1
|
Yao Y, Liu YY, Li JF, Chen YS, Shi L, Shen Y, Yang LL, Yang Q. Indoleamine 2,3-dioxygenase 1 alters the proportions of B cell subpopulations in the microenvironment of acute myeloid leukemia. MOLECULAR BIOMEDICINE 2025; 6:23. [PMID: 40234305 PMCID: PMC12000501 DOI: 10.1186/s43556-025-00262-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Acute myeloid leukemia (AML), the most common leukemia in adults, exhibits immune escape characteristics like solid tumors. The expression of indoleamine 2,3-dioxygenase 1 (IDO1), a well-recognized immune checkpoint, has been detected in AML blast cells and is associated with poor clinical outcome. Although an imbalance of B cell subpopulations exists in AML patients' bone marrow microenvironment, the role of B cells and their interaction with IDO1 in AML have yet to be elucidated. Herein, with bioinformatic analysis, we found the close correlations between IDO1 expression and survival and B cell subpopulation proportions in AML patients. Further, our investigation into IDO1 expression and activity, B cell subpopulation proportions and immunosuppressive interleukin-10 (IL-10) level in AML cells and clinical samples revealed significant findings. Using a co-culture system of healthy human PBMCs and AML cell lines, we demonstrated that high IDO1 expression in AML cells could alter the proportions of total B, regulatory B and memory B cells, and increased the level of IL-10. Finally, with the IDO1 inhibitor RY103 designed by our laboratory, we found that IDO1 inhibition had good anti-leukemic effect and restored the abnormal proportions of B cell subpopulations in AML mice. Our study is the first to reveal the modulation of IDO1 on B cell subpopulations in AML, making a significant breakthrough in understanding the immune escape mechanisms of AML. Application of IDO1 inhibitor, such as RY103, targeting the imbalance of B cell subpopulations can lead to innovative treatments for AML.
Collapse
MESH Headings
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Humans
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Tumor Microenvironment/immunology
- Animals
- Interleukin-10/metabolism
- Mice
- Cell Line, Tumor
- Male
- Female
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Middle Aged
- Adult
Collapse
Affiliation(s)
- Yu Yao
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Yu-Ying Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Jian-Feng Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yun-Shuo Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Shi
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Yang Shen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Li-Li Yang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Qing Yang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| |
Collapse
|
2
|
Liao J, Yang Y, Li J, Liu Z, Song S, Zeng Y, Wang Y. Regulatory B cells, the key regulator to induce immune tolerance in organ transplantation. Front Immunol 2025; 16:1561171. [PMID: 40264774 PMCID: PMC12011811 DOI: 10.3389/fimmu.2025.1561171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
In solid organ transplantation, especially renal transplantation, for the induction of immune tolerance, accumulating evidence has revealed that Regulatory B cells (Breg) play a crucial role in stimulating immune tolerance, alleviating immune responses, and improving graft survival. We describe the heterogeneous nature of Bregs, focusing on their defining surface markers and regulatory functions. Meanwhile, the major cytokine secretion function and the correlation between Breg and Treg or other immune checkpoints to balance the immune responses are addressed. Furthermore, we summarized the intrinsic and extrinsic pathways or costimulatory stimuli for the differentiation from naïve B cells. More importantly, we summarized the progression of the immune tolerance induction role of Breg in solid organ (kidney, liver, heart, lung, and islet) transplantation. This is an up-to-date review from the origin of Breg to the function of Breg in solid organ transplantation and how it induces immune tolerance in both murine models and human solid organ transplantation.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yixin Yang
- Department of Clinical Medicine, The First Clinical Medical College of Norman Bethune University of Medical Sciences, Jilin, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yu Zeng
- Department of Hyperbaric Oxygen, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
3
|
Altulea D, van den Born J, Bijma T, Bonasia C, Inrueangsri N, Lammerts R, Berger S, Heeringa P, Sanders JS. Comprehensive Phenotyping and Cytokine Production of Circulating B Cells Associate Resting Memory B Cells With Early Antibody-mediated Rejection in Kidney Transplant Recipients. Transplant Direct 2025; 11:e1775. [PMID: 40124243 PMCID: PMC11927649 DOI: 10.1097/txd.0000000000001775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Background B cells play a crucial role in kidney transplantation through antibody production and cytokine secretion. To better understand their impact on kidney transplantation, this retrospective study aimed to characterize circulating B-cell phenotypes and cytokine production in a cohort of kidney transplant patients to identify whether pretransplant donor-specific antibodies (DSAs) or biopsy-proven rejection is associated with different B-cell profiles. Methods Pretransplant cryopreserved peripheral blood mononuclear cells were obtained from 96 kidney transplant recipients, of whom 42 had pretransplant DSAs. The cells underwent surface marker staining using a 33-color spectral flow cytometry panel for B-cell phenotyping. Simultaneously, cells were stimulated for interleukin-10, tumor necrosis factor-α, and interleukin-6 production, and analyzed with a 6-color panel. Results Rejection was linked to decreased naive B cells and increased plasmablasts, CD27+ memory B cells, and memory B-cell subsets (all P < 0.04) compared with no rejection. Cytokine-producing B cells and immune regulatory molecule expression showed no significant differences. Multivariate analysis identified resting memory B cells (CD27+CD21+) and pretransplant DSAs as significantly associated with rejection (P = 0.01; odds ratio [OR], 1.07; P = 0.02; OR, 3.10, respectively). Cox regression analysis revealed resting memory B cells were associated with early antibody-mediated rejection (P = 0.04; OR, 1.05). Conclusions B-cell subset distributions differed between patients with and without rejection. Resting memory B-cell frequency was associated with increased early antibody-mediated rejection risk, whereas cytokine production and immune checkpoint expression did not influence rejection. The results suggest that B-cell subset composition could aid in rejection risk assessment and serve as a potential pretransplant diagnostic parameter.
Collapse
Affiliation(s)
- Dania Altulea
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joost van den Born
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Theo Bijma
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Carlo Bonasia
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nanthicha Inrueangsri
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rosa Lammerts
- Transplantation Immunology, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan-Stephan Sanders
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Ye R, Li S, Li Y, Shi K, Li L. Revealing the role of regulatory b cells in cancer: development, function and treatment significance. Cancer Immunol Immunother 2025; 74:125. [PMID: 39998678 PMCID: PMC11861783 DOI: 10.1007/s00262-025-03973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
B cells are essential components of the immune response, primarily recognized for their ability to produce antibodies. However, emerging research reveals their important roles in regulating immune responses and influencing tumor development, independent of antibodies. The connection between tumor progression and alterations in the tumor microenvironment is well-established, as immune infiltrating cells can enhance the survival of tumor cells by modifying their surroundings. Despite this, the majority of studies have focused on T cells and macrophages, creating a gap in our understanding of B cells. Regulatory B cells (Bregs) represent a crucial subpopulation that plays a significant role in maintaining immune balance. They may have a substantial impact on tumor immunity by negatively regulating tumor-infiltrating immune cells. This paper reviews the existing literature on Bregs, examining their development, phenotypes, functions, and the mechanisms through which they exert their regulatory effects. Furthermore, we highlight their potential interventional roles and prognostic significance in cancer therapy. By addressing the current gaps in knowledge regarding Bregs within tumors, we hope to inspire further research that could lead to innovative cancer treatments and improved outcomes for patients.
Collapse
Affiliation(s)
- Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yuxiao Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Kaixin Shi
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
5
|
Lin L, Luo J, Cai Y, Wu X, Zhou L, Li T, Wang X, Xu H. Mass cytometry identifies imbalance of multiple immune-cell subsets associated with biologics treatment in ankylosing spondylitis. Int J Rheum Dis 2024; 27:e15378. [PMID: 39420773 DOI: 10.1111/1756-185x.15378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE This study aims to comprehensively investigate immune-cell landscapes in ankylosing spondylitis (AS) patients and explore longitudinal immunophenotyping changes induced by biological agents. METHODS We employed mass cytometry with 35 cellular markers to analyze blood samples from 34 AS patients and 13 healthy controls (HC). Eleven AS patients were re-evaluated 1 month (4 patients) and 3 months (7 patients) after treatment with biological agents. Flow Self-Organizing Maps (FlowSOM) clustering was performed to identify specific cellular metaclusters. We compared cellular abundances across distinct subgroups and validated subset differences using gating strategies in flow cytometry scatter plots, visualized with FlowJo software. The proportions of differential subsets were then used for intercellular and clinical correlation analysis, as well as for constructing diagnostic models based on the random forest algorithm. RESULTS In AS patients, we identified and validated nine different immune-cell subsets compared to HC. Three subsets increased: helper T-cell 17 (Th17), mucosa-associated invariant T-cell (MAIT), and classical monocytes (CM). Six subsets decreased: effector memory T-cell (TEM), naïve B cells, transitional B cells, IL10+ memory B cells, non-classical monocytes (NCM), and neutrophils. Treatments with biological agents could rectify cellular abnormalities, particularly the imbalance of CM/NCM. Furthermore, these subsets may serve as biomarkers for assessing disease activity and constructing effective diagnostic models for AS. CONCLUSION These findings provide novel insights into the specific patterns of immune cell in AS, facilitating the further development of novel biomarkers and potential therapeutic targets for AS patients.
Collapse
Affiliation(s)
- Li Lin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Jing Luo
- School of Medicine, Tsinghua University, Beijing, China
| | - Yue Cai
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Ting Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Xiaobing Wang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- National Key Laboratory for Immunity and Inflammation, Shanghai, China
- School of Medicine, Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
6
|
Uvarova AN, Zheremyan EA, Ustiugova AS, Murashko MM, Bogomolova EA, Demin DE, Stasevich EM, Kuprash DV, Korneev KV. Autoimmunity-Associated SNP rs3024505 Disrupts STAT3 Binding in B Cells, Leading to IL10 Dysregulation. Int J Mol Sci 2024; 25:10196. [PMID: 39337678 PMCID: PMC11432243 DOI: 10.3390/ijms251810196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Interleukin 10 (IL10) is a major anti-inflammatory cytokine that acts as a master regulator of the immune response. A single nucleotide polymorphism rs3024505(C/T), located downstream of the IL10 gene, is associated with several aggressive inflammatory diseases, including systemic lupus erythematosus, Sjögren's syndrome, Crohn's disease, and ulcerative colitis. In such autoimmune pathologies, IL10-producing B cells play a protective role by decreasing the level of inflammation and restoring immune homeostasis. This study demonstrates that rs3024505 is located within an enhancer that augments the activity of the IL10 promoter in a reporter system based on a human B cell line. The common rs3024505(C) variant creates a functional binding site for the transcription factor STAT3, whereas the risk allele rs3024505(T) disrupts STAT3 binding, thereby reducing the IL10 promoter activity. Our findings indicate that B cells from individuals carrying the minor rs3024505(T) allele may produce less IL10 due to the disrupted STAT3 binding site, contributing to the progression of inflammatory pathologies.
Collapse
Affiliation(s)
- Aksinya N. Uvarova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elina A. Zheremyan
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alina S. Ustiugova
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Matvey M. Murashko
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Elvina A. Bogomolova
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Denis E. Demin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ekaterina M. Stasevich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Dmitry V. Kuprash
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Kirill V. Korneev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Laboratory of Intracellular Signaling in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
7
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
8
|
Sakai J, Yang J, Chou CK, Wu WW, Akkoyunlu M. B cell receptor-induced IL-10 production from neonatal mouse CD19 +CD43 - cells depends on STAT5-mediated IL-6 secretion. eLife 2023; 12:83561. [PMID: 36735294 PMCID: PMC9934864 DOI: 10.7554/elife.83561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
Newborns are unable to reach the adult-level humoral immune response partly due to the potent immunoregulatory role of IL-10. Increased IL-10 production by neonatal B cells has been attributed to the larger population of IL-10-producting CD43+ B-1 cells in neonates. Here, we show that neonatal mouse CD43- non-B-1 cells also produce substantial amounts of IL-10 following B cell antigen receptor (BCR) activation. In neonatal mouse CD43- non-B-1 cells, BCR engagement activated STAT5 under the control of phosphorylated forms of signaling molecules Syk, Btk, PKC, FAK, and Rac1. Neonatal STAT5 activation led to IL-6 production, which in turn was responsible for IL-10 production in an autocrine/paracrine fashion through the activation of STAT3. In addition to the increased IL-6 production in response to BCR stimulation, elevated expression of IL-6Rα expression in neonatal B cells rendered them highly susceptible to IL-6-mediated STAT3 phosphorylation and IL-10 production. Finally, IL-10 secreted from neonatal mouse CD43- non-B-1 cells was sufficient to inhibit TNF-α secretion by macrophages. Our results unveil a distinct mechanism of IL-6-dependent IL-10 production in BCR-stimulated neonatal CD19+CD43- B cells.
Collapse
Affiliation(s)
- Jiro Sakai
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| | - Jiyeon Yang
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| | - Chao-Kai Chou
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, United States Food and Drug AdministrationSilver SpringUnited States
| | - Wells W Wu
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, United States Food and Drug AdministrationSilver SpringUnited States
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, The US Food and Drug AdministrationSilver SpringUnited States
| |
Collapse
|
9
|
Ren J, Zhuo Y, He F, Lv L, Xing M, Guo Y, Zhang Y, Liu J, Li Y, Bai T, Chen Y, Li G, Qin Z, Zhou D. Longitudinal Immune Profiling Highlights CD4+ T Cell Exhaustion Correlated with Liver Fibrosis in Schistosoma japonicum Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:82-95. [PMID: 36445332 DOI: 10.4049/jimmunol.2200301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022]
Abstract
Schistosomiasis remains an important public health concern. The eggs deposited in livers invoke a Th2-dominant response, which mediates the fibrotic granulomatous response. However, the mechanisms involved in this immunopathological process are still not perfectly clear. Here, we report a single-cell transcriptional landscape of longitudinally collected BALB/c mouse splenocytes at different time points after Schistosoma japonicum infection. We found that exhausted CD4+ T cells were enriched after infection, changing from coproducing multiple cytokines to predominantly producing the Th2 cytokine IL-4. Regulatory B cells had high expression of Fcrl5, Ptpn22, and Lgals1, potentially regulating exhausted CD4+ T cells via direct PD-1-PD-L2 and PD-1-PD-L1 interactions. Within the myeloid compartment, the number of precursor and immature neutrophils sharply increased after infection. Moreover, dendritic cells, macrophages, and basophils showed inhibitory interactions with exhausted CD4+ T cells. Besides, in mouse livers, we found that exhausted CD4+ T cells were distributed around egg granuloma, promoting collagen expression in primary mouse hepatic stellate cells via IL-4 secretion, resulting in liver fibrosis. Our study provides comprehensive characterization of the composition and cellular states of immune cells with disease progression, which will facilitate better understanding of the mechanism underlying liver fibrotic granulomatous response in schistosomiasis.
Collapse
Affiliation(s)
- Jiling Ren
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yue Zhuo
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Furong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lihui Lv
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yingying Guo
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuchao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science, Fudan University, Shanghai, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tinghui Bai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanan Chen
- School of Medicine, Nankai University, Tianjin, China
| | - Guangru Li
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiqiang Qin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Wang C, Xu H, Gao R, Leng F, Huo F, Li Y, Liu S, Xu M, Bai J. CD19 +CD24 hiCD38 hi regulatory B cells deficiency revealed severity and poor prognosis in patients with sepsis. BMC Immunol 2022; 23:54. [PMID: 36357845 PMCID: PMC9648441 DOI: 10.1186/s12865-022-00528-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 10/20/2022] [Indexed: 11/12/2022] Open
Abstract
Background Sepsis still remains a major challenge in intensive care medicine with unacceptably high mortality among patients with septic shock. Due to current limitations of human CD19+CD24hiCD38hi Breg cells (Bregs) studies among sepsis, here, we tried to evaluate Bregs in severity and prognostic value in patients with sepsis. Methods Peripheral blood from 58 patients with sepsis and 22 healthy controls was analyzed using flow cytometry to evaluate the frequency and number of Bregs. All cases were divided into non-survived or survived group after 28 days followed up. Spearman's correlation analysis was performed on Bregs frequency and clinical indices. The area under the curve was acquired using the receiver operating characteristic analysis to assess the sensitivity and specificity of Bregs for outcome of sepsis. Survival curve analysis and binary logistic regression were applied to estimate the value of Bregs in prognosis among cases with sepsis. Results Sepsis patients had decreased proportions and number of Bregs. Sepsis patients with low frequency of Bregs were associated with an increased risk of septic shock. Bregs frequency is inversely associated with lactate, SOFA, and APACHE II and positively correlated with Tregs frequency. Low levels of Bregs closely correlated with septic outcomes. Numbers of Bregs were prediction factors for poor prognosis. Conclusions Frequency and number of Bregs decreased, and Bregs deficiency revealed poor prognosis in patients with sepsis. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-022-00528-x.
Collapse
Affiliation(s)
- Chunmei Wang
- grid.89957.3a0000 0000 9255 8984Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Nanjing Medical University, Nanjing, 211166 Jiangsu Province China ,grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Huihui Xu
- grid.9227.e0000000119573309Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Rui Gao
- grid.452252.60000 0004 8342 692XDepartment of Respiratory and Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, 272067 Shandong Province China
| | - Fengying Leng
- grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Fangjie Huo
- Department of Respiratory Medicine, Xi’an No. 4 Hospital, Xi’an, 710004 Shanxi Province China
| | - Yinzhen Li
- grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China ,grid.24516.340000000123704535Medical School, Tongji University, Shanghai, 200120 China
| | - Siting Liu
- grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Mingzheng Xu
- grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Jianwen Bai
- grid.89957.3a0000 0000 9255 8984Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Nanjing Medical University, Nanjing, 211166 Jiangsu Province China ,grid.24516.340000000123704535Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| |
Collapse
|
11
|
Wang Y, Deng W, Liu J, Yang Q, Chen Z, Su J, Xu J, Liang Q, Li T, Liu L, Li X. IKKβ increases neuropilin-2 and promotes the inhibitory function of CD9+ Bregs to control allergic diseases. Pharmacol Res 2022; 185:106517. [DOI: 10.1016/j.phrs.2022.106517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 10/31/2022]
|
12
|
Lin F, You H, Cao X, Li T, Hong X, Yang J, Huo P, Li J, Liu W, Jiang Y. Characterization of IL-10-producing regulatory B cells in thymoma. Autoimmunity 2022; 55:351-359. [PMID: 35766145 DOI: 10.1080/08916934.2022.2093862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Regulatory B cells (Bregs) are a subset of B cells that secrete interleukin 10 (IL-10) and play a vital role in suppressing the immune response. The aim of this study was to evaluate the proportion of Bregs in patients with thymoma. METHODS The proportions of subgroups of Bregs in 23 patients with thymoma and 15 healthy controls were detected by flow cytometry. The serum IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ, and TNF-α levels of the subjects were measured using a cytometric bead array (CBA). RESULTS The proportions of circulating IL-10+ B cells, IL-10+CD24hiCD38hi Bregs, and IL-10+CD24hiCD27+ Bregs and the serum IL-10 level were significantly higher in patients with thymoma than in the control group and were negatively correlated with the Karnofsky Performance Scale (KPS) score. The serum levels of cytokines IL-2, IL-6, IFN-γ, and TNF-α were higher and serum IL-17A level was lower in patients with thymoma. Patients with advanced-stage thymoma exhibited significantly higher proportions of IL-10-producing Bregs and a higher serum IL-10 level. After tumour resection, the frequency of circulating IL-10+CD24hiCD38hi Bregs and the serum IL-10 level were significantly decreased in patients with thymoma. The serum IL-10 levels exhibited the best accuracy in assessing the risk of thymoma occurrence in this study. CONCLUSIONS The expression of IL-10 produced by Bregs is increased in patients with thymoma, particularly those with advanced-stage disease, which may suggest that Bregs are involved in the pathogenesis and progression of thymoma.
Collapse
Affiliation(s)
- Fangnan Lin
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Hailong You
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Xiwen Cao
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Tingting Li
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Hong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jinli Yang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Peng Huo
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Wu M, Yu S, Chen Y, Meng W, Chen H, He J, Shen J, Lin X. Acteoside promotes B cell-derived IL-10 production and ameliorates autoimmunity. J Leukoc Biol 2022; 112:875-885. [PMID: 35638582 DOI: 10.1002/jlb.3ma0422-510r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/11/2022] [Indexed: 12/21/2022] Open
Abstract
IL-10-producing regulatory B (Breg) cells are well recognized for maintaining immune tolerance. The impaired Breg cell function with decreased IL-10-producing capacity has been found in autoimmune diseases, such as rheumatoid arthritis, lupus, and primary Sjogren's syndrome (pSS). However, seldom therapeutic agents targeting Breg cells are available to treat those autoimmune diseases. Here, we showed that acteoside (AC), a caffeoyl phenylethanoid glycoside from a medicinal herb Radix Rehmanniae, could promote IL-10 production from both human and murine B cells via critically regulating the TLR4/PI3K axis. Moreover, TLR4 was found increased in Breg cells from mice with experimental SS (ESS), a mouse model that recapitulates human pSS. Thus, B cells from the ESS mice were susceptible to AC treatment, showing higher IL-10-producing capacity than those from naïve controls. In addition, AC treatment also promoted the production of IL-10 from TLR4+ CXCR4+ plasma cells of ESS mice. Notably, we found that AC was able to enter lymphoid organs upon oral administration. AC treatment effectively increased IL-10+ B cells in ESS mice and ameliorated disease pathology accompanied by reduced T effector cells, including Th17 and T follicular helper cells in the ESS mice. In conclusion, AC could promote Breg cell function and attenuate ESS pathology in vivo, which may be a promising drug candidate for treating pSS and other autoimmune diseases.
Collapse
Affiliation(s)
- Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sulan Yu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yacun Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wei Meng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Workstation for Training and Research (Hong Kong Branch), Distinguished Professor Yu Jin Gynaecology of Chinese Medicine & Integrative Medicine, Hong Kong SAR, China.,Workstation of Zhu Nansun, National Master of Chinese Medicine, Hong Kong Branch of Zhu's School of Gynaecology of Chinese Medicine from Shanghai, Hong Kong SAR, China
| | - Haiyong Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiang Lin
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
14
|
A Comprehensive Investigation into the Distribution of Circulating B Cell Subsets in the Third Trimester of Pregnancy. J Clin Med 2022; 11:jcm11113006. [PMID: 35683395 PMCID: PMC9181443 DOI: 10.3390/jcm11113006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023] Open
Abstract
Maternal B cells play a crucial role in the development and maintenance of pregnancy, due to their humoral activities and regulatory functions. In the study, we investigated the alterations in the distributions of naïve and memory B cell subsets, as well as regulatory B (Breg) cells, in the third trimester of pregnancy. Peripheral blood from 14 healthy pregnant women in the third trimester and 7 healthy non-pregnant women was collected and examined for the frequencies of B cell subsets, including IgD+CD27− naïve, IgD+CD27+ un-switched memory, IgD−CD27+ switched memory, CD38intCD24int mature–naïve, CD38−CD24hi primarily memory and CD38hiCD24hi transitional B cells by flow cytometry. Breg cell subsets were also characterized based on the expression of CD5, CD1d and IL-10. In pregnant women, the proportions of un-switched memory and transitional B cells were significantly decreased. Additionally, the frequencies of both CD5+CD1d+ Breg and IL-10-producing B10 cells were decreased in pregnancy. Changes in the distribution of transitional B cells as well as Breg cells may be crucial contributors for the development of altered maternal immune responses and tolerance needed for the maintenance of normal pregnancy in the third trimester.
Collapse
|
15
|
Stetter N, Hartmann W, Brunn ML, Stanelle-Bertram S, Gabriel G, Breloer M. A Combination of Deworming and Prime-Boost Vaccination Regimen Restores Efficacy of Vaccination Against Influenza in Helminth-Infected Mice. Front Immunol 2022; 12:784141. [PMID: 34992602 PMCID: PMC8724120 DOI: 10.3389/fimmu.2021.784141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/03/2021] [Indexed: 11/17/2022] Open
Abstract
Helminths still infect a quarter of the human population. They manage to establish chronic infections by downmodulating the immune system of their hosts. Consequently, the immune response of helminth-infected individuals to vaccinations may be impaired as well. Here we study the impact of helminth-induced immunomodulation on vaccination efficacy in the mouse system. We have previously shown that an underlying Litomosoides sigmodontis infection reduced the antibody (Ab) response to anti-influenza vaccination in the context of a systemic expansion of type 1 regulatory T cells (Tr1). Most important, vaccine-induced protection from a challenge infection with the 2009 pandemic H1N1 influenza A virus (2009 pH1N1) was impaired in vaccinated, L. sigmodontis-infected mice. Here, we aim at the restoration of vaccination efficacy by drug-induced deworming. Treatment of mice with Flubendazole (FBZ) resulted in elimination of viable L. sigmodontis parasites in the thoracic cavity after two weeks. Simultaneous FBZ-treatment and vaccination did not restore Ab responses or protection in L. sigmodontis-infected mice. Likewise, FBZ-treatment two weeks prior to vaccination did not significantly elevate the influenza-specific Ig response and did not protect mice from a challenge infection with 2009 pH1N1. Analysis of the regulatory T cell compartment revealed that L. sigmodontis-infected and FBZ-treated mice still displayed expanded Tr1 cell populations that may contribute to the sustained suppression of vaccination responses in successfully dewormed mice. To outcompete this sustained immunomodulation in formerly helminth-infected mice, we finally combined the drug-induced deworming with an improved vaccination regimen. Two injections with the non-adjuvanted anti-influenza vaccine Begripal conferred 60% protection while MF59-adjuvanted Fluad conferred 100% protection from a 2009 pH1N1 infection in FBZ-treated, formerly L. sigmodontis-infected mice. Of note, applying this improved prime-boost regimen did not restore protection in untreated L. sigmodontis-infected mice. In summary our findings highlight the risk of failed vaccinations due to helminth infection.
Collapse
Affiliation(s)
- Nadine Stetter
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Wiebke Hartmann
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie-Luise Brunn
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Research Department for Viral Zoonoses - One Health, Leibniz Institute for Experimental Virology Heinrich Pette Institute (HPI), Hamburg, Germany
| | - Gülsah Gabriel
- Research Department for Viral Zoonoses - One Health, Leibniz Institute for Experimental Virology Heinrich Pette Institute (HPI), Hamburg, Germany.,Institute for Virology, University for Veterinary Medicine Hannover, Hannover, Germany
| | - Minka Breloer
- Section for Molecular Biology and Immunology, Helminth-Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,Department for Biology, University Hamburg, Hamburg, Germany
| |
Collapse
|
16
|
The survival and function of IL-10-producing regulatory B cells are negatively controlled by SLAMF5. Nat Commun 2021; 12:1893. [PMID: 33767202 PMCID: PMC7994628 DOI: 10.1038/s41467-021-22230-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 03/01/2021] [Indexed: 12/31/2022] Open
Abstract
B cells have essential functions in multiple sclerosis and in its mouse model, experimental autoimmune encephalomyelitis, both as drivers and suppressors of the disease. The suppressive effects are driven by a regulatory B cell (Breg) population that functions, primarily but not exclusively, via the production of IL-10. However, the mechanisms modulating IL-10-producing Breg abundance are poorly understood. Here we identify SLAMF5 for controlling IL-10+ Breg maintenance and function. In EAE, the deficiency of SLAMF5 in B cells causes accumulation of IL10+ Bregs in the central nervous system and periphery. Blocking SLAMF5 in vitro induces both human and mouse IL-10-producing Breg cells and increases their survival with a concomitant increase of a transcription factor, c-Maf. Finally, in vivo SLAMF5 blocking in EAE elevates IL-10+ Breg levels and ameliorates disease severity. Our results suggest that SLAMF5 is a negative moderator of IL-10+ Breg cells, and may serve as a therapeutic target in MS and other autoimmune diseases. Regulatory B (Breg) cells suppress excessive inflammation primary via the production of interleukin 10 (IL-10). Here the authors show that the function and homeostasis of mouse and human IL-10+ Breg cells are negatively regulated by the cell surface receptor, SLAMF5, to impact experimental autoimmunity, thereby hinting SLAMF5 as a potential target for immunotherapy.
Collapse
|
17
|
Chesneau M, Le Mai H, Brouard S. New Method for the Expansion of Highly Purified Human Regulatory Granzyme B-Expressing B Cells. Methods Mol Biol 2021; 2270:203-216. [PMID: 33479900 DOI: 10.1007/978-1-0716-1237-8_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Granzyme B (GZMB)-expressing B cells inhibit CD4+ T-lymphocyte proliferation in a contact- and GZMB-dependent manner, through degradation of TCR zeta or induction of T-cell apoptosis. This regulatory B-cell population is present in human healthy individuals and represents about 1% of circulating B cells. Their small proportion requires the development of expansion methods to enable their study and envision clinical applications. We describe here how to expand GZMB-expressing B cells to obtain more than 90% of highly purified GZMB+ B cells, and the protocol of B/T cells coculture for the evaluation of the suppressive function of the GZMB+ B-cell population.
Collapse
Affiliation(s)
- Mélanie Chesneau
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| | - Hoa Le Mai
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| | - Sophie Brouard
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France. .,Labex IGO, Nantes, France. .,Centre d'Investigation Clinique en Biothérapie, Centre de ressources biologiques (CRB), Nantes, France.
| |
Collapse
|
18
|
Yan L, Qu S, Shang J, Shi X, Kang L, Xu N, Zhu M, Zhou J, Jin S, Yao W, Yao Y, Chen G, Chang H, Zhu X, Yu L, Wu D, Fu C. Sequential CD19 and BCMA-specific CAR T-cell treatment elicits sustained remission of relapsed and/or refractory myeloma. Cancer Med 2020; 10:563-574. [PMID: 33356013 PMCID: PMC7877347 DOI: 10.1002/cam4.3624] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The low rate of durable response against relapsed and/or refractory multiple myeloma (RRMM) in recent studies indicates that chimeric antigen receptor T‐cell (CART) treatment is yet to be optimized. This study aims to investigate the safety and efficacy of sequential infusion of CD19‐CART and B‐cell maturation antigen (BCMA)‐CARTs for RRMM with a similar 3 + 3 dose escalation combined with a toxicity sentinel design. We enrolled 10 patients, among whom 7 received autologous infusion and 3 received allogeneic infusion. The median follow‐up time was 20 months. The most common grade 3/4 treatment‐emergent toxicities were hematological toxicities. Cytokine‐release syndrome (CRS) adverse reactions were grade 1/2 in 9 out of 10 subjects. No dose‐limited toxicity (DLT) was observed for BCMA‐CAR‐positive T cells ≤5 × 107/kg), while two patients with dose‐levels of 5–6.5 × 107/kg experienced DLTs. The overall response rate was 90% (five partial responses and four stringent complete responses). Three out of four patients with stringent complete responses to autologous CART had progression‐free survival for over 2 years. The three patients with allogeneic CART experienced disease progression within 2 months. These results evidence the sequential infusion's preliminarily tolerability and efficacy in RRMM, and present a simple and safe design applicable for the establishment of multiple CART therapy.
Collapse
Affiliation(s)
- Lingzhi Yan
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Su Qu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China
| | - Jingjing Shang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaolan Shi
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Liqing Kang
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China.,Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Nan Xu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China.,Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Mingqing Zhu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jin Zhou
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Song Jin
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Weiqin Yao
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ying Yao
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Guanghua Chen
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huirong Chang
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaming Zhu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Yu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China.,Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Depei Wu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chengcheng Fu
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Hartmann W, Brunn ML, Stetter N, Gagliani N, Muscate F, Stanelle-Bertram S, Gabriel G, Breloer M. Helminth Infections Suppress the Efficacy of Vaccination against Seasonal Influenza. Cell Rep 2020; 29:2243-2256.e4. [PMID: 31747598 DOI: 10.1016/j.celrep.2019.10.051] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/29/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Helminth parasites infect more than a quarter of the human population and inflict significant changes to the immunological status of their hosts. Here, we analyze the impact of helminth infections on the efficacy of vaccinations using Litomosoides sigmodontis-infected mice. Concurrent helminth infection reduces the quantity and quality of antibody responses to vaccination against seasonal influenza. Vaccination-induced protection against challenge infections with the human pathogenic 2009 pandemic H1N1 influenza A virus is drastically impaired in helminth-infected mice. Impaired responses are also observed if vaccinations are performed after clearance of a previous helminth infection, suggesting that individuals in helminth-endemic areas may not always benefit from vaccinations, even in the absence of an acute and diagnosable helminth infection. Mechanistically, the suppression is associated with a systemic and sustained expansion of interleukin (IL)-10-producing CD4+CD49+LAG-3+ type 1 regulatory T cells and partially abrogated by in vivo blockade of the IL-10 receptor.
Collapse
Affiliation(s)
- Wiebke Hartmann
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Marie-Luise Brunn
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Nadine Stetter
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Nicola Gagliani
- I Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, 17176 Stockholm, Sweden
| | - Franziska Muscate
- I Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Research Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Gülsah Gabriel
- Research Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany; Institute for Virology, University for Veterinary Medicine, Hannover, Germany
| | - Minka Breloer
- Section for Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
| |
Collapse
|
20
|
Chesneau M, Mai HL, Danger R, Le Bot S, Nguyen TVH, Bernard J, Poullaouec C, Guerrif P, Conchon S, Giral M, Charreau B, Degauque N, Brouard S. Efficient Expansion of Human Granzyme B–Expressing B Cells with Potent Regulatory Properties. THE JOURNAL OF IMMUNOLOGY 2020; 205:2391-2401. [DOI: 10.4049/jimmunol.2000335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/10/2020] [Indexed: 01/12/2023]
|
21
|
Qin J, Zhao N, Wang S, Liu S, Liu Y, Cui X, Wang S, Xiang Y, Fan C, Li Y, Shan Z, Teng W. Roles of Endogenous IL-10 and IL-10-Competent and CD5+ B Cells in Autoimmune Thyroiditis in NOD.H-2h4 Mice. Endocrinology 2020; 161:5802318. [PMID: 32152630 DOI: 10.1210/endocr/bqaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022]
Abstract
Interleukin (IL)-10 is a highly important anti-inflammatory cytokine in the immune system. CD1dhi and CD5+ B cells are both traditionally defined IL-10-secreting B cells. In recent years, a B cell group with combined markers of CD1dhi and CD5+ has been widely studied as it has been reported to suppress autoimmunity in mouse models of autoimmune diseases through IL-10 mechanisms. From the perspective of origination, CD1dhi and CD5+ B cells are developed from different B cell lineages. Whether the regulatory capacity of these 2 B cell groups is consistent with their ability to secrete IL-10 has not been determined. In this study, we generated IL-10 knockout NOD.H-2h4 mice to investigate the function of endogenous IL-10 in autoimmune thyroiditis and conducted adoptive transfer experiments to explore the respective roles of CD5+ and CD1dhi B cells. In our results, the IL-10-/- NOD.H-2h4 mice developed thyroiditis, similar to wild-type NOD.H-2h4 mice. The CD5+ B cells were more capable of secreting IL-10 than CD1dhi B cells in flow cytometric analysis, but the CD1dhi B cells showed more suppressive effects on thyroiditis development and autoantibody production, as well as Th17 cell response. In conclusion, endogenous IL-10 does not play an important role in autoimmune thyroiditis. CD1dhi B cells may play regulatory roles through mechanisms other than secreting IL-10.
Collapse
Affiliation(s)
- Jing Qin
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Zhao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuo Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shanshan Liu
- Department of Emergency, The NO.202 Hospital of People's Liberation Army, Shenyang, Liaoning, China
| | - Yongping Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejiao Cui
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shiwei Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Xiang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chenling Fan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yushu Li
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
22
|
Xiao J, Guan F, Sun L, Zhang Y, Zhang X, Lu S, Liu W. B cells induced by Schistosoma japonicum infection display diverse regulatory phenotypes and modulate CD4 + T cell response. Parasit Vectors 2020; 13:147. [PMID: 32197642 PMCID: PMC7082913 DOI: 10.1186/s13071-020-04015-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background The increased activity of regulatory B cells (Breg) is known to be involved in immunosuppression during helminth infection, which is characterized by inducing IL-10-producing Breg cells. However, the current knowledge of B cell subsets differentiation and IL-10-independent immunoregulatory mechanisms of B cells in schistosomiasis is insufficient. Methods BALB/c mice were percutaneously infected with cercariae for investigating the profile of B cell subsets during Schistosoma japonicum infection. B cells isolated from the spleen or peritoneal cavity were analyzed for the regulatory phenotype after stimulation with soluble egg antigens (SEA) in vitro. CD4+ T cells were then cocultured with B cells pretreated with or without anti-PD-L1 antibody for investigating the role of B cells from infected mice on regulating CD4+ T cells. Furthermore, the in vivo administration of anti-PD-L1 antibody was conducted to investigate the role of PD-L1 in regulating host immunity during infection. Results The percentages of peritoneal and splenic B-1a cells, as well as marginal zone B (MZB) cells were decreased at eight and twelve weeks after infection compared to those from uninfected mice. In splenic B cells, TGF-β expression was increased at eight weeks but declined at twelve weeks of infection, and PD-L1 expression was elevated at both eight and twelve weeks of infection. In addition, SEA stimulation in vitro significantly promoted the expression of IL-10 in peritoneal B cells and CD5 in splenic B cells, and the SEA-stimulated splenic and peritoneal B cells preferentially expressed PD-L1 and TGF-β. The splenic B cells from infected mice were able to suppress the function of Th1 and Th2 cells in vitro but to expand the expression of Tfh transcription factor Bcl6, which was further enhanced by blocking PD-L1 of B cells before co-cultivation. Moreover, Th2 response and Bcl6 expression in CD4+ T cells were also increased in vivo by blocking PD-L1 after infection, although the hepatic pathology was slightly influenced. Conclusions Our findings revealed that S. japonicum infection modulates the differentiation of B cell subsets that have the capability to affect the CD4+ T cell response. This study contributes to a better understanding of B cells immune response during schistosomiasis.![]()
Collapse
Affiliation(s)
- Junli Xiao
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Guan
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Sun
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijie Zhang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Zhang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengjun Lu
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenqi Liu
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Gutiérrez C, Lopez-Abente J, Pérez-Fernández V, Prieto-Sánchez A, Correa-Rocha R, Moreno-Guillen S, Muñoz-Fernández MÁ, Pion M. Analysis of the dysregulation between regulatory B and T cells (Breg and Treg) in human immunodeficiency virus (HIV)-infected patients. PLoS One 2019; 14:e0213744. [PMID: 30917149 PMCID: PMC6436717 DOI: 10.1371/journal.pone.0213744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/27/2019] [Indexed: 12/21/2022] Open
Abstract
This study examines the relationship between regulatory B (Breg) and T (Treg) compartments, which play crucial roles in the maintenance of immune homeostasis in the context of HIV. Using flow cytometry, the phenotypes of different Breg and Treg subsets from HIV-infected and healthy individuals were analyzed, along with the suppressive capacity of Breg. Peripheral blood samples of thirteen HIV+ treatment-naïve individuals, fourteen treated-HIV+ individuals with undetectable viral load and twelve healthy individuals were analyzed. The absolute counts of Breg and Treg subsets were decreased in HIV+ treatment-naïve individuals in comparison to treated-HIV+ and healthy individuals. Interestingly, correlations between Breg subsets (CD24hiCD27+ and PD-L1+ B cells) and IL-10-producing Breg observed in healthy individuals were lost in HIV+ treatment-naïve individuals. However, a correlation between frequencies of CD24hiCD38hi or TIM-1+-Breg subsets and Treg was observed in HIV+ treatment-naïve individuals and not in healthy individuals. Therefore, we hypothesized that various Breg subsets might have different functions during B and T-cell homeostasis during HIV-1 infection. In parallel, stimulated Breg from HIV-infected treatment-naïve individuals presented a decreased ability to suppress CD4+ T-cell proliferation in comparison to the stimulated Breg from treated-HIV+ or healthy individuals. We demonstrate a dysregulation between Breg and Treg subsets in HIV-infected individuals, which might participate in the hyper-activation and exhaustion of the immune system that occurs in such patients.
Collapse
Affiliation(s)
- Carolina Gutiérrez
- Molecular Immunovirology Laboratory, Department of Infectious Diseases, Ramón y Cajal Health Research Institute (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - Jacobo Lopez-Abente
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Madrid, Spain
| | - Verónica Pérez-Fernández
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Madrid, Spain
| | - Adrián Prieto-Sánchez
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Madrid, Spain
| | - Rafael Correa-Rocha
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Madrid, Spain
| | - Santiago Moreno-Guillen
- Molecular Immunovirology Laboratory, Department of Infectious Diseases, Ramón y Cajal Health Research Institute (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - María-Ángeles Muñoz-Fernández
- Molecular ImmunoBiology Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Spanish HIV HGM BioBank, Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Marjorie Pion
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Madrid, Spain
| |
Collapse
|
24
|
Sanchez LR, Godoy GJ, Gorosito Serrán M, Breser ML, Fiocca Vernengo F, Engel P, Motrich RD, Gruppi A, Rivero VE. IL-10 Producing B Cells Dampen Protective T Cell Response and Allow Chlamydia muridarum Infection of the Male Genital Tract. Front Immunol 2019; 10:356. [PMID: 30881362 PMCID: PMC6405527 DOI: 10.3389/fimmu.2019.00356] [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: 12/27/2018] [Accepted: 02/12/2019] [Indexed: 12/26/2022] Open
Abstract
A significant proportion of individuals develop chronic, persistent and recurrent genital tract infections with Chlamydia trachomatis, which has been attributed to the numerous strategies that the bacterium uses to subvert host immune responses. Animal chlamydia models have demonstrated that protective immune response is mediated by CD4+ Th1 cytokine responses. Herein, we demonstrate that early after infecting the male genital tract, C. muridarum triggers the production of IL-10 by splenic and lymph node cells. In addition, C. muridarum triggers IL-6 and TNFα secretion. Data obtained from in vitro and in vivo experiments revealed B cells as the major IL-10 contributors. Indeed, purified B cells produced high amounts of IL-10 and also exhibited enhanced expression of inhibitory molecules such as CD39, PD-L1 and PD1 after C. muridarum stimulation. In vitro experiments performed with sorted cell subsets revealed that Marginal Zone B cells were the main IL-10 producers. In vitro and in vivo studies using TLR-deficient mice indicated that TLR4 signaling pathway was essential for IL-10 production. In addition, in vivo treatments to neutralize IL-10 or deplete B cells indicated that IL-10 and B cells played a significant role in delaying bacterial clearance ability. Moreover, the latter was confirmed by adoptive cell transfer experiments in which the absence of IL-10-producing B cells conferred the host a greater capability to induce Th1 responses and clear the infection. Interestingly, NOD mice, which were the least efficient in clearing the infection, presented much more Marginal Zone B counts and also enhanced TLR4 expression on Marginal Zone B cells when compared to B6 and BALB/c mice. Besides, treatment with antibodies that selectively deplete Marginal Zone B cells rendered mice more capable of inducing enhanced IFNγ responses and clearing the infection. Our findings suggest that B cells play a detrimental role in C. muridarum infection and that activation by innate receptors like TLR4 and IL-10 production by these cells could be used by Chlamydia spp. as a strategy to modulate the immune response establishing chronic infections in susceptible hosts.
Collapse
Affiliation(s)
- Leonardo R Sanchez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gloria J Godoy
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Melisa Gorosito Serrán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria L Breser
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Facundo Fiocca Vernengo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Ruben D Motrich
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Virginia E Rivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
25
|
Esteve-Solé A, Luo Y, Vlagea A, Deyà-Martínez Á, Yagüe J, Plaza-Martín AM, Juan M, Alsina L. B Regulatory Cells: Players in Pregnancy and Early Life. Int J Mol Sci 2018; 19:ijms19072099. [PMID: 30029515 PMCID: PMC6073150 DOI: 10.3390/ijms19072099] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/17/2022] Open
Abstract
Pregnancy and early infancy represent two very particular immunological states. During pregnancy, the haploidentical fetus and the pregnant women develop tolerance mechanisms to avoid rejection; then, just after birth, the neonatal immune system must modulate the transition from the virtually sterile but haploidentical uterus to a world full of antigens and the rapid microbial colonization of the mucosa. B regulatory (Breg) cells are a recently discovered B cell subset thought to play a pivotal role in different conditions such as chronic infections, autoimmunity, cancer, and transplantation among others in addition to pregnancy. This review focuses on the role of Breg cells in pregnancy and early infancy, two special stages of life in which recent studies have positioned Breg cells as important players.
Collapse
Affiliation(s)
- Ana Esteve-Solé
- Functional Unit of Clinical Immunology and Primary Immunodeficiencies, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain.
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Yiyi Luo
- Functional Unit of Clinical Immunology and Primary Immunodeficiencies, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain.
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Alexandru Vlagea
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
- Immunology Service, Biomedic Diagnostic Center, Hospital Clínic de Barcelona, Universitat de Barcelona, IDIBAPS, 08036 Barcelona, Spain.
| | - Ángela Deyà-Martínez
- Functional Unit of Clinical Immunology and Primary Immunodeficiencies, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain.
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Jordi Yagüe
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
- Immunology Service, Biomedic Diagnostic Center, Hospital Clínic de Barcelona, Universitat de Barcelona, IDIBAPS, 08036 Barcelona, Spain.
| | - Ana María Plaza-Martín
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
- Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain.
| | - Manel Juan
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
- Immunology Service, Biomedic Diagnostic Center, Hospital Clínic de Barcelona, Universitat de Barcelona, IDIBAPS, 08036 Barcelona, Spain.
| | - Laia Alsina
- Functional Unit of Clinical Immunology and Primary Immunodeficiencies, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, University of Barcelona, Pediatric Research Institute Sant Joan de Déu, 08950 Barcelona, Spain.
- Functional Unit of Clinical Immunology, Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain.
| |
Collapse
|
26
|
Gallego-Valle J, Pérez-Fernández VA, Correa-Rocha R, Pion M. Generation of Human Breg-Like Phenotype with Regulatory Function In Vitro with Bacteria-Derived Oligodeoxynucleotides. Int J Mol Sci 2018; 19:ijms19061737. [PMID: 29895745 PMCID: PMC6032322 DOI: 10.3390/ijms19061737] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/08/2018] [Indexed: 11/24/2022] Open
Abstract
Regulatory B cells (Bregs) participate in auto-tolerance maintenance and immune homeostasis. Despite their impact on many diseases and due to the difficulty to define them, knowledge about their origin and their physiological inducers is still unclear. The incomplete understanding about the generation of Bregs and their limited numbers in periphery make it difficult to develop Breg-based therapy. Therefore, identifying factors that promote their development would allow their ex-vivo production in order to create new immunotherapy. This project aims to test the capacity of several cytokines (Interleukin 1-beta (IL-1β), Granulocyte Macrophage Colony-Stimulating Factor (GM-CSF), and Cluster of differentiation 40 ligand (CD40L)) and bacteria-derived oligodeoxynucleotides (CpG-ODN), alone or in combination, to generate B cells with regulatory phenotype and function. We have demonstrated that the Breg-associated phenotypes were heterogeneous between one and other stimulation conditions. However, the expression of other markers related to Bregs such as IL-10, CD80, CD86, CD71, Programmed cell death-1 (PD-1), and Programmed death-ligand 1 (PD-L1) was increased when cells were stimulated with CpG alone or in combination. Moreover, stimulated B cells presented a suppressive function on autologous activated peripheral blood mononuclear cells (PBMC) proliferation. Therefore, this work is the first step to demonstrate the feasibility to induce functional Breg-like cells in vitro and will then facilitate the way to produce Breg-like cells as a potential future cellular therapy.
Collapse
Affiliation(s)
- Jorge Gallego-Valle
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Calle Máiquez, 9, 28009 Madrid, Spain.
| | - Verónica Astrid Pérez-Fernández
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Calle Máiquez, 9, 28009 Madrid, Spain.
| | - Rafael Correa-Rocha
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Calle Máiquez, 9, 28009 Madrid, Spain.
| | - Marjorie Pion
- Immuno-Regulation Laboratory, University General Hospital Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Medicine and Experimental Surgery Building, Calle Máiquez, 9, 28009 Madrid, Spain.
| |
Collapse
|
27
|
Regulatory B cells: the cutting edge of immune tolerance in kidney transplantation. Cell Death Dis 2018; 9:109. [PMID: 29371592 PMCID: PMC5833552 DOI: 10.1038/s41419-017-0152-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 12/20/2022]
Abstract
Kidney transplantation is the optimal treatment for end-stage renal diseases. Although great improvement has been achieved, immune tolerance is still the Holy Grail that every organ transplant practitioner pursues. The role of B cells in transplantation has long been considered simply to serve as precursors of plasma cells, which produce alloantibodies and induce antibody-mediated rejection. Recent research indicates that a specialized subset of B cells plays an important role in immune regulation, which has been well demonstrated in autoimmune diseases, infections, and cancers. This category of regulatory B cells (Bregs) differs from conventional B cells, and they may help develop a novel immunomodulatory therapeutic strategy to achieve immune tolerance in transplantation. Here, we review the latest evidence regarding phenotypes, functions, and effectors of Bregs and discuss their diverse effects on kidney transplantation.
Collapse
|
28
|
Mavropoulos A, Varna A, Zafiriou E, Liaskos C, Alexiou I, Roussaki-Schulze A, Vlychou M, Katsiari C, Bogdanos DP, Sakkas LI. IL-10 producing Bregs are impaired in psoriatic arthritis and psoriasis and inversely correlate with IL-17- and IFNγ-producing T cells. Clin Immunol 2017; 184:33-41. [DOI: 10.1016/j.clim.2017.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/27/2017] [Indexed: 12/24/2022]
|
29
|
Wang K, Tao L, Su J, Zhang Y, Zou B, Wang Y, Zou M, Chen N, Lei L, Li X. TLR4 supports the expansion of FasL +CD5 +CD1d hi regulatory B cells, which decreases in contact hypersensitivity. Mol Immunol 2017; 87:188-199. [PMID: 28505514 DOI: 10.1016/j.molimm.2017.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 02/06/2023]
Abstract
Certain B cells termed as "regulatory B cells" (Bregs) can suppress the ongoing immune responses and a splenic CD5+CD1dhi Breg subset identified earlier was shown to exert its regulatory functions through secretion of IL-10. Though FasL expression is an alternative mechanism of immune suppression used by B cells, little is known about the FasL expressing CD5+CD1dhi Bregs. In this study, we isolated splenocytes or splenic CD19+ B cells and compared the efficiency of toll-like receptor(TLR)4 ligand (lipopolysaccharide) with TLR9 ligand (CpG), anti-CD40 and TLR9 ligand (CpG) plus anti-CD40 on the FasL expression of splenic CD5+CD1dhi Bregs by flow cytometry. FasL expression in CD5+CD1dhi B cells was rapidly increased after TLR4 ligation. Intriguingly, anti-CD40 and CpG plus anti-CD40 combinations failed to stimulate FasL expression in CD5+CD1dhi B cells although the IL-10 production was up-regulated in this subset. In addition, LPS and other B10-cell inducers increased the expression of surface molecules like CD86 and CD25, which are correlated to the regulatory functions of B cells. Furthermore, NF-κB and NF-AT inhibitors decreased the TLR4-activated FasL expression in CD5+CD1dhi B cells. Then we sorted splenic CD5+CD1dhi Bregs using flow cytometry and found that TLR4-activated CD5+CD1dhi Bregs suppressed the proliferation of CFSE-labeled CD4+ T cells in vitro, which was partly blocked by anti-FasL antibody. In oxazolone-sensitized mice having contact hypersensitivity, FasL expression in splenic CD5+CD1dhi B cells was decreased compared to the control group after TLR4 ligation. Our findings suggest that the regulatory function of CD5+CD1dhi B cells could be partly mediated by Fas-FasL pathway and this FasL expressing CD5+CD1dhi Bregs might participate in the regulation of inflammatory diseases.
Collapse
Affiliation(s)
- Keng Wang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Department of Clinical Pharmacy, The Affiliated Nanhai Hospital of Southern Medical University, Foshan 528200, PR China
| | - Lei Tao
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jianbing Su
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Yueyang Zhang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Binhua Zou
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Yiyuan Wang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Min Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Nana Chen
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Linsheng Lei
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaojuan Li
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
30
|
Al-Kufaidy R, Vazquez-Tello A, BaHammam AS, Al-Muhsen S, Hamid Q, Halwani R. IL-17 enhances the migration of B cells during asthma by inducing CXCL13 chemokine production in structural lung cells. J Allergy Clin Immunol 2016; 139:696-699.e5. [PMID: 27639935 DOI: 10.1016/j.jaci.2016.07.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/16/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Roua Al-Kufaidy
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Alejandro Vazquez-Tello
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed S BaHammam
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia; University Sleep Disorders Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saleh Al-Muhsen
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Qutayba Hamid
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada; College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
31
|
|
32
|
Gupta S. Ins and Outs of Antibodies. J Clin Immunol 2016; 36 Suppl 1:1-4. [PMID: 27169867 DOI: 10.1007/s10875-016-0295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Sudhir Gupta
- Division of Basic and Clinical Immunology, University of California at Irvine, Medical Sci. I, C-240, Irvine, CA, 92697, USA.
| |
Collapse
|