1
|
Wang F, Chen L, Tian Y. Immune traits in combination with inflammatory proteins revealing the pathogenesis of autoimmune liver diseases: A Mendelian randomization study. Cytokine 2025; 185:156815. [PMID: 39579619 DOI: 10.1016/j.cyto.2024.156815] [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: 06/02/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Prior observational research has shown relationships between immune cells, inflammatory proteins, and autoimmune liver diseases (AILD), but their causal associations remain controversial. Therefore, we aimed to clarify the causal association between them. METHODS We carried out a comprehensive Mendelian randomization (MR) analysis to clarify causal associations between 731 immune traits, 91 circulating inflammatory proteins, and AILD, including primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and autoimmune hepatitis (AIH). A two-step MR analysis was used to explore the mediating role of circulating inflammatory proteins. Additionally, we performed sensitivity analyses to evaluate the robustness of the results. RESULTS CD27 on IgD+CD24+B cell, CD27 on IgD-CD38dimB cell, CD27 on unswitched memory B cell, CD27 on switched memory B cell, and CD27 on CD24+CD27+B cell were risk factors for PBC. However, we detected protective effects of CD25 on IgD-CD27-B cell against PBC and CD28 on resting CD4+Treg cell against PSC. Circulating CD40, Interleukin-33, and Delta and Notch-like epidermal growth factor-related receptor were protective factors for PBC. Furthermore, CD40 mediated the association between immune traits and PBC, with the mediated proportions ranging from 18.3 % to 35.4 %. Tumor necrosis factor superfamily member 12 was identified as a risk factor for PSC, and monocyte chemotactic protein 3 was identified as a protective factor for PSC. Additionally, PBC and PSC had effects on eleven immune traits, which are suggested to be the consequences of them. We found no causal association between immune traits, circulating inflammatory proteins, and AIH. Sensitivity analyses demonstrated our results were robust. CONCLUSIONS Our results demonstrate the causal roles of immune traits and inflammatory proteins in PBC and PSC, which reveals their pathogenesis. It is necessary to investigate the specific mechanism by which immune cells and inflammatory proteins affecting the occurrence of AILD.
Collapse
Affiliation(s)
- Feifan Wang
- Department of Surgical, Hebei Medical University, Shijiazhuang 050017, China
| | - Lu Chen
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Yu Tian
- Department of Surgical, Hebei Medical University, Shijiazhuang 050017, China.; Department of Hand & Foot Surgery, First Hospital of Qinhuangdao, Qinhuangdao 066000, China..
| |
Collapse
|
2
|
Su QY, Jiang ZQ, Song XY, Zhang SX. Regulatory B cells in autoimmune diseases: Insights and therapeutic potential. J Autoimmun 2024; 149:103326. [PMID: 39520834 DOI: 10.1016/j.jaut.2024.103326] [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: 07/22/2024] [Revised: 10/06/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Autoimmune diseases are characterized by the body's immune system attacking its own cells, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). In recent studies, regulatory B cells (Bregs), which play a vital role in maintaining peripheral tolerance and controlling persistent autoimmune diseases (ADs), have shown great potential in treating ADs. This review synthesizes the latest advancements in targeted therapies for ADs, with a particular emphasis on the subgroups, phenotypic markers, and signal pathways associated with Bregs. Following an examination of these elements, the discussion pivots to innovative Breg-based therapeutic approaches for the management of ADs.
Collapse
Affiliation(s)
- Qin-Yi Su
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, China
| | - Zhong-Qing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, China
| | - Xuan-Yi Song
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, China
| | - Sheng-Xiao Zhang
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
3
|
von La Roche D, Schumacher M, Kohn M, Trapp J, Schusser B, Rautenschlein S, Härtle S. Characterization of class-switched B cells in chickens. Front Immunol 2024; 15:1484288. [PMID: 39640270 PMCID: PMC11617357 DOI: 10.3389/fimmu.2024.1484288] [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: 08/21/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
While B cell development in the birds' primary B cell organ, the bursa Fabricius, is relatively well understood, very little is known about post bursal B cell differentiation into plasma and memory cells though these cells are essential for a protecting antibody response and so far, no specific markers for these cells were available. Since immunoglobulin class switch is one part of the B cell differentiation process, our objective was to conduct a first detailed investigation of class-switched chicken B cells. As only very few IgY and IgA expressing cells were detected in lymphoid organs of young chickens, we used CD40L and IL-10 to establish a prolonged in vitro culture system, which induces B cell proliferation, class switch to IgY and IgA and enhanced antibody secretion. This enabled a phenotypic analysis of differentiating B cells. Importantly, these cells lost surface expression of the B cell markers chB6 and BAFF-R. B cell receptor surface expression remained unchanged, showing that while differentiating toward plasma cells, B cells can be addressed by L chain staining. Newly generated potential plasma cell markers CD138 and TACI showed only a transient expression on cultured cells and rather act as markers for B cell activation than plasma/memory cells in general. CD57 upregulation was connected to activation and blast formation but not to class switch. We also examined potential changes in class-switched cells in different age groups and post vaccination. Surprisingly, bursa involution, laying and age had no distinct effects on the presence of class-switched cells, but we detected significantly more class-switched B cells post vaccination. Hence, we are now able to generate class-switched plasmablasts in vitro for a more detailed characterization and can address them under different conditions in chickens for further analysis of their B cell response.
Collapse
Affiliation(s)
- Dominik von La Roche
- Department of Veterinary Sciences, AG Immunology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Magdalena Schumacher
- Department of Veterinary Sciences, AG Immunology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Marina Kohn
- Department of Veterinary Sciences, AG Immunology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Johanna Trapp
- Clinic for Poultry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Benjamin Schusser
- Department of Veterinary Sciences, AG Immunology, Ludwig-Maximilians-Universität München, Planegg, Germany
- Reproductive Biotechnology, TUM School of Life Sciences, Technische Universität München, Freising, Germany
- Center of Infection Prevention (ZIP), Technische Universität München, Freising, Germany
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sonja Härtle
- Department of Veterinary Sciences, AG Immunology, Ludwig-Maximilians-Universität München, Planegg, Germany
| |
Collapse
|
4
|
Breitfelder AK, Schrödl W, Baums CG, Alber G, Müller U. The immunoglobulin M-degrading enzyme of Streptococcus suis (Ide Ssuis) leads to long-lasting inhibition of the activation of porcine IgM-secreting B cells. Vet Res 2024; 55:114. [PMID: 39313819 PMCID: PMC11421183 DOI: 10.1186/s13567-024-01363-1] [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: 05/02/2024] [Accepted: 07/12/2024] [Indexed: 09/25/2024] Open
Abstract
Streptococcus suis (S. suis) is one of the most important porcine pathogens, causing severe pathologies such as meningitis or polyarthritis. It is also a very successful colonizer of mucosal surfaces. The IgM-degrading enzyme of S. suis (IdeSsuis) specifically cleaves porcine IgM, which results in complement evasion. On the basis of our previous finding that IdeSsuis also cleaves the IgM B cell receptor in vitro, we verified IgM B cell receptor cleavage ex vivo in whole regional lymph nodes and investigated the working hypothesis that this IgM B cell receptor cleavage results in a long-lasting impaired B cell function. The number of IgM-secreting cells was determined via ELISpot analysis after porcine peripheral blood mononuclear cells had initially been treated with different recombinant S. suis proteins and subsequently stimulated with interleukin-2 and the toll-like receptor 7/8 ligand R848. Compared with treatment with medium or recombinant muramidase-released protein, treatment with rIdeSsuis but also with a cleavage-deficient variant led to a reduction in the number of IgM-secreting cells as well as the level of secreted IgM. Flow cytometry analysis confirmed that the IgM B cell receptor was cleaved only by rIdeSsuis, and the receptor recovered to pretreatment levels on day 2 after treatment. Flow cytometry analysis of B and T cells incubated with fluorescein-labelled recombinant proteins revealed that different rIdeSsuis variants bind specifically to B cells, most prominently the cleavage-deficient variant. Our results indicate that in vitro interference of rIdeSsuis with the IgM B cell receptor results in long-lasting impaired IgM secretion by B cells after toll-like receptor activation. Further studies are warranted to prove that the modulation of B cell function by IdeSsuis could play a role in vivo.
Collapse
Affiliation(s)
- Annika Katharina Breitfelder
- Institute of Bacteriology and Mycology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.
| | - Wieland Schrödl
- Institute of Bacteriology and Mycology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Christoph Georg Baums
- Institute of Bacteriology and Mycology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Gottfried Alber
- Institute of Immunology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, BBZ, University of Leipzig, Leipzig, Germany
| | - Uwe Müller
- Institute of Immunology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, BBZ, University of Leipzig, Leipzig, Germany
| |
Collapse
|
5
|
Balog JÁ, Zvara Á, Bukovinszki V, Puskás LG, Balog A, Szebeni GJ. Comparative single-cell multiplex immunophenotyping of therapy-naive patients with rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus shed light on disease-specific composition of the peripheral immune system. Front Immunol 2024; 15:1376933. [PMID: 38726007 PMCID: PMC11079270 DOI: 10.3389/fimmu.2024.1376933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Systemic autoimmune diseases (SADs) are a significant burden on the healthcare system. Understanding the complexity of the peripheral immunophenotype in SADs may facilitate the differential diagnosis and identification of potential therapeutic targets. Methods Single-cell mass cytometric immunophenotyping was performed on peripheral blood mononuclear cells (PBMCs) from healthy controls (HCs) and therapy-naive patients with rheumatoid arthritis (RA), progressive systemic sclerosis (SSc), and systemic lupus erythematosus (SLE). Immunophenotyping was performed on 15,387,165 CD45+ live single cells from 52 participants (13 cases/group), using an antibody panel to detect 34 markers. Results Using the t-SNE (t-distributed stochastic neighbor embedding) algorithm, the following 17 main immune cell types were determined: CD4+/CD57- T cells, CD4+/CD57+ T cells, CD8+/CD161- T cells, CD8+/CD161+/CD28+ T cells, CD8dim T cells, CD3+/CD4-/CD8- T cells, TCRγ/δ T cells, CD4+ NKT cells, CD8+ NKT cells, classic NK cells, CD56dim/CD98dim cells, B cells, plasmablasts, monocytes, CD11cdim/CD172dim cells, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs). Seven of the 17 main cell types exhibited statistically significant frequencies in the investigated groups. The expression levels of the 34 markers in the main populations were compared between HCs and SADs. In summary, 59 scatter plots showed significant differences in the expression intensities between at least two groups. Next, each immune cell population was divided into subpopulations (metaclusters) using the FlowSOM (self-organizing map) algorithm. Finally, 121 metaclusters (MCs) of the 10 main immune cell populations were found to have significant differences to classify diseases. The single-cell T-cell heterogeneity represented 64MCs based on the expression of 34 markers, and the frequency of 23 MCs differed significantly between at least twoconditions. The CD3- non-T-cell compartment contained 57 MCs with 17 MCs differentiating at least two investigated groups. In summary, we are the first to demonstrate the complexity of the immunophenotype of 34 markers over 15 million single cells in HCs vs. therapy-naive patients with RA, SSc, and SLE. Disease specific population frequencies or expression patterns of peripheral immune cells provide a single-cell data resource to the scientific community.
Collapse
Affiliation(s)
- József Á. Balog
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Vivien Bukovinszki
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine University of Szeged, Szeged, Hungary
- Astridbio Technologies Ltd., Szeged, Hungary
| |
Collapse
|
6
|
Kaiser LM, Freeborn RA, Boss AP, Jin Y, Rockwell CE. Arsenic trioxide inhibits the response of primary human B cells to influenza virus A in vitro. Toxicol In Vitro 2024; 96:105783. [PMID: 38278458 DOI: 10.1016/j.tiv.2024.105783] [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: 07/27/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Arsenic compounds are common environmental toxicants worldwide and particularly enriched in the Northeast and the Southwestern United States, the Alps, and Bangladesh. Exposure to arsenic is linked with various detrimental health outcomes, including cancer, cognitive decline, and kidney damage. Our group has previously shown that arsenic trioxide alters T cell cytokine production. In the current study, we demonstrate that exposure to arsenic compounds alters B cell function in an in vitro influenza model. Human peripheral blood mononuclear cells (PBMCs) were isolated from blood and cultured with arsenic trioxide (As3O2) and subsequently challenged with Influenza A virus. B cells showed decreased expression of CD267, surface IgG and CD80 when treated with As3O2. Taken together, the data suggest that As3O2 affects the activation and surface antibody expression of human peripheral B cells. Overall, this suggests that As3O2 exposure could cause impaired humoral immunity.
Collapse
Affiliation(s)
- Luca M Kaiser
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States of America
| | - Robert A Freeborn
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States of America
| | - Allison P Boss
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States of America; Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan, United States of America
| | - Yining Jin
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States of America
| | - Cheryl E Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States of America; Applied Immunology Center for Research and Education, Michigan State University, East Lansing, United States of America.
| |
Collapse
|
7
|
Tompa A, Faresjö M. Shift in the B cell subsets between children with type 1 diabetes and/or celiac disease. Clin Exp Immunol 2024; 216:36-44. [PMID: 38134245 PMCID: PMC10929695 DOI: 10.1093/cei/uxad136] [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: 11/01/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023] Open
Abstract
Our purpose was to characterize the pattern of B cell subsets in children with a combined diagnosis of type 1 diabetes (T1D) and celiac disease (C) since children with single or double diagnosis of these autoimmune diseases may differ in peripheral B cell subset phenotype patterns. B cells were analyzed with flow cytometry for the expression of differentiation/maturation markers to identify transitional, naive, and memory B cells. Transitional (CD24hiCD38hiCD19+) and memory Bregs (mBregs; CD24hiCD27+CD19+, CD1d+CD27+CD19+, and CD5+CD1d+CD19+) were classified as B cells with regulatory capacity. Children with a combined diagnosis of T1D and C showed a pattern of diminished peripheral B cell subsets. The B cells compartment in children with combined diagnosis had higher percentages of memory B subsets and Bregs, including activated subsets, compared to children with either T1D or C. Children with combined diagnosis had a lower percentage of naive B cells (CD27-CD19+; IgD+CD19+) and an increased percentage of memory B cells (CD27+CD19+; IgD-CD19+). A similar alteration was seen among the CD39+ expressing naive and memory B cells. Memory Bregs (CD1d+CD27+CD19+) were more frequent, contrary to the lower percentage of CD5+ transitional Bregs in children with a combined diagnosis. In children with either T1D or C, the peripheral B cell compartment was dominated by naive cells. Differences in the pattern of heterogeneous peripheral B cell repertoire subsets reflect a shifting in the B cell compartment between children with T1D and/or C. This is an immunological challenge of impact on the pathophysiology of these autoimmune diseases.
Collapse
Affiliation(s)
- Andrea Tompa
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Division of Diagnostics, Region Jönköping County, Jönköping, Sweden
| | - Maria Faresjö
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Department of Life Sciences, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
8
|
Farahnak K, Bai YZ, Yokoyama Y, Morkan DB, Liu Z, Amrute JM, De Filippis Falcon A, Terada Y, Liao F, Li W, Shepherd HM, Hachem RR, Puri V, Lavine KJ, Gelman AE, Bharat A, Kreisel D, Nava RG. B cells mediate lung ischemia/reperfusion injury by recruiting classical monocytes via synergistic B cell receptor/TLR4 signaling. J Clin Invest 2024; 134:e170118. [PMID: 38488011 PMCID: PMC10940088 DOI: 10.1172/jci170118] [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: 03/01/2023] [Accepted: 01/17/2024] [Indexed: 03/18/2024] Open
Abstract
Ischemia/reperfusion injury-mediated (IRI-mediated) primary graft dysfunction (PGD) adversely affects both short- and long-term outcomes after lung transplantation, a procedure that remains the only treatment option for patients suffering from end-stage respiratory failure. While B cells are known to regulate adaptive immune responses, their role in lung IRI is not well understood. Here, we demonstrated by intravital imaging that B cells are rapidly recruited to injured lungs, where they extravasate into the parenchyma. Using hilar clamping and transplant models, we observed that lung-infiltrating B cells produce the monocyte chemokine CCL7 in a TLR4-TRIF-dependent fashion, a critical step contributing to classical monocyte (CM) recruitment and subsequent neutrophil extravasation, resulting in worse lung function. We found that synergistic BCR-TLR4 activation on B cells is required for the recruitment of CMs to the injured lung. Finally, we corroborated our findings in reperfused human lungs, in which we observed a correlation between B cell infiltration and CM recruitment after transplantation. This study describes a role for B cells as critical orchestrators of lung IRI. As B cells can be depleted with currently available agents, our study provides a rationale for clinical trials investigating B cell-targeting therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew E. Gelman
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Department of Surgery
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | |
Collapse
|
9
|
Pattarabanjird T, Nguyen AT, McSkimming C, Dinh HQ, Marshall MA, Ghosheh Y, Gulati R, Durant C, Vallejo J, Saigusa R, Drago F, Guy TV, Premo K, Taylor AM, Paul S, Kundu B, Berr S, Gonen A, Tsimikas S, Miller Y, Pillai S, Ley K, Hedrick CC, McNamara CA. Human circulating CD24 hi marginal zone B cells produce IgM targeting atherogenic antigens and confer protection from vascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1003-1014. [PMID: 39196097 DOI: 10.1038/s44161-023-00356-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 09/26/2023] [Indexed: 08/29/2024]
Abstract
IgMs that inactivate oxidation-specific epitopes (IgMOSE), which are secondary products of lipid peroxidization, protect against inflammatory diseases, including diet-induced atherosclerosis. However, the human B cell subtype that produces IgMOSE remains unknown. In this study, we used single-cell mass cytometry and adoptive transfer of B cell subtypes to NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice to identify B27+IgM+CD24hi cells as the major producers of IgMOSE in humans. Notably, these cells have characteristics of human circulatory marginal zone B (MZB) cells, which are known to be atheoroprotective IgM producers in mice. CD24 antibody treatment to reduce MZB cells and IgM in a hyperlipidemic humanized mouse model provides the evidence that MZB cells protect against vascular inflammation. Consistent with these findings, the frequency of B27+IgM+CD24hi cells (MZB) in patients inversely correlates with coronary artery disease severity.
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Anh Tram Nguyen
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Chantel McSkimming
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | - Huy Q Dinh
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine, Madison, WI, USA
| | - Melissa A Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | | | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Thomas V Guy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Angela M Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Soumen Paul
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Bijoy Kundu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Stuart Berr
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Ayelet Gonen
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yury Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Klaus Ley
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Coleen A McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA.
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
10
|
García-Solís B, Van Den Rym A, Martinez-Martínez L, Franco T, Pérez-Caraballo JJ, Markle J, Cubillos-Zapata C, Marín AV, Recio MJ, Regueiro JR, Navarro-Zapata A, Mestre-Durán C, Ferreras C, Martín Cotázar C, Mena R, de la Calle-Fabregat C, López-Lera A, Fernández Arquero M, Pérez-Martínez A, López-Collazo E, Sánchez-Ramón S, Casanova JL, Martínez-Barricarte R, de la Calle-Martín O, Pérez de Diego R. Inherited human ezrin deficiency impairs adaptive immunity. J Allergy Clin Immunol 2023; 152:997-1009.e11. [PMID: 37301410 PMCID: PMC11009781 DOI: 10.1016/j.jaci.2023.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Inborn errors of immunity (IEI) are a group of monogenic diseases that confer susceptibility to infection, autoimmunity, and cancer. Despite the life-threatening consequences of some IEI, their genetic cause remains unknown in many patients. OBJECTIVE We investigated a patient with an IEI of unknown genetic etiology. METHODS Whole-exome sequencing identified a homozygous missense mutation of the gene encoding ezrin (EZR), substituting a threonine for an alanine at position 129. RESULTS Ezrin is one of the subunits of the ezrin, radixin, and moesin (ERM) complex. The ERM complex links the plasma membrane to the cytoskeleton and is crucial for the assembly of an efficient immune response. The A129T mutation abolishes basal phosphorylation and decreases calcium signaling, leading to complete loss of function. Consistent with the pleiotropic function of ezrin in myriad immune cells, multidimensional immunophenotyping by mass and flow cytometry revealed that in addition to hypogammaglobulinemia, the patient had low frequencies of switched memory B cells, CD4+ and CD8+ T cells, MAIT, γδ T cells, and centralnaive CD4+ cells. CONCLUSIONS Autosomal-recessive human ezrin deficiency is a newly recognized genetic cause of B-cell deficiency affecting cellular and humoral immunity.
Collapse
Affiliation(s)
- Blanca García-Solís
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Ana Van Den Rym
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | | | - Teresa Franco
- Immunology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jareb J Pérez-Caraballo
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tenn; Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tenn
| | - Janet Markle
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tenn; Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tenn
| | - Carolina Cubillos-Zapata
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Centre for Biomedical Research Network, CIBEres, Madrid, Spain
| | - Ana V Marín
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - María J Recio
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - José R Regueiro
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Alfonso Navarro-Zapata
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Carmen Mestre-Durán
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Cristina Ferreras
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Carla Martín Cotázar
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Roció Mena
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | | | - Alberto López-Lera
- IdiPAZ Institute for Health Research, La Paz University Hospital, CIBERER U-754, Madrid, Spain
| | - Miguel Fernández Arquero
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain; Clinical Immunology Department, San Carlos Clinical Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Eduardo López-Collazo
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Silvia Sánchez-Ramón
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain; Clinical Immunology Department, San Carlos Clinical Hospital, Madrid, Spain
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Imagine Institute, University Paris Descartes, Paris, France; Howard Hughes Medical Institute, New York, NY
| | - Rubén Martínez-Barricarte
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tenn; Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tenn
| | | | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid, Spain; Interdepartmental Group of Immunodeficiencies, Madrid, Spain.
| |
Collapse
|
11
|
Vitallé J, Zenarruzabeitia O, Merino-Pérez A, Terrén I, Orrantia A, Pacho de Lucas A, Iribarren JA, García-Fraile LJ, Balsalobre L, Amo L, de Andrés B, Borrego F. Human IgM hiCD300a + B Cells Are Circulating Marginal Zone Memory B Cells That Respond to Pneumococcal Polysaccharides and Their Frequency Is Decreased in People Living with HIV. Int J Mol Sci 2023; 24:13754. [PMID: 37762055 PMCID: PMC10530418 DOI: 10.3390/ijms241813754] [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: 07/27/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
CD300a is differentially expressed among B cell subsets, although its expression in immunoglobulin (Ig)M+ B cells is not well known. We identified a B cell subset expressing CD300a and high levels of IgM (IgMhiCD300a+). The results showed that IgMhiCD300a+ B cells were CD10-CD27+CD25+IgDloCD21hiCD23-CD38loCD1chi, suggesting that they are circulating marginal zone (MZ) IgM memory B cells. Regarding the immunoglobulin repertoire, IgMhiCD300a+ B cells exhibited a higher mutation rate and usage of the IgH-VDJ genes than the IgM+CD300a- counterpart. Moreover, the shorter complementarity-determining region 3 (CDR3) amino acid (AA) length from IgMhiCD300a+ B cells together with the predicted antigen experience repertoire indicates that this B cell subset has a memory phenotype. IgM memory B cells are important in T cell-independent responses. Accordingly, we demonstrate that this particular subset secretes higher amounts of IgM after stimulation with pneumococcal polysaccharides or a toll-like receptor 9 (TLR9) agonist than IgM+CD300a- cells. Finally, the frequency of IgMhiCD300a+ B cells was lower in people living with HIV-1 (PLWH) and it was inversely correlated with the years with HIV infection. Altogether, these data help to identify a memory B cell subset that contributes to T cell-independent responses to pneumococcal infections and may explain the increase in severe pneumococcal infections and the impaired responses to pneumococcal vaccination in PLWH.
Collapse
Affiliation(s)
- Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
- Instituto de Biomedicina de Sevilla (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, 41013 Seville, Spain
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
| | - Aitana Merino-Pérez
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
| | - Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
| | - Arantza Pacho de Lucas
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Immunology Service, Cruces University Hospital, 48903 Barakaldo, Spain
| | - José A. Iribarren
- Department of Infectious Diseases, Donostia University Hospital, Biodonostia Health Research Institute, 20014 Donostia-San Sebastián, Spain;
| | - Lucio J. García-Fraile
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Internal Medicine, La Princesa University Hospital, 28006 Madrid, Spain
| | - Luz Balsalobre
- Laboratory of Microbiology, UR Salud, Infanta Sofía University Hospital, 28702 Madrid, Spain;
| | - Laura Amo
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Belén de Andrés
- Immunobiology Department, Carlos III Health Institute, 28220 Madrid, Spain;
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (O.Z.); (A.M.-P.); (I.T.); (A.O.); (L.A.)
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
12
|
Garcia-Solis B, Van Den Rym A, Pérez-Caraballo JJ, Al-Ayoubi A, Alazami AM, Lorenzo L, Cubillos-Zapata C, López-Collazo E, Pérez-Martínez A, Allende LM, Markle J, Fernández-Arquero M, Sánchez-Ramón S, Recio MJ, Casanova JL, Mohammed R, Martinez-Barricarte R, Pérez de Diego R. Clinical and Immunological Features of Human BCL10 Deficiency. Front Immunol 2021; 12:786572. [PMID: 34868072 PMCID: PMC8633570 DOI: 10.3389/fimmu.2021.786572] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022] Open
Abstract
The CARD-BCL10-MALT1 (CBM) complex is critical for the proper assembly of human immune responses. The clinical and immunological consequences of deficiencies in some of its components such as CARD9, CARD11, and MALT1 have been elucidated in detail. However, the scarcity of BCL10 deficient patients has prevented gaining detailed knowledge about this genetic disease. Only two patients with BCL10 deficiency have been reported to date. Here we provide an in-depth description of an additional patient with autosomal recessive complete BCL10 deficiency caused by a nonsense mutation that leads to a loss of expression (K63X). Using mass cytometry coupled with unsupervised clustering and machine learning computational methods, we obtained a thorough characterization of the consequences of BCL10 deficiency in different populations of leukocytes. We showed that in addition to the near absence of memory B and T cells previously reported, this patient displays a reduction in NK, γδT, Tregs, and TFH cells. The patient had recurrent respiratory infections since early childhood, and showed a family history of lethal severe infectious diseases. Fortunately, hematopoietic stem-cell transplantation (HSCT) cured her. Overall, this report highlights the importance of early genetic diagnosis for the management of BCL10 deficient patients and HSCT as the recommended treatment to cure this disease.
Collapse
Affiliation(s)
- Blanca Garcia-Solis
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Ana Van Den Rym
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Jareb J Pérez-Caraballo
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States.,Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Abdulwahab Al-Ayoubi
- Department of Pediatrics, King Saud Medical City Children's Hospital, Riyadh, Saudi Arabia
| | - Anas M Alazami
- Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Carolina Cubillos-Zapata
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Center for Biomedical Research Network, CIBEres, Madrid, Spain
| | - Eduardo López-Collazo
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Paediatric Oncology, Haematopoietic Stem Cell Transplantation, Cell Therapy, Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, La Paz University Hospital, Madrid, Spain.,Department of Paediatric Haemato-Oncology and Stem Cell Transplantation, La Paz University Hospital, Madrid, Spain
| | - Luis M Allende
- Department of Immunology, 12 de Octubre Hospital, Research Insitute imas12, Complutense University, Madrid, Spain
| | - Janet Markle
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States.,Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Miguel Fernández-Arquero
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain.,Clinical Immunology Department, San Carlos Clinical Hospital, Madrid, Spain
| | - Silvia Sánchez-Ramón
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain.,Clinical Immunology Department, San Carlos Clinical Hospital, Madrid, Spain
| | - Maria J Recio
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States.,Imagine Institute, University Paris Descartes, Paris, France.,Howard Hughes Medical Institute, New York, NY, United States
| | - Reem Mohammed
- Department of Pediatrics, Division of Allergy & Immunology King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Rubén Martinez-Barricarte
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States.,Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, Madrid, Spain.,Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| |
Collapse
|
13
|
DiSano KD, Gilli F, Pachner AR. Memory B Cells in Multiple Sclerosis: Emerging Players in Disease Pathogenesis. Front Immunol 2021; 12:676686. [PMID: 34168647 PMCID: PMC8217754 DOI: 10.3389/fimmu.2021.676686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.
Collapse
Affiliation(s)
- Krista D. DiSano
- Department of Neurology, Geisel School of Medicine & Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | | |
Collapse
|
14
|
B cells reappear less mature and more activated after their anti-CD20-mediated depletion in multiple sclerosis. Proc Natl Acad Sci U S A 2020; 117:25690-25699. [PMID: 32999069 PMCID: PMC7568262 DOI: 10.1073/pnas.2012249117] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Anti-CD20–mediated B cell depletion is a highly effective therapy in MS. However, long-term effects of this approach on the immune system are not yet characterized in detail. After cessation of anti-CD20 treatment, B cells reappear immature yet highly activated. In addition, anti-CD20 treatment exerts long-lasting effects on T cells, which may be important for its clinical effect. These findings may help in guiding therapeutic decisions with regard to treatment intervals and follow-up therapies. B cell depletion via anti-CD20 antibodies is a highly effective treatment for multiple sclerosis (MS). However, little is known about the maturation/activation stage of the returning B cell population after treatment cessation and the wider effects on other immune cells. In the present study, 15 relapsing-remitting MS patients receiving 1,000 mg of rituximab were included. B, T, and myeloid cells were analyzed before anti-CD20 administration and in different time intervals thereafter over a period of 24 mo. In comparison to the phenotype before anti-CD20 treatment, the reappearing B cell pool revealed a less mature and more activated phenotype: 1) reappearing B cells were significantly enriched in transitional (before: 10.1 ± 1.9%, after: 58.8 ± 5.2%) and mature naive phenotypes (before: 45.5 ± 3.1%, after: 25.1 ± 3.5%); 2) the frequency of memory B cells was reduced (before: 36.7 ± 3.1%, after: 8.9 ± 1.7%); and 3) reappearing B cells showed an enhanced expression of activation markers CD25 (before: 2.1 ± 0.4%, after: 9.3 ± 2.1%) and CD69 (before: 5.9 ± 1.0%, after: 21.4 ± 3.0%), and expressed significantly higher levels of costimulatory CD40 and CD86. T cells showed 1) a persistent increase in naive (CD4+: before: 11.8 ± 1.3%, after: 18.4 ± 3.4%; CD8+: before: 12.5 ± 1.4%, after: 16.5 ± 2.3%) and 2) a decrease in terminally differentiated subsets (CD4+: before: 47.3 ± 3.2%, after: 34.4 ± 3.7%; CD8+: before: 53.7 ± 2.1%, after: 49.1 ± 2.7%).
Collapse
|
15
|
Miyazaki Y, Niino M. Regulatory B cells in neuroimmunological diseases. CLINICAL AND EXPERIMENTAL NEUROIMMUNOLOGY 2020; 11:156-162. [DOI: 10.1111/cen3.12588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 01/03/2025]
Abstract
AbstractThe idea that B cells participate in immune regulation was initially postulated from observations in animals in the 1970s. It is now established that certain B‐cell populations, known as regulatory B cells, regulate immune reactions in various animal models of autoimmunity, chiefly through the production of interleukin‐10. Subsequent to these findings in animals, several B‐cell subsets have been identified in human blood that are capable of producing interleukin‐10 when stimulated ex vivo. Although we still do not have direct evidence showing that these interleukin‐10‐producing B cells regulate autoimmunity in humans, their functional and phenotypic homology to regulatory B cells in animals, their abnormalities reported in various autoimmune diseases and their alterations in response to treatments all suggest their regulatory role in humans. In this review, the role of regulatory B cells in three neuroimmunological diseases – multiple sclerosis, neuromyelitis optica spectrum disorder and myasthenia gravis – are discussed.
Collapse
Affiliation(s)
- Yusei Miyazaki
- Department of Clinical Research National Hospital Organization Hokkaido Medical Center Sapporo Japan
- Department of Neurology National Hospital Organization Hokkaido Medical Center Sapporo Japan
| | - Masaaki Niino
- Department of Clinical Research National Hospital Organization Hokkaido Medical Center Sapporo Japan
| |
Collapse
|
16
|
Figueiredo CR, Kalirai H, Sacco JJ, Azevedo RA, Duckworth A, Slupsky JR, Coulson JM, Coupland SE. Loss of BAP1 expression is associated with an immunosuppressive microenvironment in uveal melanoma, with implications for immunotherapy development. J Pathol 2020; 250:420-439. [PMID: 31960425 PMCID: PMC7216965 DOI: 10.1002/path.5384] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) induces durable responses in many metastatic cancers. Metastatic uveal melanoma (mUM), typically occurring in the liver, is one of the most refractory tumours to ICIs and has dismal outcomes. Monosomy 3 (M3), polysomy 8q, and BAP1 loss in primary uveal melanoma (pUM) are associated with poor prognoses. The presence of tumour-infiltrating lymphocytes (TILs) within pUM and surrounding mUM - and some evidence of clinical responses to adoptive TIL transfer - strongly suggests that UMs are indeed immunogenic despite their low mutational burden. The mechanisms that suppress TILs in pUM and mUM are unknown. We show that BAP1 loss is correlated with upregulation of several genes associated with suppressive immune responses, some of which build an immune suppressive axis, including HLA-DR, CD38, and CD74. Further, single-cell analysis of pUM by mass cytometry confirmed the expression of these and other markers revealing important functions of infiltrating immune cells in UM, most being regulatory CD8+ T lymphocytes and tumour-associated macrophages (TAMs). Transcriptomic analysis of hepatic mUM revealed similar immune profiles to pUM with BAP1 loss, including the expression of IDO1. At the protein level, we observed TAMs and TILs entrapped within peritumoural fibrotic areas surrounding mUM, with increased expression of IDO1, PD-L1, and β-catenin (CTNNB1), suggesting tumour-driven immune exclusion and hence the immunotherapy resistance. These findings aid the understanding of how the immune response is organised in BAP1 - mUM, which will further enable functional validation of detected biomarkers and the development of focused immunotherapeutic approaches. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carlos R Figueiredo
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of the Faculty of Medicine, MediCity Research Laboratory and Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Helen Kalirai
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph J Sacco
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of Medical OncologyThe Clatterbridge Cancer CentreWirralUK
| | - Ricardo A Azevedo
- Department of Cancer BiologyThe University of Texas–MD Anderson Cancer CenterHoustonTXUSA
| | - Andrew Duckworth
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Judy M Coulson
- Department of Cellular and Molecular PhysiologyUniversity of LiverpoolLiverpoolUK
| | - Sarah E Coupland
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| |
Collapse
|
17
|
Hamilton JA, Hsu HC, Mountz JD. Autoreactive B cells in SLE, villains or innocent bystanders? Immunol Rev 2019; 292:120-138. [PMID: 31631359 PMCID: PMC6935412 DOI: 10.1111/imr.12815] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022]
Abstract
The current concepts for development of autoreactive B cells in SLE (systemic lupus erythematosus) focus on extrinsic stimuli and factors that provoke B cells into tolerance loss. Traditionally, major tolerance loss pathways are thought to be regulated by factors outside the B cell including autoantigen engagement of the B-cell receptor (BCR) with simultaneous type I interferon (IFN) produced by dendritic cells, especially plasmacytoid dendritic cells (pDCs). Later, in autoreactive follicles, B-cells encounter T-follicular helper cells (Tfh) that produce interleukin (IL)-21, IL-4 and pathogenic cytokines, IL-17 and IFN gamma (IFNɣ). This review discusses these mechanisms and also highlights recent advances pointing to the peripheral transitional B-cell stage as a major juncture where transient autocrine IFNβ expression by developing B-cells imprints a heightened susceptibility to external factors favoring differentiation into autoantibody-producing plasmablasts. Recent studies highlight transitional B-cell heterogeneity as a determinant of intrinsic resistance or susceptibility to tolerance loss through the shaping of B-cell responsiveness to cytokines and other environment factors.
Collapse
Affiliation(s)
| | - Hui-Chen Hsu
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - John D Mountz
- University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
18
|
Mass cytometry dissects T cell heterogeneity in the immune tumor microenvironment of common dysproteinemias at diagnosis and after first line therapies. Blood Cancer J 2019; 9:72. [PMID: 31462637 PMCID: PMC6713712 DOI: 10.1038/s41408-019-0234-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022] Open
Abstract
Dysproteinemias progress through a series of clonal evolution events in the tumor cell along with the development of a progressively more “permissive” immune tumor microenvironment (iTME). Novel multiparametric cytometry approaches, such as cytometry by time-of-flight (CyTOF) combined with novel gating algorithms can rapidly characterize previously unknown phenotypes in the iTME of tumors and better capture its heterogeneity. Here, we used a 33-marker CyTOF panel to characterize the iTME of dysproteinemia patients (MGUS, multiple myeloma—MM, smoldering MM, and AL amyloidosis) at diagnosis and after standard of care first line therapies (triplet induction chemotherapy and autologous stem cell transplant—ASCT). We identify novel subsets, some of which are unique to the iTME and absent from matched peripheral blood samples, with potential roles in tumor immunosurveillance as well as tumor immune escape. We find that AL amyloidosis has a distinct iTME compared to other dysproteinemias with higher myeloid and “innate-like” T cell subset infiltration. We show that T cell immune senescence might be implicated in disease pathogenesis in patients with trisomies. Finally, we demonstrate that the early post-ASCT period is associated with an increase of senescent and exhausted subsets, which might have implications for the rational selection of post-ASCT therapies.
Collapse
|
19
|
Levels MJ, Fehres CM, van Baarsen LG, van Uden NO, Germar K, O'Toole TG, Blijdorp IC, Semmelink JF, Doorenspleet ME, Bakker AQ, Krasavin M, Tomilin A, Brouard S, Spits H, Baeten DL, Yeremenko NG. BOB.1 controls memory B-cell fate in the germinal center reaction. J Autoimmun 2019; 101:131-144. [DOI: 10.1016/j.jaut.2019.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 11/30/2022]
|
20
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
21
|
Staun-Ram E, Najjar E, Volkowich A, Miller A. Dimethyl fumarate as a first- vs second-line therapy in MS: Focus on B cells. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2018; 5:e508. [PMID: 30345334 PMCID: PMC6192691 DOI: 10.1212/nxi.0000000000000508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
Objective To elucidate the immunomodulatory effects of dimethyl fumarate (DMF) on B cells in patients with relapsing MS receiving DMF as a "1st-line" vs "2nd-line" therapy. Methods B cells were isolated from 43 patients with MS at baseline and after 15-week DMF therapy. Phenotype and functional markers and cytokine profile were assessed by flow cytometry. Analysis included clinical and MRI parameters recorded during a 1-year follow-up. Results 1st-line and 2nd-line patients presented several differences in their baseline immune profile, which corresponded with differences in their immunologic response to DMF treatment. DMF reduced the proportions of B cells and CD8 T cells whereas increased monocytes. DMF reduced memory B cells, including plasma cells in 2nd-line patients only, whereas strongly increased transitional B cells. Several IL10+ B-cell subsets and TGFβ+ B cells were increased. Proinflammatory LTα+ and TNFα+ B cells were reduced, while IL4+ B cells elevated, whereas IFNγ+ B cells showed opposite effects in 1st-line and 2nd-line patients. HLA and ICAM-1 expression was increased, but % CD86+ B cells reduced. The expression of B-cell activating factor receptor and the proportion of activated CD69 B cells were increased. Conclusions DMF is associated with increased transitional and IL10+ and TGFβ+ regulatory B cells and a shift toward a more anti-inflammatory immune profile. Cell activation with reduced costimulatory capacity may induce immune hyporesponsiveness. Carryover effects of preceding therapies in 2nd-line patients and the stage of disease influence the immune profile of the patients and the immunomodulatory effects of DMF.
Collapse
Affiliation(s)
- Elsebeth Staun-Ram
- Rappaport Faculty of Medicine (E.S.-R., E.N., A.M.), Technion-Israel Institute of Technology; and the Department of Neurology (A.V., A.M.), Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
| | - Eiman Najjar
- Rappaport Faculty of Medicine (E.S.-R., E.N., A.M.), Technion-Israel Institute of Technology; and the Department of Neurology (A.V., A.M.), Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
| | - Anat Volkowich
- Rappaport Faculty of Medicine (E.S.-R., E.N., A.M.), Technion-Israel Institute of Technology; and the Department of Neurology (A.V., A.M.), Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
| | - Ariel Miller
- Rappaport Faculty of Medicine (E.S.-R., E.N., A.M.), Technion-Israel Institute of Technology; and the Department of Neurology (A.V., A.M.), Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel
| |
Collapse
|
22
|
Negera E, Walker SL, Bekele Y, Dockrell HM, Lockwood DN. Increased activated memory B-cells in the peripheral blood of patients with erythema nodosum leprosum reactions. PLoS Negl Trop Dis 2017; 11:e0006121. [PMID: 29253897 PMCID: PMC5749895 DOI: 10.1371/journal.pntd.0006121] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/02/2018] [Accepted: 11/20/2017] [Indexed: 12/14/2022] Open
Abstract
B-cells, in addition to antibody secretion, have emerged increasingly as effector and immunoregulatory cells in several chronic inflammatory diseases. Although Erythema Nodosum Leprosum (ENL) is an inflammatory complication of leprosy, the role of B- cell subsets has never been studied in this patient group. Therefore, it would be interesting to examine the contribution of B-cells in the pathogenesis of ENL. A case-control study design was used to recruit 30 untreated patients with ENL and 30 non-reactional lepromatous leprosy (LL) patient controls at ALERT Hospital, Ethiopia. Peripheral blood samples were obtained before, during and after treatment from each patient. Peripheral blood mononuclear cells (PBMCs) were isolated and used for immunophenotyping of B- cell subsets by flow cytometry. The kinetics of B-cells in patients with ENL before, during and after Prednisolone treatment of ENL was compared with LL patient controls as well as within ENL group. Total B-cells, mature B-cells and resting memory B-cells were not significantly different between patients with ENL reactions and LL controls before treatment. Interestingly, while the percentage of naive B-cells was significantly lower in untreated ENL patients than in LL patient controls, the percentage of activated memory B-cells was significantly higher in these untreated ENL patients than in LL controls. On the other hand, the percentage of tissue-like memory B-cells was considerably low in untreated ENL patients compared to LL controls. It appears that the lower frequency of tissue-like memory B-cells in untreated ENL could promote the B-cell/T-cell interaction in these patients through downregulation of inhibitory molecules unlike in LL patients. Conversely, the increased production of activated memory B-cells in ENL patients could imply the scale up of immune activation through antigen presentation to T-cells. However, the generation and differential function of these memory B-cells need further investigation. The finding of increased percentage of activated memory B-cells in untreated patients with ENL reactions suggests the association of these cells with the ENL pathology. The mechanism by which inflammatory reactions like ENL affecting these memory cells and contributing to the disease pathology is an interesting area to be explored for and could lead to the development of novel and highly efficacious drug for ENL treatment.
Collapse
Affiliation(s)
- Edessa Negera
- London School of Hygiene and Tropical Medicine (LSHTM), Faculty of Infectious Tropical Diseases, London, United Kingdom
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Stephen L. Walker
- London School of Hygiene and Tropical Medicine (LSHTM), Faculty of Infectious Tropical Diseases, London, United Kingdom
| | - Yonas Bekele
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Hazel M. Dockrell
- London School of Hygiene and Tropical Medicine (LSHTM), Faculty of Infectious Tropical Diseases, London, United Kingdom
| | - Diana N. Lockwood
- London School of Hygiene and Tropical Medicine (LSHTM), Faculty of Infectious Tropical Diseases, London, United Kingdom
| |
Collapse
|
23
|
Gudbrandsdottir S, Brimnes M, Køllgaard T, Hasselbalch HC, Nielsen CH. Effects of rituximab and dexamethasone on regulatory and proinflammatory B-cell subsets in patients with primary immune thrombocytopenia. Eur J Haematol 2017; 100:45-52. [PMID: 28960473 DOI: 10.1111/ejh.12978] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2017] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the cytokine production and surface marker composition of B cells in adult patients with newly diagnosed primary immune thrombocytopenia (ITP) before and 12 months after treatment with rituximab + dexamethasone (RTX+DXM) or dexamethasone (DXM). METHODS Peripheral blood mononuclear cells were isolated from nine patients treated with RTX+DXM, seven patients treated with DXM, and seven healthy donors. Expression of the cell-surface markers CD5, CD27, CD25, and CD19, and intracellular content of IL-6 and IL-10 were measured by flow cytometry. RESULTS PBMCs from ITP patients at baseline contained a lower proportion of IL-10+ B cells (P < .01) and IL-6+ B cells (P < .01) than healthy controls. All patients responded to therapy and levels were normalized at 12 months. The proportion of CD5+ B cells increased (P < .01) and CD27+ memory B cells decreased (P < .05) 12 months after treatment with RTX+DXM compared to baseline, with an inverse correlation between platelet numbers and the proportion of CD27+ B cells (R = -0.71; P < .05). CONCLUSION Both treatment regimens normalized the frequencies of cytokine-producing B cells. The additional increase in CD5+ B cells after RTX+DXM is compatible with induction of Bregs.
Collapse
Affiliation(s)
- Sif Gudbrandsdottir
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Hematology, Roskilde Hospital, Copenhagen, Denmark
| | - Marie Brimnes
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tania Køllgaard
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Claus H Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
24
|
Mei HE, Hahne S, Redlin A, Hoyer BF, Wu K, Baganz L, Lisney AR, Alexander T, Rudolph B, Dörner T. Plasmablasts With a Mucosal Phenotype Contribute to Plasmacytosis in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017. [DOI: 10.1002/art.40181] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Henrik E. Mei
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Stefanie Hahne
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Andreas Redlin
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Bimba F. Hoyer
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Kaiyin Wu
- Charité University Medicine Berlin; Berlin Germany
| | - Lisa Baganz
- German Rheumatism Research Center Berlin; Berlin Germany
| | - Anna R. Lisney
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Tobias Alexander
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | | | - Thomas Dörner
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| |
Collapse
|
25
|
DiSano KD, Stohlman SA, Bergmann CC. An optimized method for enumerating CNS derived memory B cells during viral-induced inflammation. J Neurosci Methods 2017; 285:58-68. [PMID: 28495370 PMCID: PMC5545894 DOI: 10.1016/j.jneumeth.2017.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/26/2017] [Accepted: 05/07/2017] [Indexed: 11/29/2022]
Abstract
Memory B cell markers characterizing peripheral B cell phenotypes show more diverse expression patterns in the infected central nervous system (CNS). TLR7/8 stimulation for 2 days prior to ELISPOT analysis achieves optimal conversion of CNS-derived memory B cells to ASC while minimizing cell loss. In vitro stimulation allows simultaneous assessment of antibody secreting cell and memory B cell isotype, antigen specificity, and temporal alterations during CNS inflammation.
Background CNS inflammation resulting from infection, injury, or neurodegeneration leads to accumulation of diverse B cell subsets. Although antibody secreting cells (ASC) within the inflamed CNS have been extensively examined, memory B cell (Bmem) characterization has been limited as they do not secrete antibody without stimulation. Moreover, unlike human Bmem, reliable surface markers for murine Bmem remain elusive. New method Using a viral encephalomyelitis model we developed a modified limiting dilution in vitro stimulation assay to convert CNS-derived virus specific Bmem into ASC. Comparison with existing methods Stimulation methods established for lymphoid tissue cells using prolonged stimulation with viral lysate resulted in substantial ASC loss and minimal Bmem to ASC conversion of CNS-derived cells. By varying stimulation duration, TLR activators, and culture supplements, we achieved optimal conversion by culturing cells with TLR7/8 agonist R848 in the presence of feeder cells for 2 days. Results Flow cytometry markers CD38 and CD73 characterizing murine Bmem from lymphoid tissue showed more diverse expression patterns on corresponding CNS-derived B cell subsets. Using the optimized TLR7/8 stimulation protocol, we compared virus-specific IgG Bmem versus pre-existing ASC within the brain and spinal cord. Increasing Bmem frequencies during chronic infection mirrored kinetics of ASC. However, despite initially similar Bmem and ASC accumulation, Bmem prevailed in the brain, but were lower than ASC in the spinal cord during persistence. Conclusion Simultaneous enumeration of antigen-specific Bmem and ASC using the Bmem assay optimized for CNS-derived cells enables characterization of temporal changes during microbial or auto-antigen induced neuroinflammation.
Collapse
Affiliation(s)
- Krista D DiSano
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, United States; School of Biomedical Sciences, Kent State University, Kent, OH 44242, United States
| | - Stephen A Stohlman
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Cornelia C Bergmann
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, United States.
| |
Collapse
|
26
|
Staun-Ram E, Miller A. Effector and regulatory B cells in Multiple Sclerosis. Clin Immunol 2017; 184:11-25. [PMID: 28461106 DOI: 10.1016/j.clim.2017.04.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
The role of B cells in the pathogenesis of Multiple Sclerosis (MS), an autoimmune neurodegenerative disease, is becoming eminent in recent years, but the specific contribution of the distinct B cell subsets remains to be elucidated. Several B cell subsets have shown regulatory, anti-inflammatory capacities in response to stimuli in vitro, as well as in the animal model of MS: Experimental Autoimmune Encephalomyelitis (EAE). However, the functional role of the B regulatory cells (Bregs) in vivo and specifically in the human disease is yet to be clarified. In the present review, we have summarized the updated information on the roles of effector and regulatory B cells in MS and the immune-modulatory effects of MS therapeutic agents on their phenotype and function.
Collapse
Affiliation(s)
- Elsebeth Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ariel Miller
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel.
| |
Collapse
|
27
|
Bankó Z, Pozsgay J, Szili D, Tóth M, Gáti T, Nagy G, Rojkovich B, Sármay G. Induction and Differentiation of IL-10-Producing Regulatory B Cells from Healthy Blood Donors and Rheumatoid Arthritis Patients. THE JOURNAL OF IMMUNOLOGY 2017; 198:1512-1520. [PMID: 28087671 DOI: 10.4049/jimmunol.1600218] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022]
Abstract
The most important feature of B cells is the production of Abs upon activation; additionally, B cells produce pro- and anti-inflammatory cytokines in response to certain stimuli. IL-10-producing B cells represent a major subset of regulatory B cells (Bregs) that suppress autoimmune and inflammatory responses. B cells play a crucial role in the development and maintenance of the chronic inflammatory autoimmune disease rheumatoid arthritis (RA); however, controversial data are available on IL-10- producing Bregs in RA. Our aim was to identify the optimal conditions that induce IL-10+ Bregs and, furthermore, to shed light on the signaling pathways that are responsible for their expansion. The results show that dual stimulation by CpG and CD40L for 48 h is optimal for IL-10 induction, and this can be synergistically boosted by IL-21. We identified the CD19+CD27+ memory B cell population as the major source of IL-10+ Bregs. We detected significantly fewer CD19+CD27+IL-10+ cells in RA patients compared with healthy controls, and these were functionally defective in suppressing IFN-γ production by CD4+ T cells in coculture. IL-21 drastically increased the number of IL-10+ Bregs within the CD19+CD27+ and CD19+CD27- populations; furthermore, it induced the appearance of IL-10+Blimp-1+ plasmablasts. Monitoring the phosphorylation of key signaling molecules revealed that activation of ERK, p38, and CREB is indispensable for the induction of IL-10 production, whereas phosphorylation of STAT3 further enhances IL-10 expression in human Bregs. We conclude that CREB and STAT3 are the key transcription factors responsible for the expansion and differentiation of human IL-10-producing Bregs.
Collapse
Affiliation(s)
- Zsuzsanna Bankó
- Department of Immunology, Eötvös Loránd University, Budapest, 1117 Hungary
| | - Judit Pozsgay
- Department of Immunology, Eötvös Loránd University, Budapest, 1117 Hungary
| | - Dániel Szili
- Department of Immunology, Eötvös Loránd University, Budapest, 1117 Hungary
| | - Mária Tóth
- Department of Immunology, Eötvös Loránd University, Budapest, 1117 Hungary
| | - Tamás Gáti
- Buda Hospital of Hospitaller Brothers of St. John, Budapest, 1023 Hungary; and
| | - György Nagy
- Buda Hospital of Hospitaller Brothers of St. John, Budapest, 1023 Hungary; and.,Department of Rheumatology, 3rd Department of Internal Medicine, Semmelweis University, Budapest, 1023 Hungary
| | | | - Gabriella Sármay
- Department of Immunology, Eötvös Loránd University, Budapest, 1117 Hungary;
| |
Collapse
|
28
|
Rapid and strong de novo donor-specific antibody development in a lung transplant recipient: Short communication/case report. Transpl Immunol 2016; 40:17-21. [PMID: 27979771 DOI: 10.1016/j.trim.2016.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 11/24/2022]
Abstract
A 66-yo female patient (typed B*39:01, 44:02) underwent first left single lung transplant (typed B*81:01, 15:17) on 02/07/2016 with negative for DSA in current and historical samples. On 02/17/2016 strong de novo DSA (MFI=15,200, C1q+) to B81 were detected. The recipient has two children typed B*07:02, 44:02 B*27:03, 39:01, and had received multiple vaccinations. Twinrix, Zostavax and MMR vaccines contain viruses grown on live human lung fibroblasts (MRC-5, typed B*07:02, 44:02, and WI-38, typed B*08:01, 58:01). Each dose of vaccine used for injection is known to contain protein components of fibroblasts including HLA. Most likely rapid de novo DSA development is due to booster effect produced by five exposures to mismatched B locus alleles which share the following epitopes: 70IAQ, 65QIA, 65QIA+76esn, 69aa+80n, and 163ew+73te. The later three consist of paired non-self and self eplets. Although likelihood of bystander effect produced by multiple vaccinations is low its impact cannot be ruled out.
Collapse
|
29
|
Mohanram V, Demberg T, Musich T, Tuero I, Vargas-Inchaustegui DA, Miller-Novak L, Venzon D, Robert-Guroff M. B Cell Responses Associated with Vaccine-Induced Delayed SIVmac251 Acquisition in Female Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2016; 197:2316-24. [PMID: 27534560 DOI: 10.4049/jimmunol.1600544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/19/2016] [Indexed: 11/19/2022]
Abstract
An established sex bias in HIV pathogenesis is linked to immune responses. Recently we reported a vaccine-induced sex bias: vaccinated female but not male rhesus macaques exhibited delayed SIV acquisition. This outcome was correlated with SIV Env-specific rectal IgA, rectal memory B cells, and total rectal plasma cells. To uncover additional contributing factors, using samples from the same study, we investigated memory B cell population dynamics in blood, bone marrow, and rectal tissue during immunization and postchallenge; IgG subtypes and Ab avidity; and regulatory B (Breg) cell frequency and function. Few sex differences were seen in Env-specific memory B cell, plasmablast, or plasma cell frequencies in the three compartments. Males had higher IgG Ab titers and avidity indices than females. However, females had elevated levels of Env-specific IgG1, IgG2, and IgG3 Abs compared with males. gp140-specific IgG3 Abs of females but not males were correlated with Ab-dependent cell-mediated cytotoxicity activity against gp120 targets (p = 0.026) and with Ab-dependent phagocytic activity (p = 0.010). IgG3 Ab of females but not males also correlated with decreased peak viremia (p = 0.028). Peripheral blood CD19(+)CD25(+) Breg cells suppressed T cell proliferation compared with CD19(+)CD25(-) cells (p = 0.031) and exhibited increased IL-10 mRNA expression (p = 0.031). Male macaques postvaccination (p = 0.018) and postinfection (p = 0.0048) exhibited higher Breg frequencies than females. Moreover, male Breg frequencies correlated with peak viremia (p = 0.0071). Our data suggest that vaccinated females developed better Ab quality, contributing to better functionality. The elevated Breg frequencies in males may have facilitated SIV acquisition.
Collapse
Affiliation(s)
- Venkatramanan Mohanram
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Thorsten Demberg
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Thomas Musich
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Iskra Tuero
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Diego A Vargas-Inchaustegui
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Leia Miller-Novak
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - David Venzon
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, MD 20892
| | - Marjorie Robert-Guroff
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
30
|
|
31
|
Blumenfeld S, Staun-Ram E, Miller A. Fingolimod therapy modulates circulating B cell composition, increases B regulatory subsets and production of IL-10 and TGFβ in patients with Multiple Sclerosis. J Autoimmun 2016; 70:40-51. [PMID: 27055778 DOI: 10.1016/j.jaut.2016.03.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/20/2016] [Accepted: 03/23/2016] [Indexed: 01/26/2023]
Abstract
Fingolimod, an oral therapeutic agent approved for patients with relapsing-remitting Multiple Sclerosis (MS), has been shown to prevent lymphocyte egress from secondary lymphoid tissues; however the specific drug effect on B cells in fingolimod-treated patients remains to be fully elucidated. We present here a comprehensive analysis on the proportions of B cell subsets in the periphery, and the levels of activation, functional surface markers and cytokine profile of B cells in MS patients, following initiation of fingolimod therapy, using flow cytometry and cytokine bead array. Fingolimod therapy increased the ratio of naïve to memory cells, elevated the percentage of plasma cells and highly increased the proportion of transitional B cells as well as additional regulatory subsets, including: IL10(+), CD25(+) and CD5(+) B cells. The percentage of activated CD69(+) cells was highly elevated in the remaining circulating B cells, which produced increased levels of IL10, TGFβ, IL6, IL4, LTα, TNFα and IFNγ cytokines, with an overall increased ratio of TGFβ to pro-inflammatory cytokines. Furthermore, fingolimod therapy reduced ICAM-1(+) cells, suggesting a possible reduction in antigen-presenting capacity. Phosphorylated-fingolimod was shown in vitro to reduce S1PR1 RNA and protein, to slightly increase viability and to activate anti-apoptotic Bcl2 in transformed B cells of patients with MS. In conclusion, fingolimod therapy modulates significantly the composition of circulating B cells, promoting regulatory subsets and an anti-inflammatory cytokine repertoire.
Collapse
Affiliation(s)
- Shiri Blumenfeld
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Elsebeth Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ariel Miller
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel.
| |
Collapse
|
32
|
Bromberek JL, Rout ED, Agnew MR, Yoshimoto J, Morley PS, Avery AC. Breed Distribution and Clinical Characteristics of B Cell Chronic Lymphocytic Leukemia in Dogs. J Vet Intern Med 2016; 30:215-22. [PMID: 26740174 PMCID: PMC4913636 DOI: 10.1111/jvim.13814] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 10/27/2015] [Accepted: 11/17/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND B-cell chronic lymphocytic leukemia (B-CLL) is the most common hematopoietic malignancy in humans in the developed world and the primary risk factor is genetic. Dogs also develop B-CLL, but there is no systematic description of the disease in dogs. Understanding the epidemiology of B-CLL in dogs may help practitioners recognize the disease and position the dog as a model for future genetic studies. OBJECTIVES To describe B-CLL presentation in dogs, its clinicopathologic findings, and breed predisposition. ANIMALS Four hundred and ninety-one dogs with B-CLL and 5,673 control dogs with suspicion of a lymphoproliferative disorder (LPD). METHODS Retrospective cross-sectional study of dogs for which samples were submitted to the Colorado State University Clinical Immunology Laboratory for immunophenotyping between 2010 and 2014. To assess breed predilection, dogs with B-CLL were compared to those with suspicion of other LPDs using logistic regression. RESULTS The median age was 11 years with no sex predilection. Half of the dogs presented with peripheral lymphadenopathy or splenomegaly and 26% had anemia. Eleven small-breed dogs had significantly increased odds of B-CLL. In addition, English Bulldogs had an increased risk and a unique presentation: these dogs were diagnosed at a median of 6 years and expressed lower class II MHC and CD25. CONCLUSIONS B-cell chronic lymphocytic leukemia is overrepresented in small-breed dogs. Future genetic studies of these breeds may identify genetic risk factors. The unique presentation of English Bulldogs provides evidence of multiple forms of this disease. Additional studies are necessary to determine whether presenting signs are associated with survival.
Collapse
Affiliation(s)
- J L Bromberek
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - E D Rout
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - M R Agnew
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - J Yoshimoto
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - P S Morley
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - A C Avery
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
33
|
Shao HY, Huang JY, Lin YW, Yu SL, Chitra E, Chang CK, Sung WC, Chong P, Chow YH. Depletion of regulatory T-cells leads to moderate B-cell antigenicity in respiratory syncytial virus infection. Int J Infect Dis 2015; 41:56-64. [PMID: 26555647 DOI: 10.1016/j.ijid.2015.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/15/2015] [Accepted: 10/31/2015] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The regulation of the immunopathology of respiratory syncytial virus (RSV) by regulatory T-cells (CD4(+)CD25(+)Foxp3(+); Tregs) is not understood. METHODS To deduce the same, Tregs were depleted in BALB/c mice by injecting anti-CD25 antibody followed by RSV infection (anti-CD25-RSV mice). RESULTS In this model, a decrease in anti-fusion (F) antibody and neutralizing activity, and an increase in anti-nucleocapsid (N) antibody in serum, were seen. Decreased antibody-dependent cell-mediated cytotoxicity (ADCC) activity, increased IgG2a, and an influx of activated CD8(+) T-cells into the lungs were also observed. Co-culture of splenic CD45RA(+) B-cells from RSV-infected normal mice with CD4(+) cells isolated from anti-CD25-RSV mice (B/CD4) increased anti-F antibody secretion. The inclusion of CD25(+) Tregs isolated from isotype Ig-RSV mice into the B/CD4 co-culture substantially enhanced the frequency of anti-F antibody production. However, the same effect was not seen in the co-culture of CD45RA(+) B-cells with dendritic cells (DCs) (B/DCs) or CD8(+) cells (B/CD8) that were obtained from anti-CD25-RSV mice. The transfer of enriched B-cells from anti-CD25-RSV mice into RSV-infected SCID mice increased severe lung inflammation associated with the increased viral load and eosinophil number. CONCLUSIONS These results indicate that Tregs modulate B-cell activity, particularly in producing F-specific neutralizing antibodies, to regulate RSV-mediated exacerbated diseases.
Collapse
Affiliation(s)
- Hsiao-Yun Shao
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan; Graduate Program of Biotechnology in Medicine, Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Juo-Yu Huang
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Yi-Wen Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Ebenezer Chitra
- School of Medical Sciences, Division of Human Biology, International Medical University, Kuala Lumpur, Malaysia
| | - Ching-Kun Chang
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan; Graduate School of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Wang-Chou Sung
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Pele Chong
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan; Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Room No. R1-7033, No. 35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan; Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
34
|
Rincón-Arévalo H, Sanchez-Parra CC, Castaño D, Yassin L, Vásquez G. Regulatory B Cells and Mechanisms. Int Rev Immunol 2015; 35:156-76. [PMID: 25793964 DOI: 10.3109/08830185.2015.1015719] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regulatory B cells have gained prominence in their role as modulators of the immune response against tumors, infectious diseases, and autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis, among others. The concept of regulatory B cells has been strongly associated with interleukin (IL)-10 production; however, there is growing evidence that supports the existence of other regulatory mechanisms, such as the production of transforming growth factor β (TGF-β), induced cell death of effector T cells, and the induction of CD4(+)CD25(-)Foxp3(+) regulatory T cells. The regulatory function of B cells has been associated with the presence and activation of molecules such as CD40, CD19, CD1d, and BCR. Alterations in signaling by any of these pathways leads to a marked defect in regulatory B cells and to increased clinical symptoms and proinflammatory signs, both in murine models and in autoimmune diseases in humans. B cells mainly exert their regulatory effect through the inhibition of proliferation and production of proinflammatory mediators, such as TNF-α, IFN-γ, and IL-17 by CD4(+) T cells. A better understanding of how regulatory B cells function will offer new perspectives with regard to the treatment of various human diseases.
Collapse
Affiliation(s)
- Héctor Rincón-Arévalo
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Claudia C Sanchez-Parra
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Diana Castaño
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Lina Yassin
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia.,b Grupo de Ciencias Básicas, Facultad de Medicina , Universidad CES , Medellín , Colombia
| | - Gloria Vásquez
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| |
Collapse
|
35
|
Sinkora M, Sinkorova J. B Cell Lymphogenesis in Swine Is Located in the Bone Marrow. THE JOURNAL OF IMMUNOLOGY 2014; 193:5023-32. [DOI: 10.4049/jimmunol.1401152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Amu S, Lavy-Shahaf G, Cagigi A, Hejdeman B, Nozza S, Lopalco L, Mehr R, Chiodi F. Frequency and phenotype of B cell subpopulations in young and aged HIV-1 infected patients receiving ART. Retrovirology 2014; 11:76. [PMID: 25213015 PMCID: PMC4172851 DOI: 10.1186/s12977-014-0076-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/23/2014] [Indexed: 01/08/2023] Open
Abstract
Background Aged individuals respond poorly to vaccination and have a higher risk of contracting infections in comparison to younger individuals; whether age impacts on the composition and function of B cell subpopulations relevant for immune responses is still controversial. It is also not known whether increased age during HIV-1 infection further synergizes with the virus to alter B cell subpopulations. In view of the increased number of HIV-1 infected patients living to high age as a result of anti-retroviral treatment this is an important issue to clarify. Results In this work we evaluated the distribution of B cell subpopulations in young and aged healthy individuals and HIV-1 infected patients, treated and naïve to treatment. B cell populations were characterized for the expression of inhibitory molecules (PD-1 and FcRL4) and activation markers (CD25 and CD69); the capacity of B cells to respond to activation signals through up-regulation of IL-6 expression was also evaluated. Increased frequencies of activated and tissue-like memory B cells occurring during HIV-1 infection are corrected by prolonged ART therapy. Our findings also reveal that, in spite of prolonged treatment, resting memory B cells in both young and aged HIV-1 infected patients are reduced in number, and all memory B cell subsets show a low level of expression of the activation marker CD25. Conclusions The results of our study show that resting memory B cells in ART-treated young and aged HIV-1 infected patients are reduced in number and memory B cell subsets exhibit low expression of the activation marker CD25. Aging per se in the HIV-1 infected population does not worsen impairments initiated by HIV-1 in the memory B cell populations of young individuals. Electronic supplementary material The online version of this article (doi:10.1186/s12977-014-0076-x) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
de Andrés C, Tejera-Alhambra M, Alonso B, Valor L, Teijeiro R, Ramos-Medina R, Mateos D, Faure F, Sánchez-Ramón S. New regulatory CD19(+)CD25(+) B-cell subset in clinically isolated syndrome and multiple sclerosis relapse. Changes after glucocorticoids. J Neuroimmunol 2014; 270:37-44. [PMID: 24662004 DOI: 10.1016/j.jneuroim.2014.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/20/2023]
Abstract
In multiple sclerosis (MS), the immune damage to the central nervous system results from the net balance between self-reactive and immunoregulatory cells, among other factors. We identified novel perforin-expressing regulatory B-cells (BReg) in patients with clinically isolated syndrome, significantly enriched within the cerebrospinal fluid when compared to peripheral blood, of memory B cell phenotype (CD19(+)CD25(+), CD19(+)CD25(+)FoxP3(+) and CD19(+)FoxP3(+), p=0.007, p=0.06 and p=0.03, respectively). These BReg subsets were also higher in relapsing-remitting MS during relapse symptoms than in non-clinically active MS patients. Suppressive effects by CD19(+)CD25(+hi) BReg on CD4(+) T cell proliferation seem to be mediated at least in part by perforin/granzyme pathway. To our knowledge, this is the first report that shows cytolytic perforin/granzyme granule storage in B cells; the interesting point is its involvement on BReg cell immunosuppressive mechanisms, similarly to that in TReg cells. Our data may extend the understanding of pathophysiological processes in MS immunoregulation.
Collapse
Affiliation(s)
- Clara de Andrés
- Department of Neurology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain.
| | - Marta Tejera-Alhambra
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Bárbara Alonso
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Lara Valor
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Roseta Teijeiro
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Rocío Ramos-Medina
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Dolores Mateos
- Department of Neurology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain
| | | | - Silvia Sánchez-Ramón
- Department of Immunology, Hospital General Universitario Gregorio Marañón, c/Doctor Esquerdo 46, 28007 Madrid, Spain; Department of Clinical Immunology, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
38
|
Brisslert M, Rehnberg M, Bokarewa MI. Epstein-Barr virus infection transforms CD25+ B cells into antibody-secreting cells in rheumatoid arthritis patients. Immunology 2014; 140:421-9. [PMID: 23844744 DOI: 10.1111/imm.12151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 12/23/2022] Open
Abstract
Epstein-Barr virus (EBV) infection may initiate production of autoantibodies and development of cancer and autoimmune diseases. Here we outline phenotypic and functional changes in B cells of patients with rheumatoid arthritis (RA) related to EBV infection. The B-cell phenotype was analysed in blood and bone marrow (BM) of RA patients who had EBV transcripts in BM (EBV(+) , n = 13) and in EBV(-) (n = 22) patients with RA. The functional effect of EBV was studied in the sorted CD25(+) and CD25(-) peripheral B cells of RA patients (n = 18) and healthy controls (n = 9). Rituximab treatment results in enrichment of CD25(+) B cells in peripheral blood (PB) of EBV(+) RA patients. The CD25(+) B-cell subset displayed a more mature phenotype accumulating IgG-expressing cells. It was also enriched with CD27(+) and CD95(+) cells in PB and BM. EBV stimulation of the sorted CD25(+) B cells in vitro induced a polyclonal IgG and IgM secretion in RA patients, while CD25(+) B cells of healthy subjects did not respond to EBV stimulation. CD25(+) B cells were enriched in PB and synovial fluid of RA patients. EBV infection affects the B-cell phenotype in RA patients by increasing the CD25(+) subset and by inducing their immunoglobulin production. These findings clearly link CD25(+) B cells to the EBV-dependent sequence of reactions in the pathogenesis of RA.
Collapse
Affiliation(s)
- Mikael Brisslert
- EULAR Centre of Excellence, Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | | |
Collapse
|
39
|
Todd SK, Pepper RJ, Draibe J, Tanna A, Pusey CD, Mauri C, Salama AD. Regulatory B cells are numerically but not functionally deficient in anti-neutrophil cytoplasm antibody-associated vasculitis. Rheumatology (Oxford) 2014; 53:1693-703. [PMID: 24729396 DOI: 10.1093/rheumatology/keu136] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES B cells are central to the pathology of ANCA-associated vasculitis (AAV), a disease characterized by autoantibodies and effectively treated by rituximab. In addition to promoting inflammation, a subset of B cells act to suppress harmful autoimmune responses (Breg). The balance of effector and regulatory B cell subsets in AAV is not known. This study was conducted to assess the relative frequency of these subsets during different states of disease activity. METHODS B memory (Bmem), naive (Bnaive) and regulatory (Breg) subsets were defined by their relative expression of CD24 and CD38. Function was assessed by cytokine production and suppressive action on CD4(+) Th1 activation evaluated in a co-culture system. RESULTS Compared with healthy controls, the frequency of Breg (CD24(hi)CD38(hi)) was significantly reduced during disease remission in both proteinase 3 (PR3)- and MPO-ANCA patients and during acute disease in PR3-ANCA patients, while the frequency of memory cells (CD24(hi)CD38(lo)) was reduced during active disease and restored during remission. Breg cell frequency showed a positive correlation, while Bmem had an inverse correlation with IL-10 production in vitro. B and T cell co-cultures revealed that memory and naive B cell subsets augmented Th1 activation in vitro, which was prevented by Breg, and this pattern did not differ between remission AAV patients and controls. CONCLUSION In remission there is a numerical, but not functional, deficiency in Breg and preservation of Bmem associated with reduced IL-10 production and increased Th1 activation in vitro. This imbalance may contribute to the high rate of relapse observed in AAV, especially in PR3-ANCA patients.
Collapse
Affiliation(s)
- Sarah Katrina Todd
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Ruth J Pepper
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Juliana Draibe
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Anisha Tanna
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Charles D Pusey
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Claudia Mauri
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK
| | - Alan D Salama
- UCL Centre for Nephrology, Royal Free Hospital, Renal and Vascular Inflammation Group, Imperial College London, Hammersmith Hospital and Centre for Rheumatology, University College London, London, UK.
| |
Collapse
|
40
|
Vadasz Z, Haj T, Balbir A, Peri R, Rosner I, Slobodin G, Kessel A, Toubi E. A regulatory role for CD72 expression on B cells in systemic lupus erythematosus. Semin Arthritis Rheum 2013; 43:767-71. [PMID: 24461079 DOI: 10.1016/j.semarthrit.2013.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/07/2013] [Accepted: 11/22/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND B regulatory cells and their regulatory products/markers, such us semaphorin 3A (sema3A) and its receptor NP-1, FcγIIB, IL-10, and others, act at the very base of self-tolerance, maintenance, and prevention of autoimmune disease development. OBJECTIVES The aim of the present study was to assess the involvement of CD72, a regulatory receptor on B cells, in systemic lupus erythematosus (SLE). In addition, the potential of soluble sema3A in enhancing the expression of CD72 on B cells of SLE patients was investigated. RESULTS CD72 expression on activated B cells of SLE patients was significantly lower than that of normal controls. This lower expression of CD72 in SLE patients correlated inversely with SLE disease activity and was associated with lupus nephritis, the presence of anti-dsDNA antibodies, and low levels of complement. Co-culture of purified B cells from healthy controls with condition-media containing recombinant sema3A resulted in significant enhancement of CD72. Similar enhancement of CD72 on activated B cells from SLE patients, though significant, was still lower than in normal individuals. CONCLUSIONS The lower expression of CD72 on activated B cells from SLE patients correlates with SLE disease activity, lupus nephritis, the presence of anti-dsDNA antibodies, and low levels of complement. The improvement of CD72 expression following the addition of soluble semaphorin 3A suggests that CD72 may be useful as a biomarker to be followed during the treatment of SLE.
Collapse
Affiliation(s)
- Zahava Vadasz
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Tharwat Haj
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Alexandra Balbir
- Rheumatology Unit, Rambam Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Regina Peri
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Itzhak Rosner
- Rheumatology Unit, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Gleb Slobodin
- Rheumatology Unit, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Aharon Kessel
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Elias Toubi
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel; Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
41
|
Lai ZW, Borsuk R, Shadakshari A, Yu J, Dawood M, Garcia R, Francis L, Tily H, Bartos A, Faraone SV, Phillips P, Perl A. Mechanistic target of rapamycin activation triggers IL-4 production and necrotic death of double-negative T cells in patients with systemic lupus erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:2236-46. [PMID: 23913957 PMCID: PMC3777662 DOI: 10.4049/jimmunol.1301005] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is recognized as a sensor of mitochondrial dysfunction and effector of T cell lineage development; however, its role in autoimmunity, including systemic lupus erythematosus, remains unclear. In this study, we prospectively evaluated mitochondrial dysfunction and mTOR activation in PBLs relative to the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) during 274 visits of 59 patients and 54 matched healthy subjects. Partial least square-discriminant analysis identified 15 of 212 parameters that accounted for 70.2% of the total variance and discriminated lupus and control samples (p < 0.0005); increased mitochondrial mass of CD3(+)/CD4(-)/CD8(-) double-negative (DN) T cells (p = 1.1 × 10(-22)) and FOXP3 depletion in CD4(+)/CD25(+) T cells were top contributors (p = 6.7 × 10(-7)). Prominent necrosis and mTOR activation were noted in DN T cells during 15 visits characterized by flares (SLEDAI increase ≥ 4) relative to 61 visits of remission (SLEDAI decrease ≥ 4). mTOR activation in DN T cells was also noted at preflare visits of SLE patients relative to those with stable disease or healthy controls. DN lupus T cells showed increased production of IL-4, which correlated with depletion of CD25(+)/CD19(+) B cells. Rapamycin treatment in vivo blocked the IL-4 production and necrosis of DN T cells, increased the expression of FOXP3 in CD25(+)/CD4(+) T cells, and expanded CD25(+)/CD19(+) B cells. These results identify mTOR activation to be a trigger of IL-4 production and necrotic death of DN T cells in patients with SLE.
Collapse
Affiliation(s)
- Zhi-Wei Lai
- Division of Rheumatology, Department of Medicine, College of Medicine, Upstate Medical University, State University of New York, Syracuse, NY 13210, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vadasz Z, Haj T, Kessel A, Toubi E. B-regulatory cells in autoimmunity and immune mediated inflammation. FEBS Lett 2013; 587:2074-8. [PMID: 23707422 DOI: 10.1016/j.febslet.2013.05.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/08/2013] [Indexed: 12/11/2022]
Abstract
B cells are a source of inhibitory cytokines such as IL-10 and TGF-β. The ability of being B-regulatory cells (B-regs) was shown to be driven by many stimulatory factors such as toll-like receptors, CD40-ligand and others. However, the characterization of B-regs is still underway. B-regs express high levels of CD25, CD86, IL-10 and TGF-β. In addition, we propose that semaphorin3A is a regulatory molecule and therefore can serve as one of the additional markers for B-regs. This subset of B cells was able to suppress Th1 proliferation, thus contributing to the maintenance of self-tolerance. Finally, the potentiation of B-reg function should become the aim of many immunomodulatory drugs, contributing to a better control of autoimmune diseases.
Collapse
Affiliation(s)
- Zahava Vadasz
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | |
Collapse
|
43
|
Toubi E, Nussbaum S, Staun-Ram E, Snir A, Melamed D, Hayardeny L, Miller A. Laquinimod modulates B cells and their regulatory effects on T cells in multiple sclerosis. J Neuroimmunol 2012; 251:45-54. [PMID: 22846497 DOI: 10.1016/j.jneuroim.2012.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 11/19/2022]
Abstract
Laquinimod is an orally administered drug under development for the treatment of Multiple Sclerosis (MS), lacking a fully elucidated mode of action. We assessed the immunomodulatory effects of laquinimod in vitro on human B cells from healthy or MS patients, cultured alone or with CD4(+) T cells. Laquinimod modulated B cell markers, mainly by increasing the regulatory ones CD25, IL10 and CD86, and decreased IL4, while increasing IL10 and TGFβ in both B and T cells, in a B cell-mediated manner. These findings shed additional light on the mechanisms underlying the effects of laquinimod in MS and potentially other immune-mediated diseases.
Collapse
Affiliation(s)
- Elias Toubi
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
44
|
Kessel A, Haj T, Peri R, Snir A, Melamed D, Sabo E, Toubi E. Human CD19(+)CD25(high) B regulatory cells suppress proliferation of CD4(+) T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells. Autoimmun Rev 2011; 11:670-7. [PMID: 22155204 DOI: 10.1016/j.autrev.2011.11.018] [Citation(s) in RCA: 230] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2011] [Indexed: 12/12/2022]
Abstract
Studies in both animal models and humans have shown a subset of B cells behaving as immuno-regulatory cells, being a source of inhibitory cytokines such as IL-10 and TGF-β. Our aims were to establish the presence of human B regulatory (Breg) cells and to assess their ability to suppress proliferation of CD4(+) T cells and to mediate T regulatory (Treg) cells' properties. For this purpose, human Breg, CD4(+) T and Treg cells were purified using magnetic microbeads. CFSE-labeled CD4(+) T cells were stimulated and cultured alone or with Breg cells. Their proliferative response was determined 72 hours later based on the CFSE staining. In parallel, Treg cells were cultured alone or with Breg cells in different conditions for 24 hours, and then stained and analyzed for Foxp3 and CTLA-4 expression. We found that, the co-culture of Breg cells (defined as CD25(high) CD27(high) CD86(high) CD1d(high) IL-10(high) TGF-β(high)) with autologous stimulated CD4(+) T cells decreased significantly (in a dose-dependent way) the proliferative capacity of CD4(+) T cells. Furthermore, Foxp3 and CTLA-4 expression in Treg cells were enhanced by non-stimulated and further by ODN-CD40L stimulated Breg cells. The regulatory function of Breg cells on Treg cells was mainly dependent on a direct contact between Breg and Treg cells, but was also TGF-β but not IL-10 dependent. In conclusion, human Breg cells decrease the proliferation of CD4(+) T cells and also enhance the expression of Foxp3 and CTLA-4 in Treg cells by cell-to-cell contact.
Collapse
Affiliation(s)
- Aharon Kessel
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Confirmation of clinical tolerance requires the cessation of immunosuppressive drugs, which evoke immune reactivation and allograft rejection in all but the rare individuals who successfully transition into a state of operational transplantation tolerance. Therefore, the safe conduct of trials in transplantation tolerance requires two conditions: a sensitive and reliable means to identify individuals still being maintained on immunosuppression who are most likely to exhibit tolerance after immunosuppression is withdrawn and a noninvasive means that assesses the quality or robustness of the tolerant (TOL) state. Two recent studies attempting to identify a gene signature in peripheral blood of spontaneously TOL kidney transplant recipients made the unexpected observation that TOL, but not immune-suppressed transplant recipients, exhibited enriched B cells and B-cell transcripts in their blood. In concert with the emerging appreciation of a specialized subset of regulatory B cells (Bregs) that possess immune-modulatory function, these observations raise the possibility that Bregs play a critical role in the maintenance of tolerance to renal allografts in transplant patients. This review summarizes these recent findings and speculates on the relationship of Bregs to the maintenance of transplantation tolerance.
Collapse
Affiliation(s)
- A S Chong
- Department of Surgery, Section of Transplantation, The University of Chicago, IL, USA.
| | | |
Collapse
|
46
|
Santos RV, Lin KC, Mansfield K, Wachtman LM. Specific pathogen-free status alters immunophenotype in rhesus macaques: implications for the study of simian immunodeficiency virus. AIDS Res Hum Retroviruses 2011; 27:1033-42. [PMID: 21391843 DOI: 10.1089/aid.2010.0155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The repertoire of viruses to which research primates are exposed, even in the absence of clinical disease, may contribute to experimental confounding. In this study we examined whether standard specific pathogen-free (SPF) rhesus macaques exposed to a wider spectrum of enzootic viruses and expanded SPF macaques derived to exclude a greater number of viral agents would display alterations in immune activation or immune cell populations. Given the impact of immunophenotype on human immunodeficiency virus (HIV) progression and the importance of the simian immunodeficiency virus (SIV) model for the study of HIV pathogenesis, we elected to additionally examine the impact of SPF status on the capacity of peripheral blood mononuclear cells (PBMCs) to support SIV replication. The expanded SPF group displayed significant immune alterations including increased serum interleukin (IL)-15 and a greater in vitro elaboration of GM-CSF, IL1ra, VEGF, IL-10, IL12/23, and MIP-1b. Consistent with reduced viral antigenic exposure in expanded SPF macaques, decreased CD4(+) and CD8(+) transitional and effector memory (T(EM)) cell populations were observed. Expanded SPF PBMC cultures also demonstrated an increased peak (192.61 ng/ml p27) and area under the curve in in vitro SIV production (1968.64 ng/ml p27) when compared to standard SPF macaques (99.32 ng/ml p27; p=0.03 and 915.17 ng/ml p27; p=0.03, respectively). In vitro SIV replication did not correlate with CD4(+) T(EM) cell counts but was highly correlated with serum IL-15 in the subset of animals examined. Findings suggest that an altered immunophenotype associated with the maintenance of primates under differing levels of bioexclusion has the potential to impact the outcome of SIV studies and models for which the measurement of immunologic endpoints is critical.
Collapse
Affiliation(s)
- Rosemary V. Santos
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Kuei-Chin Lin
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Keith Mansfield
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| | - Lynn M. Wachtman
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts
| |
Collapse
|
47
|
Phenotype and function of CD25-expressing B lymphocytes isolated from human umbilical cord blood. Clin Dev Immunol 2011; 2011:481948. [PMID: 21941578 PMCID: PMC3175414 DOI: 10.1155/2011/481948] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 07/07/2011] [Accepted: 07/07/2011] [Indexed: 11/17/2022]
Abstract
BACKGROUND We have shown that approximately 30% of human peripheral blood B-cells express CD25. B cells expressing CD25 display a mature phenotype belonging to the memory B-cell population and have a better proliferative and antigen-presenting capacity. The aim of the present study was to characterize the CD25-expressing subset of B cells in human cord blood. MATERIAL AND METHODS Mononuclear cell fraction from human cord blood (n=34) and peripheral adult blood (n=22) was sorted into CD20+CD25+ and CD20+CD25- B-cell populations. Phenotype and function of these B-cell populations were compared using flow cytometry, proliferation, cytokine production, and immunoglobulin secretion. RESULTS CD25-expressing B cells are a limited population of cord blood mononuclear cells representing 5% of the CD20+ B cells. They are characterised by high expression of CD5 in cord blood and CD27 in adult blood. CD25-expressing B cells express a functional IL-2 receptor and high levels of CC-chemokine receptors and spontaneously produce antibodies of IgG and IgM subclass. CONCLUSIONS CD25 expression is a common denominator of a specific immunomodulatory B-cell subset ready to proliferate upon IL-2 stimulation, possibly ready to migrate and home into the peripheral tissue for further differentiation/action.
Collapse
|
48
|
B cell immunosenescence: different features of naive and memory B cells in elderly. Biogerontology 2011; 12:473-83. [PMID: 21879287 DOI: 10.1007/s10522-011-9353-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 08/02/2011] [Indexed: 01/10/2023]
Abstract
Elderly people show a reduced protection against new infections and a decreased response to vaccines as a consequence of impairment of both cellular and humoral immunity. In this paper we have studied memory/naïve B cells in the elderly, evaluating surface immunoglobulin expression, production of the pro- and anti-inflammatory cytokines, tumor necrosis factor (TNF)-α and interleukin (IL)-10, and presence of somatic hypermutation, focusing on the IgG(+)IgD(-)CD27(-) double negative (DN) B cells that are expanded in the elderly. Our results show that naïve B cells from young donors need a sufficiently strong stimulus to be activated "in vitro", while naïve B cells from old subjects are able to produce IL-10 and TNF-α when stimulated "physiologically" (α-CD40/IL-4), suggesting that these cells might play a role in the control of the immuno-inflammatory environment in the elderly. In addition, in the elderly there is an accumulation of DN B cells with a reduced rate of somatic hypermutation. Thus, DN B lymphocytes may be exhausted cells that are expanded and accumulate as a by-product of persistent stimulation or impaired germinal center formation.
Collapse
|
49
|
Abstract
We previously reported that a bioengineered interface consisting of a nano-barrier membrane (NB-LVF4) used as an artificial interface between skin allografts and wound surfaces significantly prolonged graft survival without immunosuppression. We now evaluated whether NB-LVF4 could serve as a targeted drug delivery system to further improve outcomes. Fibroblast growth factor-1 (FGF-1) was selected for its known function as a wound hormone. Full-thickness 8-mm skin grafts were cross-transplanted between out-bred mice. Controls were transplanted without treatment. In test groups, the NB-LVF4, with or without FGF-1, was applied to both basolateral skin and wound surfaces with polymerization resulting in a tridimensional transparent membrane. The mice were evaluated for T-cell activation and development of donor-specific antibody. Rejection occurred in controls by 7 days. NB-LVF4 treatment, with or without FGF-1, was found to significantly prolong allograft survival (27 and 28 days, respectively [p < 0.05]). Untreated controls stimulated 10-fold shift in the number of circulating CD4+ cells while test groups exhibited substantially reduced shifts in CD4+ cells. The group treated with FGF-1 did not develop donor-specific antibody. Treatment with the NB-LVF4 membrane delays the onset of allograft rejection in the absence of systemic immunosuppression. FGF-1 appears to prevent the development of a humoral response by preventing B cell activation.
Collapse
|
50
|
Induction of B cell responses upon experimental infection of neonatal calves with Mycobacterium avium subsp. paratuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1139-49. [PMID: 21543587 DOI: 10.1128/cvi.00058-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The objective of this study was to determine if experimental infection of neonatal calves with Mycobacterium avium subsp. paratuberculosis would invoke changes in the percentages of total B cells in the peripheral blood mononuclear cell population and of subpopulations of B cells as determined by CD5, CD25, and CD45RO markers during a 12-month period. Experimental infection groups included control (noninfected), oral (infected with M. avium subsp. paratuberculosis strain K-10), oral/DXM (pretreatment with dexamethasone before oral inoculation), i.p. (intraperitoneal inoculation), and oral/M (oral inoculation with mucosal scrapings from a cow with clinical disease) groups. Over the course of the study, the percentages of total B cells in nonstimulated and antigen-stimulated cell cultures increased for oral and i.p. group calves, with the highest percentages noted at 3 and 6 months. Oral/M group calves had increased percentages of activated B cells, as determined by CD5(dim) and CD5(bright) markers, at 9 and 12 months. Experimental infection by all methods resulted in increased expression of CD25(+) and CD45RO(+) B cells early in the study, but the most significant results were observed at 12 months for oral/DXM and oral/M group calves. Immunoblot analyses with a whole-cell sonicate of M. avium subsp. paratuberculosis demonstrated the most reactivity with sera from i.p. group calves and the least reactivity with sera from oral group calves. Further evidence of M. avium subsp. paratuberculosis-specific antibody responses in the i.p. group calves was demonstrated using the ethanol vortex enzyme-linked immunosorbent assay (EvELISA) method. In summary, an induction of B cell responses was noted after experimental infection with M. avium subsp. paratuberculosis, with differences in responses noted according to the method of experimental inoculation.
Collapse
|