have a substantial role in the pathogenesis of Multiple sclerosis. T cells could be engineered to express chimeric autoantibody receptors (CAARs), which have an epitope of autoantigens in their extracellular domain acting as bait for trapping autoreactive B cells. This study aims to assess the function of designed CAAR T cells against B cell clones reactive to the myelin basic protein (MBP) autoantigen. Methods T cells were transduced to express a CAAR consisting of MBP as the extracellular domain. experimental autoimmune encephalomyelitis (EAE) was induced by injecting MBP into mice. The cytotoxicity, proliferation, and cytokine production of the MBP-CAAR T cells were investigated in co-culture with B cells. Results MBP-CAAR T cells showed higher cytotoxic activity against autoreactive B cells in all effector-to-target ratios compared to Mock T cell (empty vector-transduced T cell) and Un-T cells (un-transduced T cell). In co-cultures containing CAAR T cells, there was more proliferation and inflammatory cytokine release as compared to Un-T and Mock T cell groups. Conclusion Based on these findings, CAAR T cells are promising for curing or modulating autoimmunity and can be served as a new approach for clone-specific B cell depletion therapy in multiple sclerosis.
Collapse
Affiliation(s)
- Maryam Sahlolbei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Javad Khanali
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Khorsand
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Computer Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Aref Shiralipour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|