Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Heart Lung Transplant ; 40(12): 1502-1516, 2021 12.
Article in English | MEDLINE | ID: mdl-34742645

ABSTRACT

BACKGROUND: Regulatory B cells (Bregs) play an important role in maintaining immune homeostasis and have the potential to induce tolerance. Previous work has found that Breg cells are involved in heart transplantation tolerance. However, the effect of Breg on the transplantation tolerance and the underlying mechanisms remain to be clarified. METHODS: Using a within-species heart transplantation model, we aimed to investigate the role of CD19+CD5+CD1dhigh Bregs isolated from transplanted mice in preventing transplant rejection in vivo. We also explored the effects of CD40 and tumor necrosis factor receptor-associated factor 6 (TRAF6) ubiquitin ligase on Breg-mediated prolongation of survival in heart transplant (HT) mice, and the regulatory effects of downstream Cdk4 and Cdk6 proteins on dendritic cells (DCs), which clarified the function and molecular mechanism of Breg cells in HT mice. RESULTS: Our data suggest that adoptive transfer of the transplanted Bregs served as an effective tolerance-inducing mechanism in HT mice and was involved in the CD40-TRAF6 signaling pathway in DCs. Moreover, DCs collected from the Breg treated HT mice also prolonged the survival of HT mice. Furthermore, DC-specific knockout of TRAF6 diminished Breg-mediated prolongation of survival in HT mice. Interestingly, gut microbes from donors increased the survival of cardiac allografts both in both the absence and presence of Bregs but were not implicated in CD40-TRAF6 signaling. CONCLUSIONS: These findings reveal a role of Breg cells in the induction of transplantation tolerance through the blockade of the CD40-TRAF6 signaling pathway, which might be used in the treatment of HT in the clinic.


Subject(s)
Adoptive Transfer , B-Lymphocytes, Regulatory/transplantation , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Transplantation Tolerance/physiology , Animals , CD40 Antigens/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Female , Gastrointestinal Microbiome , Graft Rejection/metabolism , Graft Rejection/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism
2.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33729999

ABSTRACT

Autoimmune diseases are characterized by a breakdown of immune tolerance partly due to environmental factors. The short-chain fatty acid acetate, derived mostly from gut microbial fermentation of dietary fiber, promotes antiinflammatory Tregs and protects mice from type 1 diabetes, colitis, and allergies. Here, we show that the effects of acetate extend to another important immune subset involved in tolerance, the IL-10-producing regulatory B cells (B10 cells). Acetate directly promoted B10 cell differentiation from mouse B1a cells both in vivo and in vitro. These effects were linked to metabolic changes through the increased production of acetyl-coenzyme A, which fueled the TCA cycle and promoted posttranslational lysine acetylation. Acetate also promoted B10 cells from human blood cells through similar mechanisms. Finally, we identified that dietary fiber supplementation in healthy individuals was associated with increased blood-derived B10 cells. Direct delivery of acetate or indirect delivery via diets or bacteria that produce acetate might be a promising approach to restore B10 cells in noncommunicable diseases.


Subject(s)
Acetates/metabolism , Acetates/pharmacology , Arthritis, Experimental/therapy , B-Lymphocytes, Regulatory/drug effects , Dietary Fiber/pharmacology , Acetates/blood , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Arthritis, Experimental/immunology , B-Lymphocytes, Regulatory/physiology , B-Lymphocytes, Regulatory/transplantation , Cell Differentiation/drug effects , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Female , Humans , Interleukin-10 , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Neutrophils/cytology , Neutrophils/drug effects , Receptors, G-Protein-Coupled/genetics
3.
Methods Mol Biol ; 2270: 451-467, 2021.
Article in English | MEDLINE | ID: mdl-33479913

ABSTRACT

Transplantation is still the treatment of choice for organ failure; however, allograft induces inflammatory immune responses that require immunosuppressive treatment. The role of regulatory B cells (Bregs) in downregulating inflammation has been reported to be significant in several diseases including transplant rejection. Many reports have analyzed different B-cell subpopulations, including Bregs, in tolerant, stable, and rejecting transplant recipients as well as the influence of immunosuppressant on the frequencies and functions of the different B-cell subsets. In this chapter, the key techniques required to investigate human Breg frequencies and functions in transplant patients are discussed.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/transplantation , Immunophenotyping/methods , ADP-ribosyl Cyclase 1/immunology , CD24 Antigen/immunology , Cell Count , Cell Proliferation/physiology , Female , Humans , Immunosuppressive Agents , Interleukin-10/immunology , Male , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
4.
Cardiovasc Res ; 117(3): 743-755, 2021 02 22.
Article in English | MEDLINE | ID: mdl-32219371

ABSTRACT

AIMS: B cell functions in the process of atherogenesis have been investigated but several aspects remain to be clarified. METHODS AND RESULTS: In this study, we show that follicular regulatory helper T cells (TFR) control regulatory B cell (BREG) populations in Apoe-/- mice models on a high-cholesterol diet (HCD). Feeding mice with HCD resulted in up-regulation of TFR and BREG cell populations, causing the suppression of proatherogenic follicular helper T cell (TFH) response. TFH cell modulation is correlated with the growth of atherosclerotic plaque size in thoracoabdominal aortas and aortic root plaques, suggesting that TFR cells are atheroprotective. During adoptive transfer experiments, TFR cells transferred into HCD mice decreased TFH cell populations, atherosclerotic plaque size, while BREG cell population and lymphangiogenesis are significantly increased. CONCLUSION: Our results demonstrate that, through different strategies, both TFR and TFH cells modulate anti- and pro-atherosclerotic immune processes in an Apoe-/- mice model since TFR cells are able to regulate both TFH and BREG cell populations as well as lymphangiogenesis and lipoprotein metabolism.


Subject(s)
Aorta/immunology , Aortic Diseases/immunology , Atherosclerosis/immunology , B-Lymphocytes, Regulatory/immunology , Cholesterol, Dietary , Diet, High-Fat , Plaque, Atherosclerotic , T Follicular Helper Cells/immunology , Adoptive Transfer , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/transplantation , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Lymphangiogenesis , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , T Follicular Helper Cells/metabolism , T Follicular Helper Cells/transplantation
5.
Front Immunol ; 11: 603288, 2020.
Article in English | MEDLINE | ID: mdl-33343576

ABSTRACT

CD19+CD24hiCD27+ memory Breg cells exhibit decreased abundance in patients with chronic graft-versus-host disease (cGVHD) after liver transplantation and produce less IL-10 than those from patients without cGVHD and healthy donors. Due to the lack of Breg cells and the difficulty in expanding them in vitro, in mouse models and early human clinical trials, the adoptive transfer of Breg cells to autoimmune diseases is greatly restricted. Glycogen synthase kinase 3ß (GSK-3ß) is a multifunctional serine/threonine (ser/thr) protein kinase that can participate in B cell growth, metabolic activity, and proliferation. Phosphoprotein array analysis showed that p-GSK-3ß-s9 was highly expressed in mBreg cells. Furthermore, here, we demonstrated that GSK-3ß expression in mBreg cells is lower than that observed in B cells by flow cytometry. We found that the treatment of B cells with the specific GSK-3ß inhibitor SB216763 can significantly increase the proportion and immunosuppressive function of mBreg cells in vitro. Nuclear factor of activated T cells (NFAT) is one of a pivotal regulator of gene expression in adaptive immune system. Here, we observed that inhibition of GSK-3ß by SB216763 results in enhanced expression of NFATc1 in B cells, which is essential in regulating the ability of B cells to secrete IL-10. By constructing a xGVHD mouse model, we observed that SB216763-treated mBreg cells effectively prevent xenogeneic GVHD. Here we propose a novel strategy using SB216763 to inhibit GSK-3ß and then enhance the proportion and immunosuppressive function of mBreg cells by increasing the expression of NFATc1. This approach may be used as a therapy to ameliorate GVHD and inflammatory diseases.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes, Regulatory/drug effects , CD24 Antigen/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Graft vs Host Disease/prevention & control , Indoles/pharmacology , Lymphocyte Activation/drug effects , Maleimides/pharmacology , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Adult , Aged , Animals , B-Lymphocytes, Regulatory/enzymology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/transplantation , Case-Control Studies , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Glycogen Synthase Kinase 3 beta/metabolism , Graft vs Host Disease/enzymology , Graft vs Host Disease/immunology , Humans , Male , Mice, Inbred NOD , Middle Aged , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phenotype , Signal Transduction
6.
Biomed Pharmacother ; 129: 110450, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768945

ABSTRACT

OBJECTIVES: Astilbin exerts immunoregulatory activities and plays anti-inflammatory effects in inflammation-associated diseases. IL-10-producing B cells are the major subset of regulatory B cells (Bregs) and inhibit inflammation and autoimmune diseases. This study aimed to analyse the inducing effect of astilbin on Bregs and investigate the involved molecular mechanisms. METHODS: The frequencies and activities of IL-10-producing Bregs were observed using the co-treatment of astilbin and lipopolysaccharide (LPS) ex vivo. The protective effect of astilbin/LPS-induced Bregs on dextran sulphate sodium (DSS)-induced colitis was confirmed in vivo. The molecular signalling events of Breg induction were checked via Western blot. CD40-/- and toll-like receptor (TLR) 4-/- B cells were treated with astilbin/LPS to determine the modulatory role of CD40 or TLR4 on astilbin/LPS-induced Bregs. RESULTS: Although astilbin alone could not affect Bregs, the co-treatment of astilbin and LPS remarkably induced CD19+ CD1dhi and CD19+ TIM-1+ cells which produced IL-10 ex vivo. Colonic CD19+ CD1dhi and CD19+ TIM-1+ cells were also increased in astilbin-treated mice with DSS-induced colitis. The adoptive transfer of CD19+ TIM-1+ cells pre-induced by astilbin/LPS directly suppressed the progression of DSS-induced colitis. Combined astilbin and LPS stimulated the STAT3 activation of CD19+ TIM-1+ cells but had no effects on SOCS3, AKT, NF-κB, Erk, JNK nor P38. Inhibiting the STAT3 phosphorylation of CD19+ TIM-1+ cells abolished Breg induction by astilbin/LPS. Furthermore, Breg induction was weakened in CD40-/- B cells with the decrease in STAT3 activation, but had disappeared in TLR4-/- B cells with no STAT3 activation, thereby confirming the indispensable role of TLR4 signalling in the induction of IL-10-producing Bregs. CONCLUSIONS: This study reports the new immunoregulatory role of astilbin for promoting IL-10-producing B cells and suggests the possible use of astilbin in the therapy of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , B-Lymphocytes, Regulatory/drug effects , Colitis/drug therapy , Colon/drug effects , Flavonols/pharmacology , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , STAT3 Transcription Factor/metabolism , Adoptive Transfer , Animals , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/transplantation , CD40 Antigens/deficiency , CD40 Antigens/genetics , Cells, Cultured , Coculture Techniques , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Colon/immunology , Colon/metabolism , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
7.
Front Immunol ; 11: 1051, 2020.
Article in English | MEDLINE | ID: mdl-32547555

ABSTRACT

Corticosteroids are effective therapy for autoimmune diseases but serious adverse effects preclude their prolonged use. However, immune-suppressive biologics that inhibit lymphoid proliferation are now in use as corticosteroid sparing-agents but with variable success; thus, the need to develop alternative immune-suppressive approaches including cell-based therapies. Efficacy of ex-vivo-generated IL-35-producing regulatory B-cells (i35-Bregs) in suppressing/ameliorating encephalomyelitis or uveitis in mouse models of multiple sclerosis or uveitis, respectively, is therefore a promising therapeutic approach for CNS autoimmune diseases. However, i35-Breg therapy in human uveitis would require producing autologous Bregs from each patient to avoid immune-rejection. Because exosomes exhibit minimal toxicity and immunogenicity, we investigated whether i35-Bregs release exosomes that can be exploited therapeutically. Here, we demonstrate that i35-Bregs release exosomes that contain IL-35 (i35-Exosomes). In this proof-of-concept study, we induced experimental autoimmune uveitis (EAU), monitored EAU progression by fundoscopy, histology, optical coherence tomography and electroretinography, and investigated whether i35-Exosomes treatment would suppress uveitis. Mice treated with i35-Exosomes developed mild EAU with low EAU scores and disease protection correlated with expansion of IL-10 and IL-35 secreting Treg cells with concomitant suppression of Th17 responses. In contrast, significant increase of Th17 cells in vitreous and retina of control mouse eyes was accompanied by severe choroiditis, massive retinal-folds, and photoreceptor cell damage. These hallmark features of severe uveitis were absent in exosome-treated mice and visual impairment detected by ERG was modest compared to control mice. Absence of toxicity or alloreactivity associated with exosomes thus makes i35-Exosomes attractive therapeutic option for delivering IL-35 into CNS tissues.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes, Regulatory/immunology , Exosomes/metabolism , Immunotherapy, Adoptive/methods , Interleukins/metabolism , Neurogenic Inflammation/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/therapy , B-Lymphocytes, Regulatory/transplantation , Cells, Cultured , Disease Models, Animal , Humans , Immune Tolerance , Immunomodulation , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Neurogenic Inflammation/therapy , T-Lymphocytes, Regulatory/immunology
8.
J Neurosci ; 40(26): 5105-5115, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32430295

ABSTRACT

The unmet medical need of patients with multiple sclerosis (MS) is the inexorable loss of CNS myelin and latterly neurons leading to permanent neurologic disability. Solicitation of endogenous oligodendrocytes progenitor cells, the precursor of oligodendrocytes, to remyelinate axons may abort the onset of disability. In female mice with experimental autoimmune encephalomyelitis (EAE), a murine model of MS, adoptive transfer of IL-10+ regulatory B cells (Bregs) has been shown to reverse EAE by promoting the expansion of peripheral and CNS-infiltrating IL-10+ T cells. Here, we examined whether Bregs treatment and its bystander effect on regulatory T cells are associated with CNS repair as reflected by oligodendrogenesis and remyelination. We have found that transfusion of Bregs reverses established clinical EAE and that clinical improvement is associated with a significant increase in spinal cord remyelination as reflected by g-ratio analysis within the thoracic and lumbar spine. We further observed in the spinal cords of EAE Bregs-treated mice that CNS resident CD11b/CD45intLy6C- microglia, and infiltrating CD11b+/CD45high monocytes/macrophages content reverts to normal and polarize to a M2-like CD206+ phenotype. Concurrently, there was a substantial increase in neo-oligodendrogenesis as manifest by an increase in CD45-/low CNS cells expressing A2B5, an early marker in oligodendrocytes progenitor cell differentiation as well as GalC+/O1+ premyelinating and myelin basic protein+/myelin oligodendrocyte glycoprotein+ mature oligodendrocytes with reciprocal downregulation of paired related homeobox protein 1. These results demonstrate that the clinical benefit of Bregs is associated with normalization of CNS immune milieu and concurrent activation of oligodendrocyte progenitor cells with subsequent remyelination.SIGNIFICANCE STATEMENT In multiple sclerosis patients, demyelination progresses with aging and disease course, leading to irreversible disability. In this study, we have discovered, using a mouse model of multiple sclerosis, that the transfusion of autologous regulatory B cells (Bregs) is able to ameliorate, cure, and sustain the durable remission of the disease. We show that the adoptive transfer of Bregs dramatically decreased the frequency of myeloid-derived cells, both infiltrating monocytes/macrophages and resident microglia, and converted their phenotype to an immunosuppressive-like phenotype. Moreover, we showed that CNS oligodendrocyte progenitor cells are activated following Bregs treatment and differentiate into myelinating oligodendrocytes, which results in neo-oligodendrogenesis and remyelination of spinal cords.


Subject(s)
B-Lymphocytes, Regulatory/transplantation , Encephalomyelitis, Autoimmune, Experimental/pathology , Myeloid Cells , Oligodendrocyte Precursor Cells , Remyelination/physiology , Animals , B-Lymphocytes, Regulatory/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred C57BL , Neurogenesis/physiology , Spinal Cord/pathology
9.
Int Immunopharmacol ; 78: 106111, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31881524

ABSTRACT

Over the past decade, studies have identified subset of B cells, which play suppressive functions in additions to the conventional functions of B cells: antigen processing and presentation, activation of T cells and antibody productions. Because of their regulatory function, they were named as B regulatory cells (Bregs). Bregs restrict the severity of autoimmune disorders in animal disease models such as experimental autoimmune myocarditis (EAM), experimental autoimmune encephalitis (EAE), and collagen-induced arthritis (CIA) but can contribute to the development of infection and cancer. In humans, the roles of B regulatory cells in autoimmune diseases have not been clearly established because of the inconsistent findings from many researchers. This is believed to arise from the speculated fact that Bregs lack specific marker, which can be used to identify and characterize them in human diseases. The CD19+CD24hiCD38hiCD1dhiB cells have been associated with the regulatory function. Available evidences highlight the relevance of increasing IL-10-producing B cells in autoimmune diseases and the possibility of serving as new therapeutic targets in inflammatory disorders. This review empanels the functions of Bregs in autoimmune diseases in both human and animal models, and further evaluates the possibility of Bregs as therapeutic targets in inflammatory disorders. Consequently, this might help identify possible research gaps, which need to be clarified as researchers speculate the possibility of targeting some subsets of Bregs in the treatment of inflammatory disorders.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes, Regulatory/immunology , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Interleukin-10/metabolism , Adoptive Transfer/methods , Animals , Antigen Presentation/drug effects , Autoimmune Diseases/therapy , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/transplantation , Cell Communication/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunosuppressive Agents/therapeutic use , Inflammation/therapy , Inflammation Mediators/metabolism , Interleukin-10/agonists , Interleukin-10/immunology , Th1 Cells/immunology , Th2 Cells/immunology
10.
Curr Gene Ther ; 19(2): 71-80, 2019.
Article in English | MEDLINE | ID: mdl-31161989

ABSTRACT

Transplantation is the only cure for end-stage organ failure. Current immunosuppressive drugs have two major limitations: 1) non antigen specificity, which increases the risk of cancer and infection diseases, and 2) chronic toxicity. Cell therapy appears to be an innovative and promising strategy to minimize the use of immunosuppression in transplantation and to improve long-term graft survival. Preclinical studies have shown efficacy and safety of using various suppressor cells, such as regulatory T cells, regulatory B cells and tolerogenic dendritic cells. Recent clinical trials using cellbased therapies in solid organ transplantation also hold out the promise of improving efficacy. In this review, we will briefly go over the rejection process, current immunosuppressive drugs, and the potential therapeutic use of regulatory cells in transplantation.


Subject(s)
Cell- and Tissue-Based Therapy/trends , Immunosuppression Therapy/trends , Immunosuppressive Agents/therapeutic use , Organ Transplantation/trends , B-Lymphocytes, Regulatory/transplantation , Dendritic Cells/transplantation , Graft Rejection , Humans , Immune Tolerance/genetics , T-Lymphocytes, Regulatory/transplantation
11.
J Invest Dermatol ; 139(7): 1535-1544.e1, 2019 07.
Article in English | MEDLINE | ID: mdl-30836062

ABSTRACT

In tumor immunity, the participation of IL-10-producing regulatory B cells (Bregs), which play an important role in suppressing immune responses, is unclear. In this study, we demonstrated an increase in B16F10 melanoma growth and a decrease in the proportion of IFN-γ- and TNF-α-secreting tumor-infiltrating CD8+ T cells in B cell-specific PTEN-deficient mice in which Bregs were expanded. The number of tumor-infiltrating Bregs significantly increased in B cell-specific PTEN-deficient mice. More than 50% of tumor-infiltrating B cells consisted of Bregs, predominantly CD19+CD5+CD43+ B1a Bregs, in both B cell-specific PTEN-deficient and control mice. Adoptive B1a B cell transfer, which includes >30% of Bregs, increased melanoma growth, whereas non-B1a B cell transfer, which includes <2% of Bregs, exhibited no effect. In addition, adoptive transfer of B1a B cells from wild-type mice, but not IL-10-/- mice, exacerbated B16F10 melanoma growth. The current study indicates that B1a Bregs negatively regulate anti-melanoma immunity by producing IL-10 and reducing T helper 1 type cytokine production in tumor-infiltrating CD8+ T cells. Therefore, B1a Bregs can be a potentially novel target for immunotherapy of melanomas.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Th1 Cells/immunology , Animals , B-Lymphocyte Subsets/transplantation , B-Lymphocytes, Regulatory/transplantation , Cytokines/metabolism , Humans , Immune Tolerance , Interleukin-10/genetics , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental , PTEN Phosphohydrolase/genetics
12.
Vascul Pharmacol ; 111: 54-61, 2018 12.
Article in English | MEDLINE | ID: mdl-30243560

ABSTRACT

OBJECTIVE: Intranasal immunization with a fusion protein of the ApoB100-derived peptide p210 and the cholera toxin B subunit (CTB-p210) has previously been shown to induce mucosal tolerance and reduce atherosclerosis development, but the exact mode of action remains to be elucidated. Recent studies have indicated an important role for B cells in mucosal tolerance, in particular by induction of regulatory B (Bregs) and T cells (Tregs). In this study, we aimed to investigate if transfer of B cells pulsed with CTB-p210 can protect against atherosclerosis. METHOD AND RESULTS: First, we studied if CTB-p210 can induce Bregs and Tregs in vitro. After pulsing B cells from Apobtm2Sgyldlr-/- or Apoe-/- mice with CTB-p210 for 1 h and co-culturing them with naïve T cells for 48 h, we observed increased expression of membrane bound TGFß/latency-associated peptide (mTGFß/LAP) on B cells and an increased proportion of CD25hiFoxP3+ Tregs. Adoptive transfer of B cells pulsed with CTB-p210 into high-fat diet-fed Apoe-/- mice at 8, 10 and 12 weeks of age, reduced the plaque area in the aorta at 20 weeks of age as compared with control-treated (CTB-pOVA treated B cells or PBS) mice. Moreover, mice receiving p210-CTB treated B cells had increased levels of anti-p210 IgG antibodies. CONCLUSION: Our observations suggest that CTB-p210 pulsed B cells acquire a regulatory phenotype and induce Tregs in vitro. Adoptive transfer of CTB-p210, but not control-treated, B cells into Apoe-/- mice decreased atherosclerosis development.


Subject(s)
Adoptive Transfer , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/transplantation , Cholera Toxin/pharmacology , Immunologic Factors/pharmacology , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Humans , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
Cell Immunol ; 310: 184-192, 2016 12.
Article in English | MEDLINE | ID: mdl-27667759

ABSTRACT

Recently, pancreatic islet transplantation has been shown to be a viable option for the treatment of type 1 diabetes mellitus. However, immune destruction becomes the major impediment to the clinical application of islet transplantation. Here, we evaluated changes affecting multiple types of immune cells and cytokines in allogeneic islet transplantation immunity after the administration of B10 cells alone and explored the regulatory mechanisms of B10 cells in T cell-mediated allograft rejection. In vitro assays, B10 cells significantly decreased the proliferative capacity of CD4+CD25- T cells (13.75%±0.96% vs. 32.76%±0.81%) while enhancing the proliferation of regulatory T cells (Tregs) (26.60%±1.14% vs. 21.52%±0.81%). Furthermore, after the administration of B10 cells in vivo, the frequencies of IL-10+ B cells and Tregs of islet transplant recipients were increased by the CD19+CD5+CD1dhi B cells, and the CD4+/CD8+ and IFN-γ+/IL-17+ ratios were decreased. Serum IL-10 levels were up-regulated, while IFN-γ levels were down-regulated. Grafts from 1 to 5×106 B10 cell-treated recipients exhibited a reduced level of insulitis compared with the untreated controls, although the differences of graft survival times were not statistically significant. In general, in mouse islet allograft rejection, B10 cells may alleviate T cell-mediated immune responses by promoting Treg-cell development and inhibiting Th1 cells activation, via an IL-10-dependent pathway. Development of B10 cell-targeted therapy may be benefit for modulating immune response and provide insight into the signals involved the induction of islet allograft tolerance.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Graft Rejection/immunology , Inflammation/immunology , Islets of Langerhans Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes, Regulatory/transplantation , Cell Proliferation , Cells, Cultured , Graft Rejection/prevention & control , Humans , Immune Tolerance , Inflammation/prevention & control , Interferon-gamma/metabolism , Interleukin-10/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
14.
Clin Exp Immunol ; 184(1): 50-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26671281

ABSTRACT

B7-2(-/-) non-obese diabetic (NOD) mice develop a spontaneous autoimmune polyneuropathy (SAP) that mimics the progressive form of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). In this study, we focused on the role of regulatory T cells (Tregs ) and regulatory B cells (Bregs ) in SAP. We found that deletion of B7-2 in female NOD mice led to a lower frequency and number of Tregs and Bregs in spleens and lymph nodes. Tregs but not Bregs suppressed antigen-stimulated splenocyte proliferation, whereas Bregs inhibited the T helper type 1 (Th1) cytokine response. Both Tregs and Bregs induced an increase in CD4(+) interleukin (IL)-10(+) cells, although less effectively in the absence of B7-2. Adoptive transfer studies revealed that Tregs , but not Bregs , suppressed SAP, while Bregs attenuated disease severity when given prior to symptom onset. B cell deficiency in B cell-deficient (muMT)/B7-2(-/-) NOD mice prevented the development of SAP, which would indicate that the pathogenic role of B cells predominates over its regulatory role in this model. We conclude that Bregs and Tregs control the immunopathogenesis and progression of SAP in a non-redundant fashion, and that therapies aimed at expansion of Bregs and Tregs may be an effective approach in autoimmune neuropathies.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B7-2 Antigen/immunology , Lymph Nodes/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoimmunity , B-Lymphocytes, Regulatory/pathology , B-Lymphocytes, Regulatory/transplantation , B7-2 Antigen/deficiency , B7-2 Antigen/genetics , Cell Proliferation , Disease Models, Animal , Female , Gene Expression , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Inbred NOD , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/genetics , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/pathology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Spleen/pathology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/immunology , Th1 Cells/pathology , Th1-Th2 Balance
15.
J Immunol ; 195(12): 5805-15, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26553075

ABSTRACT

In transplantation tolerance, numerous regulatory populations have the capacity to inhibit allograft rejection; however, their compensatory capacities have never been clearly evidenced. We have previously demonstrated that the tolerogenic effect mediated by CD8(+)CD45RC(low) regulatory T cells (Tregs) in a model of organ transplantation with CD40Ig could be abrogated by permanent depletion of CD8(+) cells that resulted in allograft rejection in half of the recipients. This result demonstrated that CD8(+) Tregs were essential, but also that half of the recipients still survived indefinitely. We also demonstrated that no other regulatory populations, besides CD8(+) Tregs, could induce and maintain allograft tolerance in CD40Ig-treated tolerant animals. In the current study, we analyzed the mechanisms that arose following CD8(+) Treg depletion and allowed establishment of networks of new regulatory cells to maintain allograft survival. We identified regulatory B cells (Bregs) and regulatory myeloid cells (RegMCs) as being responsible of the maintenance of the long-term allograft survival. We demonstrated that both regulatory cell subsets efficiently inhibited antidonor immune responses in adoptively transferred recipients. Although Bregs were induced, they were not essential for the maintenance of the graft as demonstrated in IgM-deficient recipients. In addition, we showed that RegMCs were the most suppressive and acted alone, whereas Bregs activity was associated with increased suppressive activity of other subsets in adoptively transferred recipients. Altogether, to our knowledge, we demonstrated in this study for the first time the emergence of both Bregs and RegMCs following Tregs depletion and highlighted the importance of regulatory cell networks and their synergistic potential in transplantation.


Subject(s)
B-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation , Myeloid Cells/immunology , Adoptive Transfer , Animals , B-Lymphocytes, Regulatory/transplantation , Cell Communication , Cells, Cultured , Immunomodulation , Myeloid Cells/transplantation , Rats , Rats, Inbred Lew , Transplantation Tolerance
16.
Crit Rev Immunol ; 35(1): 49-57, 2015.
Article in English | MEDLINE | ID: mdl-25746047

ABSTRACT

Neuroinflammation contributes to neuronal deficits in neurodegenerative CNS (central nervous system) autoimmune diseases, such as multiple sclerosis and uveitis. The major goal of most treatment modalities for CNS autoimmune diseases is to limit inflammatory responses in the CNS; immune-suppressive drugs are the therapy of choice. However, lifelong immunosuppression increases the occurrence of infections, nephrotoxicity, malignancies, cataractogenesis, and glaucoma, which can greatly impair quality of life for the patient. Biologics that target pathogenic T cells is an alternative approach that is gaining wide acceptance as indicated by the popularity of a variety of Food and Drug Administration (FDA)-approved anti-inflammatory compounds and humanized antibodies such as Zenapax, Etanercept, Remicade, anti-ICAM, rapamycin, or tacrolimus. B cells are also potential therapeutic targets because they provide costimulatory signals that activate pathogenic T cells and secrete cytokines that promote autoimmune pathology. B cells also produce autoreactive antibodies implicated in several organ-specific and systemic autoimmune diseases including lupus erythematosus, Graves' disease, and Hashimoto's thyroiditis. On the other hand, recent studies have led to the discovery of several regulatory B-cell (Breg) populations that suppress immune responses and autoimmune diseases. In this review, we present a brief overview of Breg phenotypes and in particular, the newly discovered IL35-producing regulatory B cell (i35-Breg). We discuss the critical roles played by i35-Bregs in regulating autoimmune diseases and the potential use of adoptive Breg therapy in CNS autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/immunology , Central Nervous System Diseases/immunology , Immunotherapy, Adoptive , Interleukins/metabolism , Animals , Autoantibodies/immunology , Autoimmune Diseases/therapy , B-Lymphocytes, Regulatory/transplantation , Central Nervous System Diseases/therapy , Humans , Neurogenic Inflammation
17.
J Immunol ; 193(8): 3947-58, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25210119

ABSTRACT

Biologic drugs, including enzyme-replacement therapies, can elicit anti-drug Abs (ADA) that may interfere with drug efficacy and impact patient safety. In an effort to control ADA, we focused on identifying regimens of immune tolerance induction that may be readily available for clinical use. Data generated in both wild-type mice and a Pompe disease mouse model demonstrate that single-cycle, low-dose methotrexate can be as effective as three cycles of methotrexate in providing a long-lived reduction in alglucosidase alfa-specific ADA. In addition, we show that methotrexate induces Ag-specific tolerance as mice generate similar Ab responses to an irrelevant Ag regardless of prior methotrexate treatment. Methotrexate-induced immune tolerance does not seem to involve cell depletion, but rather a specific expansion of IL-10- and TGF-ß-secreting B cells that express Foxp3, suggesting an induction of regulatory B cells. The mechanism of immune tolerance induction appears to be IL-10 dependent, as methotrexate does not induce immune tolerance in IL-10 knockout mice. Splenic B cells from animals that have been tolerized to alglucosidase alfa with methotrexate can transfer tolerance to naive hosts. We hypothesize that methotrexate induction treatment concomitant with initial exposure to the biotherapeutic can induce Ag-specific immune tolerance in mice through a mechanism that appears to involve the induction of regulatory B cells.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Folic Acid Antagonists/pharmacology , Immune Tolerance/drug effects , Methotrexate/pharmacology , alpha-Glucosidases/immunology , Adoptive Transfer , Animals , Antigens, CD1d/immunology , Antimetabolites, Antineoplastic/pharmacology , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/transplantation , CD5 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism
18.
J Autoimmun ; 50: 107-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24491821

ABSTRACT

Regulatory B cells (Breg) have attracted increasing attention for their roles in maintaining peripheral tolerance. Interleukin 33 (IL-33) is a recently identified IL-1 family member, which leads a double-life with both pro- and anti-inflammatory properties. We report here that peritoneal injection of IL-33 exacerbated inflammatory bowel disease in IL-10-deficient (IL-10(-/-)) mice, whereas IL-33-treated IL-10-sufficient (wild type) mice were protected from the disease induction. A phenotypically unconventional subset(s) (CD19(+)CD25(+)CD1d(hi)IgM(hi)CD5(-)CD23(-)Tim-1(-)) of IL-10 producing Breg-like cells (Breg(IL-33)) was identified responsible for the protection. We demonstrated further that Breg(IL-33) isolated from these mice could suppress immune effector cell expansion and functions and, upon adoptive transfer, effectively blocked the development of spontaneous colitis in IL-10(-/-) mice. Our findings indicate an essential protective role, hence therapeutic potential, of Breg(IL-33) against mucosal inflammatory disorders in the gut.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Colitis/immunology , Gastric Mucosa/drug effects , Interleukin-10/immunology , Interleukins/pharmacology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/transplantation , Colitis/genetics , Colitis/pathology , Female , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gene Expression , Injections, Intraperitoneal , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-33 , Interleukins/immunology , Lymphocyte Activation , Mice , Mice, Knockout
19.
Biol Reprod ; 89(4): 90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23986569

ABSTRACT

During mammalian pregnancy, the immune system defies a double challenge: to tolerate the foreign growing fetus and to fight off infections that could affect both mother and fetus. Minimal disturbances to the fine equilibrium between immune activation and tolerance would compromise fetal survival. Here, we show that regulatory B10 cells are important for pregnancy tolerance in mice. The frequency of these cells increases during normal murine pregnancies, while mice presenting spontaneous abortion do not show elevated levels of regulatory B10 cells. When B10 cells are transferred to the abortion-prone mice, dendritic cells are kept in an immature state, and regulatory T cells increase, thus avoiding immunological rejection of the fetuses. In vitro, we could identify IL-10 secreted by B10 cells as the main mediator of these salutary effects. Our data add an important piece of information to the complex immune crosstalk during pregnancy. This study opens novel lines of work to better understand how to help women who have trouble in maintaining a pregnancy.


Subject(s)
Abortion, Spontaneous/prevention & control , B-Lymphocytes, Regulatory/transplantation , Disease Models, Animal , Immunologic Deficiency Syndromes/therapy , Interleukin-10/metabolism , Abortion, Spontaneous/etiology , Animals , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Crosses, Genetic , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Interleukin-10/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Knockout , Pregnancy , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
20.
Eur J Immunol ; 43(11): 2907-18, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23893352

ABSTRACT

Cerebral malaria (CM) is a neurological syndrome often occurring in severe malaria. Although CM is known as an immunopathology in brain tissue mediated by excessive proinflammatory cytokines, the immunoregulatory mechanism is poorly understood. Here, we investigated the role of IL-10-producing regulatory B (Breg) cells in modulating CM development in a murine model of Plasmodium berghei ANKA infection. We observed that blood-stage P. berghei induced expansion of IL-10-producing Breg cells in C57BL/6 mice. Adoptive transfer of IL-10(+) Breg cells to P. berghei infected mice significantly reduced the accumulation of NK and CD8(+) T cells and hemorrhage in brain tissue, and improved the survival of the mice compared with control groups, although parasitemia levels were not altered. Treatment of Breg-cell recipient mice with anti-IL-10 receptor mAb blocked the protective effect of Breg cells. Adoptive transfer of CD4(+) CD25(+) Treg cells failed to prevent CM in infected mice. Spleen cells from Breg-cell recipient mice produced increased levels of IL-10 in vitro. Cell co-culture showed that purified IL-10(+) B cells, but not IL-10(-) B cells, promoted IL-10 production by CD4(+) T cells. These results demonstrate that IL-10-producing Breg cells may represent an important mechanism for controlling the immunopathology and prevention of CM associated with P. berghei infection.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Interleukin-10/metabolism , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , B-Lymphocytes, Regulatory/transplantation , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Interleukin-10/genetics , Interleukin-10/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-10/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...