51
|
Valizadeh A, Sanaei R, Rezaei N, Azizi G, Fekrvand S, Aghamohammadi A, Yazdani R. Potential role of regulatory B cells in immunological diseases. Immunol Lett 2019; 215:48-59. [PMID: 31442542 DOI: 10.1016/j.imlet.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/04/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022]
Abstract
Regulatory B cells (Bregs) are immune-modulating cells that affect the immune system by producing cytokines or cellular interactions. These cells have immunomodulatory effects on the immune system by cytokine production. The abnormalities in Bregs could be involved in various disorders such as autoimmunity, chronic infectious disease, malignancies, allergies, and primary immunodeficiencies are immune-related scenarios. Ongoing investigation could disclose the biology and the exact phenotype of these cells and also the assigned mechanisms of action of each subset, as a result, potential therapeutic strategies for treating immune-related anomalies. In this review, we collect the findings of human and mouse Bregs and the therapeutic efforts to change the pathogenicity of these cells in diverse disease.
Collapse
Affiliation(s)
- Amir Valizadeh
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Roozbeh Sanaei
- Immunology Research Center (IRC), Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
52
|
IL-10-producing regulatory B cells and plasmocytes: Molecular mechanisms and disease relevance. Semin Immunol 2019; 44:101323. [DOI: 10.1016/j.smim.2019.101323] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022]
|
53
|
Wang X, Wang G, Wang Z, Liu B, Han N, Li J, Lu C, Liu X, Zhang Q, Yang Q, Wang G. PD-1-expressing B cells suppress CD4+ and CD8+ T cells via PD-1/PD-L1-dependent pathway. Mol Immunol 2019; 109:20-26. [DOI: 10.1016/j.molimm.2019.02.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/01/2019] [Accepted: 02/13/2019] [Indexed: 10/27/2022]
|
54
|
Zhu G, Liu Y, Zhang W, Huang Y, Li K. CD27 +TIM-1 + memory B cells promoted the development of Foxp3 + Tregs and were associated with better survival in acute respiratory distress syndrome. Immunol Res 2019; 66:281-287. [PMID: 29392553 DOI: 10.1007/s12026-017-8983-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a rapid onset life-threatening condition involving uncontrolled propagation of inflammatory responses. Here, we observed that ARDS patients that survived presented significantly higher frequencies of TIM-1+ B cells, especially the CD27+TIM-1+ B cells, than the ARDS patients who succumbed to the condition. We then found that using BCR/CD40 antigen-dependent stimulation or Staphylococcus aureus Cowan (SAC) antigen-independent stimulation, TIM-1+ B cells presented significantly higher IL-10 secretion and/or TGF-β1 secretion, with SAC stimulation being more effective. CD4+ T cells that incubated with TIM-1+ B cells presented significantly elevated IL-10 secretion, TGF-β1 secretion, and Foxp3 expression, than CD4+ T cells that incubated with TIM-1- B cells, suggesting TIM-1+ B cells promoted the in vitro development of Foxp3+ Treg cells. Interestingly, this TIM-1+ B cell-mediated promotion of Foxp3 expression was mostly dependent on TGF-β1 but not IL-10, since neutralization of TGF-β1, but not IL-10, resulted in the suppression of Foxp3 expression. We further showed that in TIM-1+ B cells, the CD27+ classical memory B cell subset demonstrated more regulatory potency than the CD27- subset. Together, our results suggested that the TIM-1+ B cells, especially those that expressed CD27, could promote Foxp3 expression. Their clinical efficacy in treating ARDS should be examined in in vivo experiments.
Collapse
Affiliation(s)
- Guangfa Zhu
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, 2 Anzhen Road, Beijing, 100029, People's Republic of China. .,Department of Infectious Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, 100029, People's Republic of China.
| | - Yan Liu
- Department of Infectious Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, 100029, People's Republic of China
| | - Wenmei Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, 2 Anzhen Road, Beijing, 100029, People's Republic of China
| | - Yan Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, 2 Anzhen Road, Beijing, 100029, People's Republic of China
| | - Keng Li
- Department of Infectious Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, 100029, People's Republic of China
| |
Collapse
|
55
|
Pang XX, Luo SD, Zhang T, Shi F, Wang CF, Chen XH, Wei YX, Qin L, Wei JX, Luo XQ, Wang JL. Association of interleukin-27 gene polymorphisms with susceptibility to HIV infection and disease progression. J Cell Mol Med 2019; 23:2410-2418. [PMID: 30632263 PMCID: PMC6433771 DOI: 10.1111/jcmm.14067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/26/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin‐27 (IL‐27) gene polymorphisms are linked to infectious disease susceptibility and IL‐27 plasma level is associated with HIV infection. Therefore, we aimed to investigate the association between IL‐27 polymorphisms and susceptibility to HIV infection and disease progression. A total of 300 patients with HIV infection (48 long‐term nonprogressors and 252 typical progressors) and 300 healthy controls were genotyped for three IL‐27 polymorphisms, rs17855750, rs181206, rs40837 which were performed by using multiple single nucleotide primer extension technique. Significant association was found between IL‐27 rs40837 polymorphisms with susceptibility to HIV infection (AG vs AA: adjusted OR = 1.60, 95% CI, 1.11‐2.30, P = 0.012; AG+GG vs AA: adjusted OR = 1.44, 95% CI, 1.02‐2.03, P = 0.038) and disease progression (LTNP: AG vs AA: adjusted OR = 2.33, 95% CI, 1.13‐4.80, P = 0.021; TP: AG vs AA: adjusted OR = 1.50, 95% CI, 1.04‐2.24, P = 0.030). Serum IL‐27 levels were significantly lower in cases compared to controls (P < 0.001). There were lower serum IL‐27 levels in TPs than in LTNPs (P < 0.001). We further found that LTNPs with rs40837 AG or GG genotype had lower serum IL‐27 levels than with AA genotype (P < 0.05). The CD4+T counts in cases were significantly lower than controls (P < 0.001). In contrast, individuals with rs40837 AG genotype had lower CD4+T counts than with AA genotype in cases (P < 0.05). In addition, CD4+T counts in TPs were significantly lower than LTNPs (P < 0.001). IL‐27 rs40837 polymorphism might influence the susceptibility to HIV infection and disease progression probably by regulating the level of serum IL‐27 or the quantity of CD4+T.
Collapse
Affiliation(s)
- Xiao-Xia Pang
- Youjiang Medical University for Nationalities, Baise, China.,Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Shun-Da Luo
- Department of Laboratory Medicine, The Fourth People's Hospital of Nanning, Nanning, China
| | - Ting Zhang
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Feng Shi
- Youjiang Medical University for Nationalities, Baise, China.,Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Chun-Fang Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xing-Hong Chen
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yu-Xia Wei
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Li Qin
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jing-Xi Wei
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xiao-Qiong Luo
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jun-Li Wang
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
56
|
Miller NJ, Church CD, Fling SP, Kulikauskas R, Ramchurren N, Shinohara MM, Kluger HM, Bhatia S, Lundgren L, Cheever MA, Topalian SL, Nghiem P. Merkel cell polyomavirus-specific immune responses in patients with Merkel cell carcinoma receiving anti-PD-1 therapy. J Immunother Cancer 2018; 6:131. [PMID: 30482247 PMCID: PMC6258401 DOI: 10.1186/s40425-018-0450-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/12/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is an aggressive skin cancer that frequently responds to anti-PD-1 therapy. MCC is associated with sun exposure and, in 80% of cases, Merkel cell polyomavirus (MCPyV). MCPyV-specific T and B cell responses provide a unique opportunity to study cancer-specific immunity throughout PD-1 blockade therapy. METHODS Immune responses were assessed in patients (n = 26) with advanced MCC receiving pembrolizumab. Peripheral blood mononuclear cells (PBMC) were collected at baseline and throughout treatment. MCPyV-oncoprotein antibodies were quantified and T cells were assessed for MCPyV-specificity via tetramer staining and/or cytokine secretion. Pre-treatment tumor biopsies were analyzed for T cell receptor clonality. RESULTS MCPyV oncoprotein antibodies were detectable in 15 of 17 (88%) of virus-positive MCC (VP-MCC) patients. Antibodies decreased in 10 of 11 (91%) patients with responding tumors. Virus-specific T cells decreased over time in patients who had a complete response, and increased in patients who had persistent disease. Tumors that were MCPyV(+) had a strikingly more clonal (less diverse) intratumoral TCR repertoire than virus-negative tumors (p = 0.0001). CONCLUSIONS Cancer-specific T and B cell responses generally track with disease burden during PD-1 blockade, in proportion to presence of antigen. Intratumoral TCR clonality was significantly greater in VP-MCC than VN-MCC tumors, suggesting expansion of a limited number of dominant clones in response to fewer immunogenic MCPyV antigens. In contrast, VN-MCC tumors had lower clonality, suggesting a diverse T cell response to numerous neoantigens. These findings reveal differences in tumor-specific immunity for VP-MCC and VN-MCC, both of which often respond to anti-PD-1 therapy.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biomarkers, Tumor
- Carcinoma, Merkel Cell/diagnosis
- Carcinoma, Merkel Cell/drug therapy
- Carcinoma, Merkel Cell/etiology
- Humans
- Immunomodulation/drug effects
- Lymphocyte Activation/immunology
- Merkel cell polyomavirus/immunology
- Molecular Targeted Therapy
- Polyomavirus Infections/complications
- Polyomavirus Infections/immunology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Cell Antigen Receptor Specificity/genetics
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
- Tumor Virus Infections/complications
- Tumor Virus Infections/immunology
Collapse
Affiliation(s)
- Natalie J. Miller
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| | - Candice D. Church
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| | - Steven P. Fling
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Rima Kulikauskas
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| | - Nirasha Ramchurren
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Michi M. Shinohara
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| | - Harriet M. Kluger
- Comprehensive Cancer Center, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT USA
| | - Shailender Bhatia
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| | - Lisa Lundgren
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Martin A. Cheever
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, WA USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA USA
| | - Suzanne L. Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, and Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD USA
| | - Paul Nghiem
- Department of Medicine, Divisions of Dermatology and Medical Oncology, University of Washington, 850 Republican Street, Seattle, WA 98109 USA
| |
Collapse
|
57
|
Yu Y, Zhu C, Zhou S, Chi S. Association Between C1q, TRAIL, and Tim-1 Gene Polymorphisms and Systemic Lupus Erythematosus. Genet Test Mol Biomarkers 2018; 22:546-553. [PMID: 30183357 DOI: 10.1089/gtmb.2018.0056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIM The present study was designed to examine the relationship between gene polymorphisms of C1q, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), T cell immunoglobulin mucin (Tim-1), and systemic lupus erythematosus (SLE). MATERIALS AND METHODS A total of 245 SLE patients were selected from February 2012 to August 2016, along with 245 healthy donors as the control group. Genomic DNA was extracted from peripheral blood samples from all subjects followed by mutational analyses. Gene polymorphisms of the C1q gene (rs292001, rs631090, rs294223 loci); the TRAIL gene (1525A/G, 1588A/G, 1595T/C locus); and the Tim-1 gene were detected by sequencing after polymerase chain reaction amplification. The concentration of anti-C1q antibody and the protein levels of sTRAIL/Tim-1 in serum of all subjects were measured by enzyme-linked immunosorbent assay. RESULTS As for the C1q gene, the frequency of the T allele at the rs631090 locus in the study group was lower than that in the controls, and the frequency of the C allele was higher in the study group than in the healthy donors. The frequency of the G allele at the 1525A/G locus of TRAIL gene in the study group was significantly higher than those in the control group. The frequency of the G allele at -1454G/A of Tim-1 was dramatically higher in the study group than in the control group. Anti-C1q antibody concentrations of subjects carrying CC and CT genotype at the rs631090 locus were statistically higher than TT genotype carriers. The sTRAIL protein level of the TRAIL 1525A/G GG genotype carriers was significantly higher than that of GA and AA genotype carriers, as well as CC genotype carriers at 1595T/C site compared with CT/TT genotype carriers. GG genotype carriers at -1454G/A had higher Tim-1 expression levels than GA/AA genotype carriers. CONCLUSION The C allele at the rs631090 locus of C1q, the G allele at 1525A/G site of TRAIL, and the G allele of Tim-1 at -1454G/A site are susceptibility variants associated with SLE.
Collapse
Affiliation(s)
- Yunxia Yu
- Department of Rheumatology, General Hospital of Ningxia Medical University , Yinchuan, Ningxia, China
| | - Caixia Zhu
- Department of Rheumatology, General Hospital of Ningxia Medical University , Yinchuan, Ningxia, China
| | - Shaolan Zhou
- Department of Rheumatology, General Hospital of Ningxia Medical University , Yinchuan, Ningxia, China
| | - Shuhong Chi
- Department of Rheumatology, General Hospital of Ningxia Medical University , Yinchuan, Ningxia, China
| |
Collapse
|
58
|
Yin W, Zhang PL, Macknis JK, Lin F, Bonventre JV. Kidney injury molecule-1 identifies antemortem injury in postmortem adult and fetal kidney. Am J Physiol Renal Physiol 2018; 315:F1637-F1643. [PMID: 30110569 DOI: 10.1152/ajprenal.00060.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is currently no technique to unambiguously diagnose antemortem kidney injury on postmortem examination since postmortem tissue damage and autolysis are common. We assessed the ability to detect kidney injury molecule-1 (KIM-1) expression in adult and fetal kidneys examined at autopsy. In adult kidneys ( n = 52 subjects), we found that the intensity of KIM-1 staining significantly correlated with the antemortem level of serum creatinine, and this was independent of the extent of tissue autolysis. In addition, kidneys from a total of 52 fetal/neonatal subjects, 30 stillborns and 22 liveborns, were assessed for KIM-1 staining. Given that serum creatinine is unreliable and often unavailable in fetuses and newborns, we assessed preterminal hypoxia in fetuses by the presence of squames in pulmonary alveoli and by required intubation. KIM-1 expression correlated with these clinical indexes of hypoxia. The expression of KIM-1 was seen in a majority of the fetal and neonatal autopsy kidneys (77%, 40/52) as early as 16 wk of gestation, even in the presence of autolysis. Thus KIM-1 is a specific and stable marker of antemortem tubular injury in kidneys of adults and fetuses despite postmortem autolysis.
Collapse
Affiliation(s)
- Wenqing Yin
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Ping L Zhang
- Department of Anatomic Pathology, William Beaumont Hospital, Royal Oak, Minnesota
| | - Jacqueline K Macknis
- Department of Anatomic Pathology, William Beaumont Hospital, Royal Oak, Minnesota
| | - Fan Lin
- Division of Laboratory Medicine, Geisinger Medical Center , Danville, Pennsylvania
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts.,Harvard Stem Cell Institute , Cambridge, Massachusetts.,Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology , Cambridge, Massachusetts
| |
Collapse
|
59
|
Chang-Panesso M, Kadyrov FF, Machado FG, Kumar A, Humphreys BD. Meis1 is specifically upregulated in kidney myofibroblasts during aging and injury but is not required for kidney homeostasis or fibrotic response. Am J Physiol Renal Physiol 2018; 315:F275-F290. [PMID: 29592525 PMCID: PMC6139520 DOI: 10.1152/ajprenal.00030.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/21/2018] [Accepted: 03/26/2018] [Indexed: 11/22/2022] Open
Abstract
The homeobox transcription factor Meis1 is required for mammalian development, and its overexpression plays a role in tumorigenesis, especially leukemia. Meis1 is known to be expressed in kidney stroma, but its function in kidney is undefined. We hypothesized that Meis1 may regulate stromal cell proliferation in kidney development and disease and tested the hypothesis using cell lineage tracing and cell-specific Meis1 deletion in development, aging, and fibrotic disease. We observed strong expression of Meis1 in platelet-derived growth factor receptor-β-positive pericytes and perivascular fibroblasts, both in adult mouse kidney and to a lesser degree in human kidney. Either bilateral ischemia-reperfusion injury or aging itself led to strong upregulation of Meis1 protein and mRNA in kidney myofibroblasts, and genetic lineage analysis confirmed that Meis1-positive cells proliferate as they differentiate into myofibroblasts after injury. Conditional deletion of Meis1 in all kidney stroma with two separate tamoxifen-inducible Cre recombinase drivers had no phenotype with the exception of consistent induction of the tubular injury marker kidney injury molecule-1 (Kim-1) only in Meis1 mutants. Further examination of Kim-1 expression revealed linkage disequilibrium of Kim-1 and Meis1, such that Meis1 mutants carried the longer BALB/c Kim-1 allele. Unexpectedly, we report that this Kim-1 allele is expressed at baseline in wild-type BALB/c mice, without any associated abnormalities, including long-term fibrosis, as predicted from the literature. We conclude that Meis1 is specifically expressed in stroma and myofibroblasts of mouse and human kidney, that it is not required for kidney development, disease, or aging, and that BALB/c mice unexpectedly express Kim-1 protein at baseline without other kidney abnormality.
Collapse
Affiliation(s)
- Monica Chang-Panesso
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine , St. Louis, Missouri
| | - Farid F Kadyrov
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine , St. Louis, Missouri
| | - Flavia G Machado
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine , St. Louis, Missouri
| | - Ashish Kumar
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine , St. Louis, Missouri
| |
Collapse
|
60
|
Czaja AJ. Under-Evaluated or Unassessed Pathogenic Pathways in Autoimmune Hepatitis and Implications for Future Management. Dig Dis Sci 2018; 63:1706-1725. [PMID: 29671161 DOI: 10.1007/s10620-018-5072-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autoimmune hepatitis is a consequence of perturbations in homeostatic mechanisms that maintain self-tolerance but are incompletely understood. The goals of this review are to describe key pathogenic pathways that have been under-evaluated or unassessed in autoimmune hepatitis, describe insights that may shape future therapies, and encourage investigational efforts. The T cell immunoglobulin mucin proteins constitute a family that modulates immune tolerance by limiting the survival of immune effector cells, clearing apoptotic bodies, and expanding the population of granulocytic myeloid-derived suppressor cells. Galectins influence immune cell migration, activation, proliferation, and survival, and T cell exhaustion can be induced and exploited as a possible management strategy. The programmed cell death-1 protein and its ligands comprise an antigen-independent inhibitory axis that can limit the performance of activated T cells by altering their metabolism, and epigenetic changes can silence pro-inflammatory genes or de-repress anti-inflammatory genes that affect disease severity. Changes in the intestinal microbiota and permeability of the intestinal mucosal barrier can be causative or consequential events that affect the occurrence and phenotype of immune-mediated disease, and they may help explain the female propensity for autoimmune hepatitis. Perturbations within these homeostatic mechanisms have been implicated in experimental models and limited clinical experiences, and they have been favorably manipulated by monoclonal antibodies, recombinant molecules, pharmacological agents or dietary supplements. In conclusion, pathogenic mechanisms that have been implicated in other systemic immune-mediated and liver diseases but under-evaluated or unassessed in autoimmune hepatitis warrant consideration and rigorous evaluation.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
61
|
Le Gallou S, Zhou Z, Thai LH, Fritzen R, de Los Aires AV, Mégret J, Yu P, Kitamura D, Bille E, Tros F, Nassif X, Charbit A, Weller S, Weill JC, Reynaud CA. A splenic IgM memory subset with antibacterial specificities is sustained from persistent mucosal responses. J Exp Med 2018; 215:2035-2053. [PMID: 29959173 PMCID: PMC6080908 DOI: 10.1084/jem.20180977] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
Le Gallou et al. use an AID fate-mapping model to identify an IgM memory population in the spleen of unimmunized mice, originating from persistent gut immune responses and endowed with cross-reactivity against bacteria. To what extent immune responses against the gut flora are compartmentalized within mucosal tissues in homeostatic conditions remains a much-debated issue. We describe here, based on an inducible AID fate-mapping mouse model, that systemic memory B cell subsets, including mainly IgM+ B cells in spleen, together with IgA+ plasma cells in spleen and bone marrow, are generated in mice in the absence of deliberate immunization. While the IgA component appears dependent on the gut flora, IgM memory B cells are still generated in germ-free mice, albeit to a reduced extent. Clonal relationships and renewal kinetics after anti-CD20 treatment reveal that this long-lasting splenic population is mainly sustained by output of B cell clones persisting in mucosal germinal centers. IgM-secreting hybridomas established from splenic IgM memory B cells showed reactivity against various bacterial isolates and endogenous retroviruses. Ongoing activation of B cells in gut-associated lymphoid tissues thus generates a diversified systemic compartment showing long-lasting clonal persistence and protective capacity against systemic bacterial infections.
Collapse
Affiliation(s)
- Simon Le Gallou
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Zhicheng Zhou
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lan-Huong Thai
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Remi Fritzen
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alba Verge de Los Aires
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérôme Mégret
- Flow Cytometry Core Facility, Structure Fédérative de Recherche Necker, Institut National de la Santé et de la Recherche Médicale US24-Centre National de la Recherche Scientifique UMS 3633, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Philipp Yu
- Institute of Immunology, Philipps-Universität Marburg, Marburg, Germany
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Emmanuelle Bille
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fabiola Tros
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xavier Nassif
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Charbit
- Team "Pathogeny of Systemic Infections", Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sandra Weller
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Claude Weill
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claude-Agnès Reynaud
- Team "Development of the Immune System," Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151-Centre National de la Recherche Scientifique UMR 8253, Faculté de Médecine Paris Descartes, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
62
|
Said SS, Barut GT, Mansur N, Korkmaz A, Sayi-Yazgan A. Bacterially activated B-cells drive T cell differentiation towards Tr1 through PD-1/PD-L1 expression. Mol Immunol 2018; 96:48-60. [DOI: 10.1016/j.molimm.2018.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/20/2018] [Accepted: 02/10/2018] [Indexed: 01/08/2023]
|
63
|
Decreased Breg/Th17 Ratio Improved the Prognosis of Patients with Ulcerative Colitis. Can J Gastroenterol Hepatol 2018; 2018:5760849. [PMID: 29765931 PMCID: PMC5885404 DOI: 10.1155/2018/5760849] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/26/2017] [Accepted: 12/18/2017] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the effects of regulatory B (Breg) cells and T helper 17 (Th17) cells on pathogenesis of ulcerative colitis, explore the clinical significance of Breg/Th17 ratio on the prognosis of ulcerative colitis, and provide the theoretical basis for the targeted therapy, diagnosis, and prognosis of the disease. METHODS Peripheral blood and colonic mucosa were collected from patients with ulcerative colitis. Hematoxylin-eosin staining was used to observe the pathological changes of colonic mucosa. Flow cytometry was utilized to analyze the percentages of Breg cells and Th17 cells. Real-time fluorescent quantitative polymerase chain reaction and immunohistochemistry were applied to determine the expression of Breg cells-related cytokines IL-10 and Th17 cell transcription factor RORγT. Enzyme-linked immunosorbent assay was employed to detect serum IL-10 and IL-17 levels. RESULTS The colonic mucosa of ulcerative colitis patients presented massive inflammatory cell infiltration and hemorrhagic necrosis. The number of Breg cells and Th17 cells, the gene expressions of IL-10 and RORγT, and serum levels of IL-10 and IL-17 all increased in peripheral blood. Compared with nonremission group, the remission group showed that the percentage of Breg cells reduced, the percentage of Th17 cells increased, and thus the B10/Th17 ratio was significantly decreased in peripheral blood. In addition, serum IL-10 levels diminished, IL-17 levels increased, and thus IL-10/IL-17 ratio was remarkably reduced in remission group. B10/Th17 ratio and IL-10/IL-17 ratio were positively correlated with the severity of disease. CONCLUSIONS Breg and Th17 cells participate in the occurrence and development of ulcerative colitis. B10/Th17 ratio and IL-10/IL-17 ratio can be used as prognostic markers for ulcerative colitis. This provides a theoretical basis for design of targeted treatment and prognosis assessment of the disease.
Collapse
|
64
|
Alhabbab R, Blair P, Smyth LA, Ratnasothy K, Peng Q, Moreau A, Lechler R, Elgueta R, Lombardi G. Galectin-1 is required for the regulatory function of B cells. Sci Rep 2018; 8:2725. [PMID: 29426942 PMCID: PMC5807431 DOI: 10.1038/s41598-018-19965-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022] Open
Abstract
Galectin-1 (Gal-1) is required for the development of B cells in the bone marrow (BM), however very little is known about the contribution of Gal-1 to the development of B cell regulatory function. Here, we report an important role for Gal-1 in the induction of B cells regulatory function. Mice deficient of Gal-1 (Gal-1−/−) showed significant loss of Transitional-2 (T2) B cells, previously reported to include IL-10+ regulatory B cells. Gal-1−/− B cells stimulated in vitro via CD40 molecules have impaired IL-10 and Tim-1 expression, the latter reported to be required for IL-10 production in regulatory B cells, and increased TNF-α expression compared to wild type (WT) B cells. Unlike their WT counterparts, T2 and T1 Gal-1−/− B cells did not suppress TNF-α expression by CD4+ T cells activated in vitro with allogenic DCs (allo-DCs), nor were they suppressive in vivo, being unable to delay MHC-class I mismatched skin allograft rejection following adoptive transfer. Moreover, T cells stimulated with allo-DCs show an increase in their survival when co-cultured with Gal-1−/− T2 and MZ B cells compared to WT T2 and MZ B cells. Collectively, these data suggest that Gal-1 contributes to the induction of B cells regulatory function.
Collapse
Affiliation(s)
- R Alhabbab
- Infectious Disease Unit & Division of Applied Medical Sciences, King Fahad Centre for medical research, King Abdulaziz University, Jeddah, Saudi Arabia. .,Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.
| | - P Blair
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.,Centre for Rheumatology, Division of Medicine, University College London, London, WC1E 6JF, UK
| | - L A Smyth
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.,School of Health, Sports and Biosciences, University of East London, Stratford, E15 4LZ, UK
| | - K Ratnasothy
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK
| | - Q Peng
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK
| | - A Moreau
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.,Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, CHU, Nantes, France
| | - R Lechler
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK
| | - R Elgueta
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.
| | - G Lombardi
- Division of Transplantation Immunology & Mucosal Biology, King's College London, King's Health Partners, Guy's Hospital, London, SE1 9RT, UK.
| |
Collapse
|
65
|
Chu Z, Zou W, Xu Y, Sun Q, Zhao Y. The regulatory roles of B cell subsets in transplantation. Expert Rev Clin Immunol 2018; 14:115-125. [PMID: 29338551 DOI: 10.1080/1744666x.2018.1426461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Zhulang Chu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Department of Pathology, Beijing University of Chinese Medicine, Beijing, China
| | - Weilong Zou
- Surgery of Transplant and Hepatopancrobiliary, The General Hospital of Chinese People’s Armed Police Forces, Beijing, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qiquan Sun
- Department of Renal Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
66
|
Miles K, Simpson J, Brown S, Cowan G, Gray D, Gray M. Immune Tolerance to Apoptotic Self Is Mediated Primarily by Regulatory B1a Cells. Front Immunol 2018; 8:1952. [PMID: 29403471 PMCID: PMC5780629 DOI: 10.3389/fimmu.2017.01952] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/18/2017] [Indexed: 01/24/2023] Open
Abstract
The chronic autoimmune inflammatory diseases, systemic lupus erythematosus and Sjogren's syndrome, develop when tolerance to apoptotic cells (ACs) is lost. We have previously reported that this tolerance is maintained by innate-like, IL-10 secreting regulatory B cells. Two questions remained. First, do these regulatory B cells belong predominantly to a single subset of steady-state B cells and second, what is their specificity? We report here that innate-like B cells with markers characteristic for B1a cells (CD43+veCD19hiCD5+veIgMhiIgDlo) constitute 80% of splenic and 96% of peritoneal B cells that respond to ACs by secreting IL-10. AC responsive B1a cells secrete self-reactive natural antibodies (NAbs) and IL-10, which is augmented by toll-like receptor (TLR) 7 or TLR9 stimulation. In so doing, they both accelerate the clearance of dying cells by macrophages and inhibit their potential to mount proinflammatory immune responses. While B1a cells make prolonged contact with ACs, they do not require TIM1 or complement to mediate their regulatory function. In an animal model of neural inflammation (experimental autoimmune encephalomyelitis), just 105 activated B1a B cells was sufficient to restrain inflammation. Activated B1a B cells also induced antigen-specific T cells to secrete IL-10. Hence, regulatory B1a cells specifically recognize and augment tolerance to apoptotic self via IL-10 and NAbs; but once activated, can also prevent autoimmune mediated inflammation.
Collapse
Affiliation(s)
- Katherine Miles
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanne Simpson
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sheila Brown
- School of Biological Sciences, Institute of Immunology and Infection Research, Ashworth Laboratories, The University of Edinburgh, Edinburgh, United Kingdom
| | - Graeme Cowan
- School of Biological Sciences, Institute of Immunology and Infection Research, Ashworth Laboratories, The University of Edinburgh, Edinburgh, United Kingdom
| | - David Gray
- School of Biological Sciences, Institute of Immunology and Infection Research, Ashworth Laboratories, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mohini Gray
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
67
|
CD19+ Tim-1+ B cells are decreased and negatively correlated with disease severity in Myasthenia Gravis patients. Immunol Res 2017; 64:1216-1224. [PMID: 27677768 DOI: 10.1007/s12026-016-8872-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
T cell immunoglobulin mucin domain-1(Tim-1) was recently identified to be critical and essential for optimal regulatory B cells function in maintaining immune tolerance. We aimed to measure the expression levels of Tim-1 on B cells from patients with Myasthenia Gravis (MG) and to investigate whether the expression of Tim-1 is associated with pathogenesis of MG. A total of 34 patients with MG (18 generalized MG (GMG) and 16 ocular MG (OMG) and 24 healthy donors were recruited in this study. The quantitative myasthenia gravis score (QMGS) was used to evaluate the clinical severity. Real-time PCR and flow cytometry were used to measure the levels of Tim-1 expressed on peripheral B cells. Peripheral CD138+ plasma cells were assayed by flow cytometry. Serum Th17-related cytokines (IL-6, IL-1β and IL-17) and anti-AChR antibody (Ab) titers were tested by enzyme-linked immunosorbent assay (ELISA). Our data demonstrated that the mRNA and protein expression levels of B cell Tim-1 in both the GMG and OMG groups were significantly lower than those in healthy controls, with lower expression in GMG than in OMG. Tim-1 expression on B cells from OMG/GMG was negatively correlated with clinical severity, plasma cells frequency, serum Th17-related cytokines and anti-AChR Ab levels. Our results indicated that aberrant expression of Tim-1 exists on B cells and may contribute to the Th17 polarization and antibody-secreting plasma cells differentiation in MG patients.
Collapse
|
68
|
Wang H, Xu W, Shao Q, Ding Q. miR-21 silencing ameliorates experimental autoimmune encephalomyelitis by promoting the differentiation of IL-10-producing B cells. Oncotarget 2017; 8:94069-94079. [PMID: 29212210 PMCID: PMC5706856 DOI: 10.18632/oncotarget.21578] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/18/2017] [Indexed: 01/04/2023] Open
Abstract
IL-10-producing regulatory B (IL-10+ Breg) cells promote tolerance in autoimmune diseases and transplantation. However, it remains unclear whether microRNAs are involved in the development of IL-10+ Breg cells. Here, we found that microRNA-21 (miR-21) acts as an upstream regulator of IL-10 by targeting the 3' untranslated region of IL-10 mRNA. We also demonstrated that IL-10+ Breg cells exhibit lower miR-21 expression than non-Breg cells and that miR-21 acts as a potent negative regulator of the differentiation of IL-10+ Breg cells. Accordingly, specific inhibition of miR-21 using antisense oligonucleotides markedly promoted B cell IL-10 expression. Thus, IL-10 is a direct target of miR-21. Moreover, silencing of miR-21 significantly alleviated the severity of experimental autoimmune encephalomyelitis (EAE), and this change was associated with an increase in the number of IL-10+ Breg cells. Finally, we demonstrated that miR-21-silenced B cells exert their suppressive activity through effector T cells in an IL-10-dependent manner. Thus, we characterized a B cell-intrinsic microRNA pathway that inhibits the differentiation of IL-10+ Breg cells and promotes autoimmunity. miR-21 silencing therefore represents a new therapeutic strategy for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Hui Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Qixiang Shao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Qing Ding
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
69
|
Hu Y, Yu P, Yu X, Hu X, Kawai T, Han X. IL-21/anti-Tim1/CD40 ligand promotes B10 activity in vitro and alleviates bone loss in experimental periodontitis in vivo. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2149-2157. [PMID: 28583714 DOI: 10.1016/j.bbadis.2017.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/12/2017] [Accepted: 06/01/2017] [Indexed: 12/20/2022]
Abstract
IL-10-expressing regulatory B cells (B10) play an essential role in immune system balance by suppressing excessive inflammatory responses. In this study, we investigated induction of B 10 cell's IL-10 competency in vitro and its effect on ligature-induced experimental periodontitis in vivo. Spleen B cells were isolated from C57BL/6J mice and cultured for 48h under the following conditions: control, CD40L, IL-21, anti-Tim1, CD40L+IL-21, CD40L+anti-Tim1, CD40L+IL-21+anti-Tim1. Silk ligatures were tied around both maxillary second molars of C57BL/6J mice for two weeks. Optimized combination of CD40L, IL-21 and anti-Tim1 and vehicle were injected into contralateral side of palatal gingiva on days 3, 6 and 9. The palatal gingival tissues and maxillary bone were collected on day 14 to determine expressions of IL-10 and periodontal bone resorption respectively. Our results demonstrated that IL-10 expressions of cultured spleen B cells were significantly increased in the presence of CD40L, IL-21 and anti-Tim1 combination when compared with control groups. Gingival IL-10 mRNA and protein expressions were significantly increased after injection of CD40L, IL-21 and anti-Tim1 combination, when compared to the control side. The gingival RANKL expression and periodontal bone loss were significantly decreased on the combination treatment side, as compared to the control side. These results suggest that combination of IL-21, anti-Tim1 and CD40L treatment induced B10 cell's IL-10 competency in vitro and inhibited periodontal bone loss in ligature-induced experimental periodontitis.
Collapse
Affiliation(s)
- Yang Hu
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States; Harvard University, School of Dental Medicine, Cambridge, MA, United States
| | - Pei Yu
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States; State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinbo Yu
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States; Department of Periodontology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xingxue Hu
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States
| | - Toshihisa Kawai
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States; Harvard University, School of Dental Medicine, Cambridge, MA, United States
| | - Xiaozhe Han
- Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States; Harvard University, School of Dental Medicine, Cambridge, MA, United States.
| |
Collapse
|
70
|
Foks AC, Kuiper J. Immune checkpoint proteins: exploring their therapeutic potential to regulate atherosclerosis. Br J Pharmacol 2017; 174:3940-3955. [PMID: 28369782 DOI: 10.1111/bph.13802] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/17/2017] [Accepted: 03/15/2017] [Indexed: 12/23/2022] Open
Abstract
The immune system provides a large variety of immune checkpoint proteins, which involve both costimulatory and inhibitory proteins. Costimulatory proteins can promote cell survival, cell cycle progression and differentiation to effector and memory cells, whereas inhibitory proteins terminate these processes to halt ongoing inflammation. Immune checkpoint proteins play a pivotal role in atherosclerosis by regulating the activation and proliferation of various immune and non-immune cells, such as T-cells, macrophages and platelets. Upon activation within the atherosclerotic lesions or in secondary lymphoid organs, these cells produce large amounts of pro-atherogenic cytokines that contribute to the growth and destabilization of lesions, which can result in rupture of the lesion causing acute coronary syndromes, such as a myocardial infarction. Given the presence and regulatory capacity of immune checkpoint proteins in the circulation and atherosclerotic lesions of cardiovascular patients, modulation of these proteins by, for example, the use of monoclonal antibodies, offers unique opportunities to regulate pro-inflammatory immune responses in atherosclerosis. In this review, we highlight the latest advances on the role of immune checkpoint proteins, such as OX40-OX40L, CTLA-4 and TIM proteins, in atherosclerosis and discuss their therapeutic potential as promising immunotherapies to treat or prevent cardiovascular disease. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- A C Foks
- Division of Biopharmaceutics, LACDR, Leiden University, Leiden, The Netherlands
| | - J Kuiper
- Division of Biopharmaceutics, LACDR, Leiden University, Leiden, The Netherlands
| |
Collapse
|
71
|
Soares RR, Antinarelli LMR, Abramo C, Macedo GC, Coimbra ES, Scopel KKG. What do we know about the role of regulatory B cells (Breg) during the course of infection of two major parasitic diseases, malaria and leishmaniasis? Pathog Glob Health 2017; 111:107-115. [PMID: 28353409 PMCID: PMC5445636 DOI: 10.1080/20477724.2017.1308902] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Parasitic diseases, such as malaria and leishmaniasis, are relevant public health problems worldwide. For both diseases, the alarming number of clinical cases and deaths reported annually has justified the incentives directed to better understanding of host's factors associated with susceptibility to infection or protection. In this context, over recent years, some studies have given special attention to B lymphocytes with a regulator phenotype, known as Breg cells. Essentially important in the maintenance of immunological tolerance, especially in autoimmune disease models such as rheumatoid arthritis and experimentally induced autoimmune encephalomyelitis, the function of these lymphocytes has so far been poorly explored during the course of diseases caused by parasites. As the activation of Breg cells has been proposed as a possible therapeutic or vaccine strategy against several diseases, here we reviewed studies focused on understanding the relation of parasite and Breg cells in malaria and leishmaniasis, and the possible implications of these strategies in the course of both infections.
Collapse
|
72
|
Schistosome-induced pulmonary B cells inhibit allergic airway inflammation and display a reduced Th2-driving function. Int J Parasitol 2017; 47:545-554. [PMID: 28385494 DOI: 10.1016/j.ijpara.2017.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/31/2022]
Abstract
Chronic schistosome infections protect against allergic airway inflammation (AAI) via the induction of IL-10-producing splenic regulatory B (Breg) cells. Previous experiments have demonstrated that schistosome-induced pulmonary B cells can also reduce AAI, but act independently of IL-10. We have now further characterized the phenotype and inhibitory activity of these protective pulmonary B cells. We excluded a role for regulatory T (Treg) cell induction as putative AAI-protective mechanisms. Schistosome-induced B cells showed increased CD86 expression and reduced cytokine expression in response to Toll-like receptor (TLR) ligands compared with control B cells. To investigate the consequences for T cell activation we cultured ovalbumin (OVA)-pulsed, schistosome-induced B cells with OVA-specific transgenic T cells and observed less Th2 cytokine expression and T cell proliferation compared with control conditions. This suppressive effect was preserved even under optimal T cell stimulation by anti-CD3/28. Blocking of the inhibitory cytokines IL-10 or TGF-β only marginally restored Th2 cytokine induction. These data suggest that schistosome-induced pulmonary B cells are impaired in their capacity to produce cytokines to TLR ligands and to induce Th2 cytokine responses independent of their antigen-presenting function. These findings underline the presence of distinct B cell subsets with different stimulatory or inhibitory properties even if induced by the same type of helminth.
Collapse
|
73
|
Wortel CM, Heidt S. Regulatory B cells: Phenotype, function and role in transplantation. Transpl Immunol 2017; 41:1-9. [PMID: 28257995 DOI: 10.1016/j.trim.2017.02.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 02/27/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
While B cells are traditionally known for their roles in antibody production, antigen presentation and cytokine production, recent studies have highlighted the existence of B cells with regulatory properties, which have been termed Bregs, analogous to regulatory T cells (Tregs). Bregs have been found to play a role in autoimmune disease, malignancies, infections, and may also be involved in solid organ transplantation. Their main mechanism of action is by promoting the development of Tregs while suppressing effector CD4+ and CD8+ T cells, primarily by IL-10 secretion. In the field of transplantation evidence for an active role of Bregs is scarce. While the presence of Bregs has been associated with improved graft survival and operational tolerance in kidney transplant recipients, these findings are not without controversy. Since the majority of fundamental research on Bregs has been performed in the fields in autoimmunity and infectious diseases, we will first focus on what these fields taught us on basic Breg biology, after which the relevance for the transplant setting is discussed.
Collapse
Affiliation(s)
- C M Wortel
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, The Netherlands
| | - S Heidt
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, The Netherlands.
| |
Collapse
|
74
|
Aravena O, Ferrier A, Menon M, Mauri C, Aguillón JC, Soto L, Catalán D. TIM-1 defines a human regulatory B cell population that is altered in frequency and function in systemic sclerosis patients. Arthritis Res Ther 2017; 19:8. [PMID: 28103916 PMCID: PMC5248463 DOI: 10.1186/s13075-016-1213-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/28/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a systemic autoimmune disease characterized by excessive production of extracellular matrix by fibroblasts on skin and internal organs. Although Th2 cells have been involved in fibroblast stimulation, hyperactivated B cells may also play an important role. Regulatory B cells (Bregs) are cells capable of inhibiting inflammatory responses and controlling autoimmune diseases. Although many Breg populations have in common the ability to produce high amounts of IL-10, a unique surface marker defining most human Bregs is lacking. It has been described in mice that T cell Ig and mucin domain protein 1 (TIM-1) is an inclusive marker for Bregs, and that TIM-1+ B cells are able to prevent the development of autoimmunity. The aim of this work was to evaluate TIM-1 as a marker for human IL-10+ Bregs, and to determine whether TIM-1+ B cells are defective in SSc patients. METHODS SSc patients (n = 39) and 53 healthy subjects were recruited. TIM-1 and IL-10 expression was assessed in resting or activated peripheral blood CD19+ B cells by flow cytometry. The regulatory function of TIM-1+ or activated B cells from SSc patients and healthy subjects was assessed in autologous and allogenic co-cultures with CD4+ T cells, where T cell proliferation and IFN-γ, IL-17, TNF-α and IL-4 production by T cells was measured by flow cytometry. RESULTS TIM-1 and IL-10 were preferentially expressed in transitional B cells, but were upregulated in naïve and memory B cells upon stimulation. The frequency of transitional TIM-1+ IL-10+ B cells was significantly decreased in SSc patients compared to healthy controls. In addition, activated B cells from SSc patients induced stronger allogenic Th1 and Th2 responses than activated B cells from healthy controls. Finally, TIM-1+ B cells, including transitional and non-transitional cells, exhibited a higher CD4+ T cell suppressive ability than TIM-1- B cells in healthy controls, but not in SSc patients. CONCLUSIONS TIM-1 is a unique marker for the identification of a human IL-10+ Breg subpopulation which is partially superimposed with transitional B cells. Alterations in TIM-1+ B cells could contribute to the development of autoimmune diseases such as SSc.
Collapse
Affiliation(s)
- Octavio Aravena
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, and Millennium Institute in Immunology and Immunotherapy, Santiago, Chile
| | - Ashley Ferrier
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, and Millennium Institute in Immunology and Immunotherapy, Santiago, Chile
| | - Madhvi Menon
- Centre for Rheumatology Research, Department of Medicine, University College London, London, UK
| | - Claudia Mauri
- Centre for Rheumatology Research, Department of Medicine, University College London, London, UK
| | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, and Millennium Institute in Immunology and Immunotherapy, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, and Millennium Institute in Immunology and Immunotherapy, Santiago, Chile.,Departamento de Medicina, Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, and Millennium Institute in Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
75
|
Biragyn A, Aliseychik M, Rogaev E. Potential importance of B cells in aging and aging-associated neurodegenerative diseases. Semin Immunopathol 2017; 39:283-294. [PMID: 28083646 DOI: 10.1007/s00281-016-0615-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/20/2022]
Abstract
Our understanding of B cells as merely antibody producers is slowly changing. Alone or in concert with antibody, they control outcomes of seemingly different diseases such as cancer, rheumatoid arthritis, diabetes, and multiple sclerosis. While their role in activation of effector immune cells is beneficial in cancer but bad in autoimmune diseases, their immunosuppressive and regulatory subsets (Bregs) inhibit autoimmune and anticancer responses. These pathogenic and suppressive functions are not static and appear to be regulated by the nature and strength of inflammation. Although aging increases inflammation and changes the composition and function of B cells, surprisingly, little is known whether the change affects aging-associated neurodegenerative disease, such as Alzheimer's disease (AD). Here, by analyzing B cells in cancer and autoimmune and neuroinflammatory diseases, we elucidate their potential importance in AD and other aging-associated neuroinflammatory diseases.
Collapse
Affiliation(s)
- Arya Biragyn
- Immunoregulation section, National Institute on Aging, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Maria Aliseychik
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Evgeny Rogaev
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Genomics and Human Genetics, Russian Academy of Sciences, Institute of General Genetics, Moscow, Russia.,Center for Brain Neurobiology and Neurogenetics, Siberian Branch of the Russian Academy of Sciences, Institute of Cytology and Genetics, Novosibirsk, Russia
| |
Collapse
|
76
|
|
77
|
Kishimoto W, Nishikori M, Arima H, Miyoshi H, Sasaki Y, Kitawaki T, Shirakawa K, Kato T, Imaizumi Y, Ishikawa T, Ohno H, Haga H, Ohshima K, Takaori-Kondo A. Expression of Tim-1 in primary CNS lymphoma. Cancer Med 2016; 5:3235-3245. [PMID: 27709813 PMCID: PMC5119979 DOI: 10.1002/cam4.930] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 11/16/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a distinct subtype of extranodal lymphoma with aggressive clinical course and poor outcome. As increased IL‐10/IL‐6 ratio is recognized in the cerebrospinal fluid (CSF) of PCNSL patients, we hypothesized that PCNSL might originate from a population of B cells with high IL‐10‐producing capacity, an equivalent of “regulatory B cells” in mice. We intended in this study to clarify whether Tim‐1, a molecule known as a marker for regulatory B cells in mice, is expressed in PCNSL. By immunohistochemical analysis, Tim‐1 was shown to be positive in as high as 54.2% of PCNSL (26 of 58 samples), while it was positive in 19.1% of systemic diffuse large B‐cell lymphoma (DLBCL) samples (17 of 89 samples; P < 0.001). Tim‐1 expression positively correlated with IL‐10 expression in PCNSL (Cramer's V = 0.55, P < 0.001), and forced expression of Tim‐1 in a PCNSL cell line resulted in increased IL‐10 secretion, suggesting that Tim‐1 is functionally linked with IL‐10 production in PCNSL. Moreover, soluble Tim‐1 was detectable in the CSF of PCNSL patients, and was suggested to parallel disease activity. In summary, PCNSL is characterized by frequent Tim‐1 expression, and its soluble form in CSF may become a useful biomarker for PCNSL.
Collapse
Affiliation(s)
- Wataru Kishimoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Momoko Nishikori
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Arima
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Yuya Sasaki
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeharu Kato
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Takayuki Ishikawa
- Department of Hematology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Hitoshi Ohno
- Department of Hematology, Tenri Hospital, Mishima-cho, Tenri, Nara, Japan
| | - Hironori Haga
- Department of Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
78
|
MESH Headings
- Antigen Presentation
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/pathology
- Colitis/genetics
- Colitis/immunology
- Colitis/pathology
- Cytokines/genetics
- Cytokines/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation/immunology
- Humans
- Hypersensitivity, Delayed/genetics
- Hypersensitivity, Delayed/immunology
- Hypersensitivity, Delayed/pathology
- Immune Tolerance
- Immunity, Humoral
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Cytokine/genetics
- Receptors, Cytokine/immunology
- Signal Transduction
Collapse
|
79
|
New insights into B cell biology in systemic lupus erythematosus and Sjögren's syndrome. Curr Opin Rheumatol 2016; 27:461-7. [PMID: 26164595 DOI: 10.1097/bor.0000000000000201] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Our understanding of the physiological and pathogenic functions of B cells in systemic lupus erythematosus (SLE) and Primary Sjögren's syndrome (pSS) continues to expand. In this review, we discuss novel insights published in the last 18 months into the roles of B cells in systemic autoimmunity. RECENT FINDINGS Data have continued to expand regarding the diverse mechanisms by which innate immune signals including Toll-like receptors (TLRs) regulate the B cell compartment. Localized B cells and long-lived plasma cells have been identified as playing an important role in target tissue including the development of ectopic lymphoid structures in kidney and salivary gland. In addition to pathogenic roles for B cells, there is mounting evidence for regulatory B cell subsets that play a protective role and new insights into the signals that regulate their development. SUMMARY The past few years have provided insights into the multiple paths by which innate immune signals can lead to B cell activation in SLE and pSS and the increasingly diverse ways in which B cells contribute to disease expression. Further understanding the imbalance between protective and pathogenic functions for B cells in disease including in understudied target tissue should yield new treatment approaches.
Collapse
|
80
|
Immune Regulation and Antitumor Effect of TIM-1. J Immunol Res 2016; 2016:8605134. [PMID: 27413764 PMCID: PMC4931049 DOI: 10.1155/2016/8605134] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/10/2016] [Accepted: 04/28/2016] [Indexed: 01/25/2023] Open
Abstract
T cells play an important role in antitumor immunity, and the T cell immunoglobulin domain and the mucin domain protein-1 (TIM-1) on its surface, as a costimulatory molecule, has a strong regulatory effect on T cells. TIM-1 can regulate and enhance type 1 immune response of tumor association. Therefore, TIM-1 costimulatory pathways may be a promising therapeutic target in future tumor immunotherapy. This review describes the immune regulation and antitumor effect of TIM-1.
Collapse
|
81
|
Tebbe B, Wilde B, Ye Z, Wang J, Wang X, Jian F, Dolff S, Schedlowski M, Hoyer PF, Kribben A, Witzke O, Hoerning A. Renal Transplant Recipients Treated with Calcineurin-Inhibitors Lack Circulating Immature Transitional CD19+CD24hiCD38hi Regulatory B-Lymphocytes. PLoS One 2016; 11:e0153170. [PMID: 27045291 PMCID: PMC4821620 DOI: 10.1371/journal.pone.0153170] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 03/24/2016] [Indexed: 12/26/2022] Open
Abstract
Background CD19+CD24hiCD38hi transitional immature B-lymphocytes have been demonstrated to play an important role in regulating the alloimmune response in transplant recipients. Here, we analyzed the effect of calcineurin inhibition on these peripherally circulating regulatory B-cells (Breg) in renal transplant recipients receiving cyclosporine A (CsA) or tacrolimus. Methods PBMCs from healthy subjects (HS) (n = 16) and renal transplant recipients (n = 46) were isolated. Flow cytometry was performed for CD19, CD24, CD38 and IL-10 either after isolation or after 72 hours of co-culture in presence of PMA/Ionomycin and TLR9-ligand in presence or absence of increasing concentrations of tacrolimus or CsA. Results The amount of CD19+ B-cells among lymphocytes was ∼9.1% in HS, ∼3.6% in CsA (n = 11, p<0.05) and ∼6.4% in TAC (n = 35, p<0.05) treated patients. Among B-cells, a distinct subset of Breg was found to be 4.7% in HS, 1.4% in tacrolimus treated patients and almost blunted in patients receiving CsA. Similarily, ∼4% of B-cells in HS and even fewer in CsA or tacrolimus treated patients produced IL-10 (0.5% and 1.5%, p<0.05) and this was confirmed both in non-transplanted CsA-treated healthy subjects and in in vitro co-culture experiments. Among 29 patients with <1% of Breg, 9 cases (31%) displayed an allograft rejection in contrast to only one case of rejection (6%) among 17 patients with >1%. Conclusion Calcineurin inhibitors reduce number and IL-10 production of Bregs in the peripheral circulation of both renal transplant recipients and non-transplanted healthy subjects. CNI induced Breg reduction is not restricted to a solid organ transplant setting and is not mediated by co-medication with steroids or MPA. A low proportion of Breg cells is associated with an elevated frequency of allograft rejection events.
Collapse
Affiliation(s)
- Bastian Tebbe
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Zeng Ye
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Junyu Wang
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Xinning Wang
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children’s Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Fu Jian
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Dolff
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter F. Hoyer
- Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children’s Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - André Hoerning
- Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children’s Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
82
|
The clearance of dying cells: table for two. Cell Death Differ 2016; 23:915-26. [PMID: 26990661 PMCID: PMC4987729 DOI: 10.1038/cdd.2015.172] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells of the immune system must constantly survey for, recognize, and efficiently clear the billions of cellular corpses that arise as a result of development, stress, infection, or normal homeostasis. This process, termed efferocytosis, is critical for the prevention of autoimmune and inflammatory disorders, and persistence of dead cells in tissue is characteristic of many human autoimmune diseases, notably systemic lupus erythematosus. The most notable characteristic of the efferocytosis of apoptotic cells is its ‘immunologically silent' response. Although the mechanisms by which phagocytes facilitate engulfment of dead cells has been a well-studied area, the pathways that coordinate to process the ingested corpse and direct the subsequent immune response is an area of growing interest. The recently described pathway of LC3 (microtubule-associated protein 1A/1B-light chain 3)-associated phagocytosis (LAP) has shed some light on this issue. LAP is triggered when an extracellular particle, such as a dead cell, engages an extracellular receptor during phagocytosis, induces the translocation of autophagy machinery, and ultimately LC3 to the cargo-containing phagosome, termed the LAPosome. In this review, we will examine efferocytosis and the impact of LAP on efferocytosis, allowing us to reimagine the impact of the autophagy machinery on innate host defense mechanisms.
Collapse
|
83
|
Abstract
Polymorphisms in the T cell (or transmembrane) immunoglobulin and mucin
domain 1 ( TIM-1) gene, particularly in the mucin
domain, have been associated with atopy and allergic diseases in mice and human.
Genetic- and antibody-mediated studies revealed that Tim-1 functions as a
positive regulator of Th2 responses, while certain antibodies to Tim-1 can
exacerbate or reduce allergic lung inflammation. Tim-1 can also positively
regulate the function of B cells, NKT cells, dendritic cells and mast cells.
However, the precise molecular mechanisms by which Tim-1 modulates immune cell
function are currently unknown. In this study, we have focused on defining
Tim-1-mediated signaling pathways that enhance mast cell activation through the
high affinity IgE receptor (FceRI). Using a Tim-1 mouse model lacking the mucin
domain (Tim-1 Dmucin), we show for the first time that the
polymorphic Tim-1 mucin region is dispensable for normal mast cell activation.
We further show that Tim-4 cross-linking of Tim-1 enhances select signaling
pathways downstream of FceRI in mast cells, including mTOR-dependent signaling,
leading to increased cytokine production but without affecting
degranulation.
Collapse
Affiliation(s)
- Binh Phong
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Immunology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
84
|
Apoptosis inhibitor of macrophage protein enhances intraluminal debris clearance and ameliorates acute kidney injury in mice. Nat Med 2016; 22:183-93. [PMID: 26726878 DOI: 10.1038/nm.4012] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 11/17/2015] [Indexed: 12/13/2022]
Abstract
Acute kidney injury (AKI) is associated with prolonged hospitalization and high mortality, and it predisposes individuals to chronic kidney disease. To date, no effective AKI treatments have been established. Here we show that the apoptosis inhibitor of macrophage (AIM) protein on intraluminal debris interacts with kidney injury molecule (KIM)-1 and promotes recovery from AKI. During AKI, the concentration of AIM increases in the urine, and AIM accumulates on necrotic cell debris within the kidney proximal tubules. The AIM present in this cellular debris binds to KIM-1, which is expressed on injured tubular epithelial cells, and enhances the phagocytic removal of the debris by the epithelial cells, thus contributing to kidney tissue repair. When subjected to ischemia-reperfusion (IR)-induced AKI, AIM-deficient mice exhibited abrogated debris clearance and persistent renal inflammation, resulting in higher mortality than wild-type (WT) mice due to progressive renal dysfunction. Treatment of mice with IR-induced AKI using recombinant AIM resulted in the removal of the debris, thereby ameliorating renal pathology. We observed this effect in both AIM-deficient and WT mice, but not in KIM-1-deficient mice. Our findings provide a basis for the development of potentially novel therapies for AKI.
Collapse
|
85
|
Śledzińska A, Menger L, Bergerhoff K, Peggs KS, Quezada SA. Negative immune checkpoints on T lymphocytes and their relevance to cancer immunotherapy. Mol Oncol 2015; 9:1936-65. [PMID: 26578451 DOI: 10.1016/j.molonc.2015.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/09/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
The term 'inhibitory checkpoint' refers to the broad spectrum of co-receptors expressed by T cells that negatively regulate T cell activation thus playing a crucial role in maintaining peripheral self-tolerance. Co-inhibitory receptor ligands are highly expressed by a variety of malignancies allowing evasion of anti-tumour immunity. Recent studies demonstrate that manipulation of these co-inhibitory pathways can remove the immunological brakes that impede endogenous immune responses against tumours. Antibodies that block the interactions between co-inhibitory receptors and their ligands have delivered very promising clinical responses, as has been shown by recent successful trials targeting the CTLA-4 and PD-1 pathways. In this review, we discuss the mechanisms of action and expression pattern of co-inhibitory receptors on different T cells subsets, emphasising differences between CD4(+) and CD8(+) T cells. We also summarise recent clinical findings utilising immune checkpoint blockade.
Collapse
Affiliation(s)
- Anna Śledzińska
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | - Laurie Menger
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | | | - Karl S Peggs
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK.
| | | |
Collapse
|
86
|
Abstract
Over the last decade it has become evident that in addition to producing antibody, B cells activate the immune system by producing cytokines and via antigen presentation. In addition, B cells also exhibit immunosuppressive functions via diverse regulatory mechanisms. This subset of B cells, known as regulatory B cells (Bregs), contributes to the maintenance of tolerance, primarily via the production of IL-10. Studies in experimental animal models, as well as in patients with autoimmune diseases, have identified multiple Breg subsets exhibiting diverse mechanisms of immune suppression. In this review, we describe the different Breg subsets identified in mice and humans, and their diverse mechanisms of suppression in different disease settings.
Collapse
Affiliation(s)
- Claudia Mauri
- Centre for Rheumatology, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Madhvi Menon
- Centre for Rheumatology, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| |
Collapse
|
87
|
Ding T, Yan F, Cao S, Ren X. Regulatory B cell: New member of immunosuppressive cell club. Hum Immunol 2015; 76:615-21. [PMID: 26385479 DOI: 10.1016/j.humimm.2015.09.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/17/2015] [Accepted: 09/12/2015] [Indexed: 02/07/2023]
Abstract
Historically, the pivotal role of B cells or B lymphocytes in immunity has been attributed to the production of antibodies. They were also demonstrated to present antigens to T cells and to secrete cytokines, thereby acting as positive regulators in immune responses. A series of studies on autoimmune diseases, however, led researchers to find a unique subset of B cells, later described as "regulatory B cells" (Bregs), that has the ability to suppress immune responses. Bregs occur not only in autoimmune diseases, but also in inflammation and transplantation. Furthermore, recently published literatures suggested that Bregs contributed to the growth and metastasis of certain cancers. In this review, we will discuss these unique subsets of B cells in different kinds of disorders, with particular emphasis on the mechanisms of their immunoregulatory role that were collected from mice and humans.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China; National Clinical Research Center of Cancer, China
| | - Fan Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China; National Clinical Research Center of Cancer, China
| | - Shui Cao
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China; National Clinical Research Center of Cancer, China.
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China; National Clinical Research Center of Cancer, China.
| |
Collapse
|
88
|
Brooks CR, Yeung MY, Brooks YS, Chen H, Ichimura T, Henderson JM, Bonventre JV. KIM-1-/TIM-1-mediated phagocytosis links ATG5-/ULK1-dependent clearance of apoptotic cells to antigen presentation. EMBO J 2015; 34:2441-64. [PMID: 26282792 DOI: 10.15252/embj.201489838] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 07/01/2015] [Indexed: 12/14/2022] Open
Abstract
Phagocytosis of apoptotic cells by both professional and semi-professional phagocytes is required for resolution of organ damage and maintenance of immune tolerance. KIM-1/TIM-1 is a phosphatidylserine receptor that is expressed on epithelial cells and can transform the cells into phagocytes. Here, we demonstrate that KIM-1 phosphorylation and association with p85 results in encapsulation of phagosomes by lipidated LC3 in multi-membrane organelles. KIM-1-mediated phagocytosis is not associated with increased ROS production, and NOX inhibition does not block LC3 lipidation. Autophagy gene expression is required for efficient clearance of apoptotic cells and phagosome maturation. KIM-1-mediated phagocytosis leads to pro-tolerogenic antigen presentation, which suppresses CD4 T-cell proliferation and increases the percentage of regulatory T cells in an autophagy gene-dependent manner. Taken together, these data reveal a novel mechanism of epithelial biology linking phagocytosis, autophagy and antigen presentation to regulation of the inflammatory response.
Collapse
Affiliation(s)
- Craig R Brooks
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Transplantation Research Center, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Yang S Brooks
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA, USA Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Takaharu Ichimura
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Joseph V Bonventre
- Department of Medicine, Renal Division, Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
89
|
Abstract
PURPOSE OF REVIEW B cells are known to play a central role in humoral immunity and to boost cellular immunity, however, in a variety of experimental models, B-cell subsets ameliorate inflammation and autoimmune disease, indicating that they can also play a regulatory role. Here, we highlight the advances in regulatory B-cell (Breg) biology of the past year with an emphasis on findings pertinent to transplantation. Several recent observations highlight the relevance to clinical transplantation. Data from at least three independent groups demonstrated that spontaneously tolerant renal transplant recipients exhibit a peripheral blood B-cell signature although the significance of these data remains unclear. Moreover, new data suggest that regulatory B cells may serve as a biomarker for long-term allograft outcomes. Finally, recent evidence suggesting that plasma cells may be an essential component of Bregs raises new concerns about targeting antibody producing cells. RECENT FINDINGS We describe new information on Breg mechanisms of action to suppress the alloresponse, signals to expand Bregs in vitro, and more functional evidence of Breg involvement in operationally tolerant kidney patients and in maintaining stable allograft function. SUMMARY Although lymphocyte depletion remains central to tolerance induction therapy, the sparing or expansion of regulatory B cells may be an additional strategy to preempt graft rejection.
Collapse
Affiliation(s)
- James I. Kim
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Thier 8, Boston, MA 02114
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical School, 200 Lothrop Street, E1555 Biomedical Science Tower, Pittsburgh, PA 15261
| | - James F. Markmann
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Thier 8, Boston, MA 02114
| |
Collapse
|
90
|
Li G, Wulan H, Song Z, Paik PA, Tsao ML, Goodman GM, MacEachern PT, Downey RS, Jankowska AJ, Rabinowitz YM, Learch TB, Song DZ, Yuan JJ, Zheng S, Zheng Z. Regulatory B Cell Function Is Suppressed by Smoking and Obesity in H. pylori-Infected Subjects and Is Correlated with Elevated Risk of Gastric Cancer. PLoS One 2015; 10:e0134591. [PMID: 26226399 PMCID: PMC4520600 DOI: 10.1371/journal.pone.0134591] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 07/13/2015] [Indexed: 12/29/2022] Open
Abstract
Helicobacter pylori infection occurs in more than half of the world’s population and is the main cause for gastric cancer. A series of lifestyle and nutritional factors, such as tobacco smoking and obesity, have been found to elevate the risk for cancer development. In this study, we sought to determine the immunological aspects during H. pylori infection and gastric cancer development. We found that B cells from H. pylori-infected patients presented altered composition and function compared to uninfected patients. IL-10-expressing CD24+CD38+ B cells were upregulated in H. pylori-infected patients, contained potent regulatory activity in inhibiting T cell pro-inflammatory cytokine secretion, and responded directly to H. pylori antigen stimulation. Interestingly, in H. pylori-infected smoking subjects and obese subjects, the number of IL-10+ B cells and CD24+CD38+ B cells were reduced compared to H. pylori-infected asymptomatic subjects. Regulatory functions mediated by CD24+CD38+ B cells were also impaired. In addition, gastric cancer positive patients had reduced IL-10-producing B cell frequencies after H. pylori-stimulation. Altogether, these data suggest that in H. pylori-infection, CD24+CD38+ B cell is upregulated and plays a role in suppressing pro-inflammatory responses, possibly through IL-10 production, a feature that was not observed in smoking and obese patients.
Collapse
Affiliation(s)
- Guanggang Li
- Affiliated Bayi Brain Hospital, General Hospital of Beijing Military Command, Beijing, 100700, China
| | - Hasi Wulan
- Department of Plastic and Reconstructive Surgery, PLA General Hospital, Beijing, 100853, China
| | - Zongchang Song
- Department of Oncology, The 155th Central Hospital of PLA, Kaifeng, 475003, China
| | - Paul A Paik
- Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Ming L Tsao
- Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Gary M Goodman
- Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Paul T MacEachern
- Georgetown University Medical Center, Washington DC, United States of America
| | - Robert S Downey
- Georgetown University Medical Center, Washington DC, United States of America
| | - Anna J Jankowska
- Georgetown University Medical Center, Washington DC, United States of America
| | - Yaron M Rabinowitz
- Georgetown University Medical Center, Washington DC, United States of America
| | - Thomas B Learch
- University of Maryland Medical Center. Baltimore, MD, United States of America
| | - David Z Song
- University Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Ji J Yuan
- University Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Shihang Zheng
- Department of Gastroenterology, Shengjing Hospital, China Medical University, Shenyang, 110022, China
| | - Zhendong Zheng
- Department of Medical Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, 200070, China; Department of Oncology, General Hospital of Shenyang Military Area Command, Shenyang, 110840, China
| |
Collapse
|
91
|
Abstract
Regulatory B (Breg) cells are immunosuppressive cells that support immunological tolerance. Through the production of interleukin-10 (IL-10), IL-35, and transforming growth factor β (TGF-β), Breg cells suppress immunopathology by prohibiting the expansion of pathogenic T cells and other pro-inflammatory lymphocytes. Recent work has shown that different inflammatory environments induce distinct Breg cell populations. Although these findings highlight the relevance of inflammatory signals in the differentiation of Breg cells, they also raise other questions about Breg cell biology and phenotype. For example, what are the functional properties and phenotype of Breg cells? Can a Breg cell arise at every stage in B cell development? Is inflammation the primary requisite for Breg cell differentiation? Here, we use these questions to discuss the advances in understanding Breg cell biology, with a particular emphasis on their ontogeny; we propose that multiple Breg cell subsets can be induced in response to inflammation at different stages in development.
Collapse
|
92
|
Baba Y, Matsumoto M, Kurosaki T. Signals controlling the development and activity of regulatory B-lineage cells. Int Immunol 2015; 27:487-93. [PMID: 25957265 DOI: 10.1093/intimm/dxv027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/01/2015] [Indexed: 12/25/2022] Open
Abstract
The fundamental concepts surrounding B cells with inhibitory function (regulatory B cells) are now being established. In the context of autoimmune and inflammatory animal models, B cells play an immunomodulatory role via IL-10 production and contribute to limitation of the pathogenesis. Recent studies have notably identified the human counterparts of these cells, which have been suggested to be relevant to the pathophysiology of disease. Clear criteria to identify these cell subsets and the key molecular mechanisms underlying their physiological features are required for understanding the big picture of regulatory B cells. Plasmablasts have recently been identified as a major IL-10-producing regulatory B-cell subset and Ca(2+) signaling has furthermore been found to contribute to B-cell IL-10 expression. In this review, the signaling components controlling IL-10-dependent B-cell regulatory function and the development of IL-10-competent/-producing B cells and plasmablasts are discussed.
Collapse
Affiliation(s)
- Yoshihiro Baba
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Masanori Matsumoto
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
93
|
Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D, Willimsky G, Ammirante M, Strasner A, Hansel DE, Jamieson C, Kane CJ, Klatte T, Birner P, Kenner L, Karin M. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature 2015; 521:94-8. [PMID: 25924065 PMCID: PMC4501632 DOI: 10.1038/nature14395] [Citation(s) in RCA: 440] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 03/09/2015] [Indexed: 12/14/2022]
Abstract
Cancer-associated genetic alterations induce expression of tumour antigens that can activate CD8(+) cytotoxic T cells (CTLs), but the microenvironment of established tumours promotes immune tolerance through poorly understood mechanisms. Recently developed therapeutics that overcome tolerogenic mechanisms activate tumour-directed CTLs and are effective in some human cancers. Immune mechanisms also affect treatment outcome, and certain chemotherapeutic drugs stimulate cancer-specific immune responses by inducing immunogenic cell death and other effector mechanisms. Our previous studies revealed that B cells recruited by the chemokine CXCL13 into prostate cancer tumours promote the progression of castrate-resistant prostate cancer by producing lymphotoxin, which activates an IκB kinase α (IKKα)-BMI1 module in prostate cancer stem cells. Because castrate-resistant prostate cancer is refractory to most therapies, we examined B cell involvement in the acquisition of chemotherapy resistance. Here we focus on oxaliplatin, an immunogenic chemotherapeutic agent that is effective in aggressive prostate cancer. We show that mouse B cells modulate the response to low-dose oxaliplatin, which promotes tumour-directed CTL activation by inducing immunogenic cell death. Three different mouse prostate cancer models were refractory to oxaliplatin unless genetically or pharmacologically depleted of B cells. The crucial immunosuppressive B cells are plasmocytes that express IgA, interleukin (IL)-10 and programmed death ligand 1 (PD-L1), the appearance of which depends on TGFβ receptor signalling. Elimination of these cells, which also infiltrate human-therapy-resistant prostate cancer, allows CTL-dependent eradication of oxaliplatin-treated tumours.
Collapse
Affiliation(s)
- Shabnam Shalapour
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Joan Font-Burgada
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Giuseppe Di Caro
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Zhenyu Zhong
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Elsa Sanchez-Lopez
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Debanjan Dhar
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Gerald Willimsky
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
| | - Massimo Ammirante
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Amy Strasner
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Donna E Hansel
- Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| | - Christina Jamieson
- Department of Surgery, Urology Division, University of California San Diego, 3855 Health Sciences Drive, San Diego, California 92093, USA
| | - Christopher J Kane
- Department of Surgery, Urology Division, University of California San Diego, 3855 Health Sciences Drive, San Diego, California 92093, USA
| | - Tobias Klatte
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Birner
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Lukas Kenner
- 1] Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria [2] Clinical Institute of Pathology, Ludwig Boltzmann Institute for Cancer Research, Medical University of Vienna, Unit of Pathology of Laboratory Animals (UPLA), University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Michael Karin
- 1] Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, San Diego, California 92093, USA [2] Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, San Diego, California 92093, USA
| |
Collapse
|
94
|
Yeung MY, Ding Q, Brooks CR, Xiao S, Workman CJ, Vignali DA, Ueno T, Padera RF, Kuchroo VK, Najafian N, Rothstein DM. TIM-1 signaling is required for maintenance and induction of regulatory B cells. Am J Transplant 2015; 15:942-53. [PMID: 25645598 PMCID: PMC4530122 DOI: 10.1111/ajt.13087] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 01/25/2023]
Abstract
Apart from their role in humoral immunity, B cells can exhibit IL-10-dependent regulatory activity (Bregs). These regulatory subpopulations have been shown to inhibit inflammation and allograft rejection. However, our understanding of Bregs has been hampered by their rarity, lack of a specific marker, and poor insight into their induction and maintenance. We previously demonstrated that T cell immunoglobulin mucin domain-1 (TIM-1) identifies over 70% of IL-10-producing B cells, irrespective of other markers. We now show that TIM-1 is the primary receptor responsible for Breg induction by apoptotic cells (ACs). However, B cells that express a mutant form of TIM-1 lacking the mucin domain (TIM-1(Δmucin) ) exhibit decreased phosphatidylserine binding and are unable to produce IL-10 in response to ACs or by specific ligation with anti-TIM-1. TIM-1(Δmucin) mice also exhibit accelerated allograft rejection, which appears to be due in part to their defect in both baseline and induced IL-10(+) Bregs, since a single transfer of WT TIM-1(+) B cells can restore long-term graft survival. These data suggest that TIM-1 signaling plays a direct role in Breg maintenance and induction both under physiological conditions (in response to ACs) and in response to therapy through TIM-1 ligation. Moreover, they directly demonstrate that the mucin domain regulates TIM-1 signaling.
Collapse
Affiliation(s)
- Melissa Y. Yeung
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Qing Ding
- Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Craig R. Brooks
- Renal Division, Harvard Medical School, Boston, Massachusetts, USA
| | - Sheng Xiao
- Center for Neurologic Disease, Harvard Medical School, Boston, Massachusetts, USA
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Dario A.A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Takuya Ueno
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert F. Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vijay K. Kuchroo
- Center for Neurologic Disease, Harvard Medical School, Boston, Massachusetts, USA
| | - Nader Najafian
- Transplantation Research Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Nephrology, Cleveland Clinic Florida, Weston, FL, USA
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
95
|
Yang L, Brooks CR, Xiao S, Sabbisetti V, Yeung MY, Hsiao LL, Ichimura T, Kuchroo V, Bonventre JV. KIM-1-mediated phagocytosis reduces acute injury to the kidney. J Clin Invest 2015; 125:1620-36. [PMID: 25751064 DOI: 10.1172/jci75417] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/22/2015] [Indexed: 12/13/2022] Open
Abstract
Kidney injury molecule 1 (KIM-1, also known as TIM-1) is markedly upregulated in the proximal tubule after injury and is maladaptive when chronically expressed. Here, we determined that early in the injury process, however, KIM-1 expression is antiinflammatory due to its mediation of phagocytic processes in tubule cells. Using various models of acute kidney injury (AKI) and mice expressing mutant forms of KIM-1, we demonstrated a mucin domain-dependent protective effect of epithelial KIM-1 expression that involves downregulation of innate immunity. Deletion of the mucin domain markedly impaired KIM-1-mediated phagocytic function, resulting in increased proinflammatory cytokine production, decreased antiinflammatory growth factor secretion by proximal epithelial cells, and a subsequent increase in tissue macrophages. Mice expressing KIM-1Δmucin had greater functional impairment, inflammatory responses, and mortality in response to ischemia- and cisplatin-induced AKI. Compared with primary renal proximal tubule cells isolated from KIM-1Δmucin mice, those from WT mice had reduced proinflammatory cytokine secretion and impaired macrophage activation. The antiinflammatory effect of KIM-1 expression was due to the interaction of KIM-1 with p85 and subsequent PI3K-dependent downmodulation of NF-κB. Hence, KIM-1-mediated epithelial cell phagocytosis of apoptotic cells protects the kidney after acute injury by downregulating innate immunity and inflammation.
Collapse
|
96
|
Xiao S, Brooks CR, Sobel RA, Kuchroo VK. Tim-1 is essential for induction and maintenance of IL-10 in regulatory B cells and their regulation of tissue inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 194:1602-8. [PMID: 25582854 DOI: 10.4049/jimmunol.1402632] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T cell Ig and mucin domain (Tim)-1 identifies IL-10-producing regulatory B cells (Bregs). Mice on the C57BL/6 background harboring a loss-of-function Tim-1 mutant showed progressive loss of IL-10 production in B cells and with age developed severe multiorgan tissue inflammation. We demonstrate that Tim-1 expression and signaling in Bregs are required for optimal production of IL-10. B cells with Tim-1 defects have impaired IL-10 production but increased proinflammatory cytokine production, including IL-1 and IL-6. Tim-1-deficient B cells promote Th1 and Th17 responses but inhibit the generation of regulatory T cells (Foxp3(+) and IL-10-producing type 1 regulatory T cells) and enhance the severity of experimental autoimmune encephalomyelitis. Mechanistically, Tim-1 on Bregs is required for apoptotic cell (AC) binding to Bregs and for AC-induced IL-10 production in Bregs. Treatment with ACs reduces the severity of experimental autoimmune encephalomyelitis in hosts with wild-type but not Tim-1-deficient Bregs. Collectively, these findings suggest that in addition to serving as a marker for identifying IL-10-producing Bregs, Tim-1 is also critical for maintaining self-tolerance by regulating IL-10 production in Bregs.
Collapse
Affiliation(s)
- Sheng Xiao
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115;
| | - Craig R Brooks
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Raymond A Sobel
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304; and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115;
| |
Collapse
|
97
|
Regulation of T cell trafficking by the T cell immunoglobulin and mucin domain 1 glycoprotein. Trends Mol Med 2014; 20:675-84. [DOI: 10.1016/j.molmed.2014.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/30/2022]
|
98
|
Kimani SG, Geng K, Kasikara C, Kumar S, Sriram G, Wu Y, Birge RB. Contribution of Defective PS Recognition and Efferocytosis to Chronic Inflammation and Autoimmunity. Front Immunol 2014; 5:566. [PMID: 25426118 PMCID: PMC4226236 DOI: 10.3389/fimmu.2014.00566] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/23/2014] [Indexed: 12/14/2022] Open
Abstract
The rapid and efficient clearance of apoptotic cells results in the elimination of auto-antigens and provides a strong anti-inflammatory and immunosuppressive signal to prevent autoimmunity. While professional and non-professional phagocytes utilize a wide array of surface receptors to recognize apoptotic cells, the recognition of phosphatidylserine (PS) on apoptotic cells by PS receptors on phagocytes is the emblematic signal for efferocytosis in metazoans. PS-dependent efferocytosis is associated with the production of anti-inflammatory factors such as IL-10 and TGF-β that function, in part, to maintain tolerance to auto-antigens. In contrast, when apoptotic cells fail to be recognized and processed for degradation, auto-antigens persist, such as self-nucleic acids, which can trigger immune activation leading to autoantibody production and autoimmunity. Despite the fact that genetic mouse models clearly demonstrate that loss of PS receptors can lead to age-dependent auto-immune diseases reminiscent of systemic lupus erythematosus (SLE), the link between PS and defective clearance in chronic inflammation and human autoimmunity is not well delineated. In this perspective, we review emerging questions developing in the field that may be of relevance to SLE and human autoimmunity.
Collapse
Affiliation(s)
- Stanley Gititu Kimani
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| | - Ke Geng
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| | - Canan Kasikara
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| | - Ganapathy Sriram
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| | - Yi Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University , Suzhou , China ; Sol Sherry Thrombosis Research Center, Temple University School of Medicine , Philadelphia, PA , USA
| | - Raymond B Birge
- Department of Biochemistry and Molecular Biology, Rutgers School of Biomedical and Health Sciences - Cancer Center , Newark, NJ , USA
| |
Collapse
|
99
|
Braza F, Chesne J, Castagnet S, Magnan A, Brouard S. Regulatory functions of B cells in allergic diseases. Allergy 2014; 69:1454-63. [PMID: 25060230 DOI: 10.1111/all.12490] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 12/24/2022]
Abstract
B cells are essentially described for their capacity to produce antibodies ensuring anti-infectious immunity or deleterious responses in the case of autoimmunity or allergy. However, abundant data described their ability to restrain inflammation by diverse mechanisms. In allergy, some regulatory B-cell subsets producing IL-10 have been recently described as potent suppressive cells able to restrain inflammatory responses both in vitro and in vivo by regulatory T-cell differentiation or directly inhibiting T-cell-mediated inflammation. A specific deficit in regulatory B cells participates to more severe allergic inflammation. Induction of allergen tolerance through specific immunotherapy induces a specific expansion of these cells supporting their role in establishment of allergen tolerance. However, the regulatory functions carried out by B cells are not exclusively IL-10 dependent. Indeed, other regulatory mechanisms mediated by B cells are (i) the production of TGF-β, (ii) the promotion of T-cell apoptosis by Fas-Fas ligand or granzyme-B pathways, and (iii) their capacity to produce inhibitory IgG4 and sialylated IgG able to mediate anti-inflammatory mechanisms. This points to Bregs as interesting targets for the development of new therapies to induce allergen tolerance. In this review, we highlight advances in the study of regulatory mechanisms mediated by B cells and outline what is known about their phenotype as well as their suppressive role in allergy from studies in both mice and humans.
Collapse
Affiliation(s)
- F. Braza
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| | - J. Chesne
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| | - S. Castagnet
- Laboratoire HLA; Établissement Français du Sang; Nantes France
| | - A. Magnan
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
- CHU Nantes; l'institut du Thorax; Service de Pneumologie; Nantes France
| | - S. Brouard
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| |
Collapse
|
100
|
Moller-Tank S, Maury W. Phosphatidylserine receptors: enhancers of enveloped virus entry and infection. Virology 2014; 468-470:565-580. [PMID: 25277499 PMCID: PMC4252826 DOI: 10.1016/j.virol.2014.09.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 07/15/2014] [Accepted: 09/08/2014] [Indexed: 12/23/2022]
Abstract
A variety of both RNA and DNA viruses envelop their capsids in a lipid bilayer. One of the more recently appreciated benefits this envelope is incorporation of phosphatidylserine (PtdSer). Surface exposure of PtdSer disguises viruses as apoptotic bodies; tricking cells into engulfing virions. This mechanism is termed apoptotic mimicry. Several PtdSer receptors have been identified to enhance virus entry and we have termed this group of proteins PtdSer-mediated virus entry enhancing receptors or PVEERs. These receptors enhance entry of a range of enveloped viruses. Internalization of virions by PVEERs provides a broad mechanism of entry with little investment by the virus itself. PVEERs may allow some viruses to attach to cells, thereby making viral glycoprotein/cellular receptor interactions more probable. Alternatively, other viruses may rely entirely on PVEERs for internalization into endosomes. This review provides an overview of PtdSer receptors that serve as PVEERs and the biology behind virion/PVEER interaction. Phosphatidylserine (PtdSer) receptors can mediate entry of enveloped viruses. PtdSer is present on the outer leaflet of the virion envelope. PtdSer receptors are expressed on a variety of primary cells and cell lines. Characteristics of PtdSer receptors that mediate virus entry are defined.
Collapse
Affiliation(s)
- Sven Moller-Tank
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Wendy Maury
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|