1
|
Zhang L, Zhong H, Fan J, Mao J, Li Y. Clinical significance of T helper cell subsets in the peripheral blood and bone marrow of patients with multiple myeloma. Front Immunol 2024; 15:1445530. [PMID: 39324138 PMCID: PMC11422089 DOI: 10.3389/fimmu.2024.1445530] [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: 06/07/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Background T helper (Th) cell subsets primarily assist B cells in differentiating into plasma cells in the germinal center. The mechanism of malignant transformation of plasma cells is an important target for the clinical treatment of MM; however, the mechanism remains unclear. Methods We collected the peripheral blood (PB) and bone marrow (BM) samples of 33 patients with MM. In addition, the PB was also collected from 25 normal healthy controls (HCs). We analyzed the percentages of Th cell subsets in the PB and BM samples of patients with MM. Results Tfh/CD4+ were positively correlated with the proportion of myeloma cells in the BM and PB samples (r = 0.592, P = 0.002 and r = 0.510, P = 0.010 respectively), and showed a strong correlation between the BM and PB samples (r = 0.6559, P = 0.0095). In the PB samples, the percentages of Th2/CD4+ and Tfh2/Tfh cells were significantly lower in patients with MM than in HCs (P = 0.00013 and P = 0.0004, respectively), whereas the percentage of Th17/CD4+ and Tfh17/Tfh was significantly higher in newly diagnosed patients with MM than in HCs (P = 0.0037 and P = 0.03, respectively), and all these cells showed a good predictive value for MM (area under the curve [AUC] 0.781, = 0.792, = 0.837, and 0.723 respectively). In the PB samples, all subsets of PD-1+ICOS- Tfh showed a noticeable downward trend in MM from newly diagnosed to non-remission and remission groups. In contrast, all subsets of PD-1-ICOS+ Tfh increased gradually. Conclusion Th cell subsets play an important role in the occurrence and development of MM and may provide a fundamental basis for identifying new immunotherapy targets and prognosis.
Collapse
Affiliation(s)
- Liangjun Zhang
- Department of Laboratory Medicine, Zigong First People’s Hospital, Zigong, China
| | - Huixiu Zhong
- Department of Laboratory Medicine, Zigong First People’s Hospital, Zigong, China
| | - Jiwen Fan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiansen Mao
- Department of Laboratory Medicine, Nanjing International School, Nanjing, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Mayer DP, Nelson ME, Andriyanova D, Filler RB, Ökten A, Antao OQ, Chen JS, Scumpia PO, Weaver WM, Wilen CB, Deshayes S, Weinstein JS. A novel microporous biomaterial vaccine platform for long-lasting antibody mediated immunity against viral infection. J Control Release 2024; 370:570-582. [PMID: 38734312 DOI: 10.1016/j.jconrel.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Current antigen delivery platforms, such as alum and nanoparticles, are not readily tunable, thus may not generate optimal adaptive immune responses. We created an antigen delivery platform by loading lyophilized Microporous Annealed Particle (MAP) with aqueous solution containing target antigens. Upon administration of antigen loaded MAP (VaxMAP), the biomaterial reconstitution forms an instant antigen-loaded porous scaffold area with a sustained release profile to maximize humoral immunity. VaxMAP induced CD4+ T follicular helper (Tfh) cells and germinal center (GC) B cell responses in the lymph nodes similar to Alum. VaxMAP loaded with SARS-CoV-2 spike protein improved the magnitude, neutralization, and duration of anti-receptor binding domain antibodies compared to Alum vaccinated mice. A single injection of Influenza specific HA1-loaded-VaxMAP enhanced neutralizing antibodies and elicited greater protection against influenza virus challenge than HA1-loaded-Alum. Thus, VaxMAP is a platform that can be used to promote adaptive immune cell responses to generate more robust neutralizing antibodies, and better protection upon pathogen challenge.
Collapse
Affiliation(s)
- Daniel P Mayer
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Mariah E Nelson
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Daria Andriyanova
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Renata B Filler
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Arya Ökten
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Olivia Q Antao
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Jennifer S Chen
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Philip O Scumpia
- Department of Medicine, Division of Dermatology, University of California Los Angeles, Los Angeles, California, United States of America; Department of Dermatology, West Los Angeles Veteran Affairs Medical Center, Los Angeles, California, United States of America
| | - Westbrook M Weaver
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Craig B Wilen
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Stephanie Deshayes
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Jason S Weinstein
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America.
| |
Collapse
|
3
|
Liu Y, Ren S, Ma L, Lin X, Lu J, Cao Z, Zheng S, Hu Z, Xu X, Chen X. Peg-IFNα combined with hepatitis B vaccination contributes to HBsAg seroconversion and improved immune function. Virol J 2024; 21:77. [PMID: 38555445 PMCID: PMC10981809 DOI: 10.1186/s12985-024-02344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/17/2024] [Indexed: 04/02/2024] Open
Abstract
PURPOSE The purpose of this study was to investigate immunological variations between a group that received the hepatitis B vaccine and a non-vaccine group. We focused on a cohort that achieved HBsAg seroclearance after Peg-IFNα treatment of CHB. METHODS We enrolled twenty-eight individuals who achieved HBsAg seroclearance after Peg-IFNα treatment. They were divided into two groups: a vaccine group (n = 14) and a non-vaccine group (n = 14). We assessed lymphocyte subpopulations, B cell- and T cell-surface costimulatory/inhibitory factors, cytokines and immunoglobulin levels were detected at different time points to explore immune-function differences between both groups. RESULTS The seroconversion rate in the vaccine group at 24 weeks post-vaccination was 100%, which was significantly higher (p = 0.006) than that of the non-vaccine group (50%). Additionally, more individuals in the vaccine group exhibited anti-HBs levels exceeding 100 IUs/L and 300 IUs/L compared to the non-vaccine group (p < 0.05). The vaccine group demonstrated significantly increase total B cells and class-switched B cells at 24 weeks and plasma cells, CD80+B cells, Tfh cells, and ICOS+Tfh cell at 12 weeks, compared with baseline levels (p < 0.05). Conversely, Bregs (CD24+CD27+ and CD24+CD38high) decreased significantly at 24 weeks (p < 0.05). None of the above changes were statistically significance in the non-vaccine group (p > 0.05). Total IgG increased significantly in the vaccine group, and IL-2, IL-5, and IL-6 concentrations increased significantly at week 24 (p < 0.05). Differences in various types of cytokines and immunoglobulins in the plasma of the non-vaccine group were not significant (p > 0.05). Anti-HBs titers positively correlated with Th1/Th2 cells at 24 weeks (r = 0.448 and 0.458, respectively, p = 0.022 and 0.019, respectively), and negatively with CD24+CD38highBreg cells (r = -0.402, p = 0.042). CONCLUSIONS After achieving HBsAg seroclearance through Peg-IFNα treatment for CHB, administering the hepatitis B vaccine significantly increased anti-HBs-seroconversion rates and antibody levels. We also observed significant immunological differences between the vaccine and non-vaccine groups. Specifically, the vaccine group exhibited significant increases in B cells, plasma cells, and Tfh cells, while Breg levels was significantly lower. These immunological changes are likely conducive to the production of anti-HBs antibodies. However, in the non-vaccine group, the observed changes were not significantlly significant.
Collapse
Affiliation(s)
- Yisi Liu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Shan Ren
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Lina Ma
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xiao Lin
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Junfeng Lu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Zhenhuan Cao
- Third Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Sujun Zheng
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Zhongjie Hu
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xiaoxue Xu
- Department of Core Facility Center, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China
| | - Xinyue Chen
- First Department of Liver Disease Center, Beijing Youan Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen wai, Beijing, 100069, China.
| |
Collapse
|
4
|
Hinrichs AC, Kruize AA, Lafeber FPJG, Leavis HL, van Roon JAG. CCR9/CXCR5 Co-Expressing CD4 T Cells Are Increased in Primary Sjögren's Syndrome and Are Enriched in PD-1/ICOS-Expressing Effector T Cells. Int J Mol Sci 2023; 24:11952. [PMID: 37569326 PMCID: PMC10418442 DOI: 10.3390/ijms241511952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/21/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune disease characterised by B cell hyperactivity. CXCR5+ follicular helper T cells (Tfh), CXCR5-PD-1hi peripheral helper T cells (Tph) and CCR9+ Tfh-like cells have been implicated in driving B cell hyperactivity in pSS; however, their potential overlap has not been evaluated. Our aim was to study the overlap between the two CXCR5- cell subsets and to study their PD-1/ICOS expression compared to "true" CXCR5/PD-1/ICOS-expressing Tfh cells. CXCR5- Tph and CCR9+ Tfh-like cell populations from peripheral blood mononuclear cells of pSS patients and healthy controls (HC) were compared using flow cytometry. PD-1/ICOS expression from these cell subsets was compared to each other and to CXCR5+ Tfh cells, taking into account their differentiation status. CXCR5- Tph cells and CCR9+ Tfh-like cells, both in pSS patients and HC, showed limited overlap. PD-1/ICOS expression was higher in memory cells expressing CXCR5 or CCR9. However, the highest expression was found in CXCR5/CCR9 co-expressing T cells, which are enriched in the circulation of pSS patients. CXCR5- Tph and CCR9+ Tfh-like cells are two distinct cell populations that both are enriched in pSS patients and can drive B cell hyperactivity in pSS. The known upregulated expression of CCL25 and CXCL13, ligands of CCR9 and CXCR5, at pSS inflammatory sites suggests concerted action to facilitate the migration of CXCR5+CCR9+ T cells, which are characterised by the highest frequencies of PD-1/ICOS-positive cells. Hence, these co-expressing effector T cells may significantly contribute to the ongoing immune responses in pSS.
Collapse
Affiliation(s)
- Anneline C. Hinrichs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
| | - Aike A. Kruize
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
| | - Floris P. J. G. Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
| | - Helen L. Leavis
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
| | - Joel A. G. van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, The Netherlands
| |
Collapse
|
5
|
Seth A, Yokokura Y, Choi JY, Shyer JA, Vidyarthi A, Craft J. AP-1-independent NFAT signaling maintains follicular T cell function in infection and autoimmunity. J Exp Med 2023; 220:e20211110. [PMID: 36820828 PMCID: PMC9998660 DOI: 10.1084/jem.20211110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/05/2022] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
Coordinated gene expression programs enable development and function of T cell subsets. Follicular helper T (Tfh) cells coordinate humoral immune responses by providing selective and instructive cues to germinal center B cells. Here, we show that AP-1-independent NFAT gene expression, a program associated with hyporesponsive T cell states like anergy or exhaustion, is also a distinguishing feature of Tfh cells. NFAT signaling in Tfh cells, maintained by NFAT2 autoamplification, is required for their survival. ICOS signaling upregulates Bcl6 and induces an AP-1-independent NFAT program in primary T cells. Using lupus-prone mice, we demonstrate that genetic disruption or pharmacologic inhibition of NFAT signaling specifically impacts Tfh cell maintenance and leads to amelioration of autoantibody production and renal injury. Our data provide important conceptual and therapeutic insights into the signaling mechanisms that regulate Tfh cell development and function.
Collapse
Affiliation(s)
- Abhinav Seth
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yoshiyuki Yokokura
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jin-Young Choi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Justin A. Shyer
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Aurobind Vidyarthi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Joe Craft
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
6
|
Feng H, Zhao Z, Dong C. Adapting to the world: The determination and plasticity of T follicular helper cells. J Allergy Clin Immunol 2022; 150:981-989. [DOI: 10.1016/j.jaci.2022.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022]
|
7
|
The Regulation between CD4+CXCR5+ Follicular Helper T (Tfh) Cells and CD19+CD24hiCD38hi Regulatory B (Breg) Cells in Gastric Cancer. J Immunol Res 2022; 2022:9003902. [PMID: 36339942 PMCID: PMC9629923 DOI: 10.1155/2022/9003902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/22/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose T follicular helper (Tfh) cells and regulatory B (Breg) cells are reported to play essential roles in humoral immunity, especially in inflammation, autoimmune diseases, and cancer. Hence, we sought to investigate the involvement of CXCR5+CD4+ Tfh cells and CD19+CD24hiCD38hi Breg cells in gastric cancer. Methods The blood samples were obtained from 36 gastric cancer patients and 18 healthy individuals. The percentage of Tfh cells (Tfh%) and Breg cells (Breg%) was detected via flow cytometry, while IL-21, IL-10, and CXCL13 levels were examined with ELISA. The association between them and clinical parameters of patients was also assessed. The in vitro Tfh-B cell coculture experiments were performed for six days, and then, Tfh%, Breg%, and cytokines were valued by flow cytometry and ELISA, respectively. Results Tfh%, Breg%, and CXCL13 level were significantly increased among gastric cancer patients. Moreover, higher Tfh% was associated with lymphatic metastasis, patients' worse outcomes and Breg%. Tfh differentiation and CXCL13 were upregulated by cocultured B cells in vitro, while Tfh cells seem to not participate in Breg cell differentiation from B cells. Conclusion Altogether, increased Tfh and Breg cells could be involved in immune suppression in gastric cancer. Moreover, B cell may be a potential regulator for Tfh differentiation, while Tfh cells had no significant effects on the regulation of Breg cells.
Collapse
|
8
|
Tan Z, Wang L, Li X. Composition and regulation of the immune microenvironment of salivary gland in Sjögren’s syndrome. Front Immunol 2022; 13:967304. [PMID: 36177010 PMCID: PMC9513852 DOI: 10.3389/fimmu.2022.967304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Primary Sjögren’s syndrome (pSS) is a systemic autoimmune disease characterized by exocrine gland dysfunction and inflammation. Patients often have dry mouth and dry eye symptoms, which seriously affect their lives. Improving dry mouth and eye symptoms has become a common demand from patients. For this reason, researchers have conducted many studies on external secretory glands. In this paper, we summarize recent studies on the salivary glands of pSS patients from the perspective of the immune microenvironment. These studies showed that hypoxia, senescence, and chronic inflammation are the essential characteristics of the salivary gland immune microenvironment. In the SG of pSS, genes related to lymphocyte chemotaxis, antigen presentation, and lymphocyte activation are upregulated. Interferon (IFN)-related genes, DNA methylation, sRNA downregulation, and mitochondrial-related differentially expressed genes are also involved in forming the immune microenvironment of pSS, while multiple signaling pathways are involved in regulation. We further elucidated the regulation of the salivary gland immune microenvironment in pSS and relevant, targeted treatments.
Collapse
|
9
|
Gonzalez-Visiedo M, Li X, Munoz-Melero M, Kulis MD, Daniell H, Markusic DM. Single-dose AAV vector gene immunotherapy to treat food allergy. Mol Ther Methods Clin Dev 2022; 26:309-322. [PMID: 35990748 PMCID: PMC9361215 DOI: 10.1016/j.omtm.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022]
Abstract
Immunotherapies for patients with food allergy have shown some success in limiting allergic responses. However, these approaches require lengthy protocols with repeated allergen dosing and patients can relapse following discontinuation of treatment. The purpose of this study was to test if a single dose of an adeno-associated virus (AAV) vector can safely prevent and treat egg allergy in a mouse model. AAV vectors expressing ovalbumin (OVA) under an ubiquitous or liver-specific promoter were injected prior to or after epicutaneous sensitization with OVA. Mice treated with either AAV8-OVA vector were completely protected from allergy sensitization. These animals had a significant reduction in anaphylaxis mediated by a reduction in OVA-specific IgE titers. In mice with established OVA allergy, allergic responses were mitigated only in mice treated with an AAV8-OVA vector expressing OVA from an ubiquitous promoter. In conclusion, an AAV vector with a liver-specific promoter was more effective for allergy prevention, but higher OVA levels were necessary for reducing symptoms in preexisting allergy. Overall, our AAV gene immunotherapy resulted in an expansion of OVA-specific FoxP3+ CD4+ T cells, an increase in the regulatory cytokine IL-10, and a reduction in the IgE promoting cytokine IL-13.
Collapse
Affiliation(s)
- Miguel Gonzalez-Visiedo
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, R4-155, Indianapolis, IN 46202, USA
| | - Xin Li
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, R4-155, Indianapolis, IN 46202, USA
| | - Maite Munoz-Melero
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, R4-155, Indianapolis, IN 46202, USA
| | - Michael D Kulis
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M Markusic
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, R4-155, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Grydziuszko E, Phelps A, Bruton K, Jordana M, Koenig JFE. Heterogeneity, subsets, and plasticity of T follicular helper cells in allergy. J Allergy Clin Immunol 2022; 150:990-998. [PMID: 36070826 DOI: 10.1016/j.jaci.2022.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 10/14/2022]
Abstract
Antibody responses are critical for protection against pathogens. However, diseases such as allergic rhinitis or food allergy result from aberrant production of IgE antibodies against otherwise innocuous environmental antigens. The production of allergen-specific IgE requires interaction between B cells and CD4+ T cells, and a granular understanding of these interactions is required to develop novel therapies for allergic disease. CD4+ T cells are exceptionally heterogeneous in their transcriptional, epigenetic, and proteomic profiles, which poses significant challenges when attempting to define subsets relevant to the study of allergy among a continuum of cells. Defining subsets such as the T follicular helper (TFH) cell cluster provides a shorthand to understand the functions of CD4+ T cells in antibody production and supports mechanistic experimentation for hypothesis-driven discovery. With a focus on allergic disease, this Rostrum article broadly discusses heterogeneity among CD4+ T cells and provides a rationale for subdividing TFH cells into both functional and cytokine-skewed subsets. Further, it highlights the plasticity demonstrated by TFH cells during the primary response and after recall, and it explores the possibility of harnessing this plasticity to reprogram immunity for therapeutic benefit in allergic disease.
Collapse
Affiliation(s)
- Emily Grydziuszko
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Allyssa Phelps
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Kelly Bruton
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Manel Jordana
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Joshua F E Koenig
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
11
|
Lombard‐Vadnais F, Lacombe J, Chabot‐Roy G, Ferron M, Lesage S. OCA‐B does not act as a transcriptional coactivator in T cells. Immunol Cell Biol 2022; 100:338-351. [DOI: 10.1111/imcb.12543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/14/2022] [Accepted: 03/09/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Félix Lombard‐Vadnais
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
- Department of Microbiology & Immunology McGill University Montreal QC H3A 0G4 Canada
| | - Julie Lacombe
- Molecular Physiology Research Unit Institut de recherches cliniques de Montréal Montréal QC H2W 1R7 Canada
| | - Geneviève Chabot‐Roy
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit Institut de recherches cliniques de Montréal Montréal QC H2W 1R7 Canada
- Département de médecine Université de Montréal Montréal QC H3T 1J4 Canada
- Division of Experimental Medicine McGill University Montreal QC H3A 0G4 Canada
| | - Sylvie Lesage
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
- Département de microbiologie, infectiologie et immunologie Université de Montréal Montréal QC H3T 1J4 Canada
| |
Collapse
|
12
|
O’Neal KA, Latham LE, Ntirandekura E, Foscue CL, Stumhofer JS. ICOS Expression Is Required for Maintenance but Not the Formation of Germinal Centers in the Spleen in Response to Plasmodium yoelii Infection. Infect Immun 2022; 90:e0046821. [PMID: 35007126 PMCID: PMC8929343 DOI: 10.1128/iai.00468-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
Inducible T cell costimulator (ICOS) plays a key role in the differentiation and maintenance of follicular helper T (Tfh) cells and, thus, germinal center (GC) formation. Previously, our laboratory showed in a Plasmodium chabaudi infection model that Icos-/- mice were significantly impaired in their ability to form GCs despite persistent infection and, thus, a continued antigen (Ag) load. Here, we show that the resolution of primary infection with Plasmodium yoelii was delayed in Icos-/- mice. This phenotype was associated with a reduction in the accumulation of Tfh-like and GC Tfh cells and an early deficiency in Ag-specific antibody (Ab) production. However, Icos-/- mice could form GCs, although they were less frequent in number than in wild-type (WT) mice. Nonetheless, the Ag-specific Abs from Icos-/- mice lacked signs of affinity maturation, suggesting functional defects associated with these GCs. Eventually, these GC structures dissipated more rapidly in Icos-/- mice than in WT mice. Moreover, the ability of Icos-/- mice to form these GC structures is not reliant on the high Ag loads associated with P. yoelii infections, as GC formation was preserved in Icos-/- mice treated with atovaquone. Finally, mice were unable to form secondary GCs in the absence of ICOS after rechallenge. Overall, these data demonstrate the necessity of ICOS in the maintenance of Tfh cells, the formation and maintenance of sufficient numbers of functioning GCs, and the ability to generate new GC structures after reinfection with P. yoelii.
Collapse
Affiliation(s)
- Kara A. O’Neal
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, Little Rock, Arkansas, USA
| | - Leah E. Latham
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, Little Rock, Arkansas, USA
| | - Enatha Ntirandekura
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, Little Rock, Arkansas, USA
| | - Camille L. Foscue
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, Little Rock, Arkansas, USA
| | - Jason S. Stumhofer
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, Little Rock, Arkansas, USA
| |
Collapse
|
13
|
Londregan J, Maslanka J, Goldman N, Somerville J, Riggs JE. IgD ligation allows peritoneal cavity B cell proliferation. Immunobiology 2022; 227:152181. [PMID: 35077917 PMCID: PMC8918009 DOI: 10.1016/j.imbio.2022.152181] [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: 10/27/2021] [Revised: 12/24/2021] [Accepted: 01/17/2022] [Indexed: 11/19/2022]
Abstract
Atypical cytokine production and immune cell subset ratios, particularly those that include high proportions of macrophages, characterize tumor microenvironments (TMEs). TMEs can be modeled by culturing peritoneal cavity (PerC) cells which have a high macrophage to lymphocyte ratio. With TCR or BCR ligation, PerC lymphocyte proliferation is tempered by macrophages. However, PHA (T cells) and anti-CD40 (B cells) are activators that induce proliferation. Herein, we report that ligating IgD, in contrast to IgM, triggers PerC B cell proliferation. IL-4 addition enhanced the IgD response for BALB/c PerC B cells but suppressed that of C57BL/6 mice. Intriguingly, concurrent ligation of IgD and CD3ε rescued a PerC T cell proliferative response. These results serve to expand the list of targets for promoting cellular and humoral immunity in conditions that model macrophage-rich TMEs.
Collapse
Affiliation(s)
| | - Jeffrey Maslanka
- Biology Department, Rider University, Lawrenceville, NJ 08648, USA
| | - Naomi Goldman
- Biology Department, Rider University, Lawrenceville, NJ 08648, USA
| | - John Somerville
- Biology Department, Rider University, Lawrenceville, NJ 08648, USA
| | - James E Riggs
- Biology Department, Rider University, Lawrenceville, NJ 08648, USA.
| |
Collapse
|
14
|
Pontarini E, Coleby R, Bombardieri M. Cellular and molecular diversity in Sjogren's syndrome salivary glands: Towards a better definition of disease subsets. Semin Immunol 2021; 58:101547. [PMID: 34876330 DOI: 10.1016/j.smim.2021.101547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Primary Sjögren's syndrome (pSS) is a highly heterogeneous disease in terms of clinical presentation ranging from a mild disease localised to the salivary and lacrimal glands, to multiorgan complications of various degrees of severity, finishing with the evolution, in around 5% of pSS patients, to B cell lymphomas most commonly arising in the inflamed salivary glands. Currently, there are poor positive or negative predictors of disease evolution able to guide patient management and treatment at early stages of the diseases. Recent understanding of the pathogenic mechanisms driving immunopathology in pSS, particularly through histological and transcriptomic analysis of minor and parotid salivary gland (SG) biopsies, has highlighted a high degree of cellular and molecular heterogeneity of the inflammatory lesions but also allowed the identification of clusters of patients with similar underlying SG immunopathology. In particular, patients presenting with high degrees of B/T cell infiltration and the formation of ectopic lymphoid structures (ELS) in the SG have been associated, albeit with conflicting results, with higher degree of disease severity and enhanced risk of lymphoma evolution, suggesting that a dysregulated adaptive immune response plays a key role in driving disease manifestations in pSS. Recent data from randomised clinical trials with novel biological therapies in pSS have also highlighted the potential role of SG immunopathology and molecular pathology in stratifying patients for trial inclusion as well as assessing proof of mechanisms in longitudinal SG biopsies before and after treatment. Although significant progress has been made in the understanding of disease pathogenesis and heterogeneity through cellular and molecular SG pathology, further work is needed to validate their clinical utility in routine clinical settings and in randomised clinical trials.
Collapse
Affiliation(s)
- Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rachel Coleby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
15
|
Louis K, Macedo C, Metes D. Targeting T Follicular Helper Cells to Control Humoral Allogeneic Immunity. Transplantation 2021; 105:e168-e180. [PMID: 33909968 PMCID: PMC8484368 DOI: 10.1097/tp.0000000000003776] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Humoral allogeneic immunity driven by anti-HLA donor-specific antibodies and antibody-mediated rejection (AMR) significantly impede prolonged survival of organ allografts after transplantation. Although the importance of T follicular helper (TFH) cells in controlling antibody responses has been long established, their role in directing donor-specific antibody generation leading to AMR was only recently appreciated in the clinical setting of organ transplantation. In this review, we provide a comprehensive summary of the current knowledge on the biology of human TFH cells as well as their circulating counterparts and describe their pivotal role in driving humoral alloimmunity. In addition, we discuss the intrinsic effects of current induction therapies and maintenance immunosuppressive drugs as well as of biotherapies on TFH cells and provide future directions and novel opportunities of biotherapeutic targeting of TFH cells that have the potential of bringing the prophylactic and curative treatments of AMR toward personalized and precision medicine.
Collapse
Affiliation(s)
- Kevin Louis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Human Immunology and Immunopathology, Inserm UMR 976, Université de Paris, Paris, France
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diana Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Steines L, Poth H, Schuster A, Amann K, Banas B, Bergler T. Disruption of Tfh:B Cell Interactions Prevents Antibody-Mediated Rejection in a Kidney Transplant Model in Rats: Impact of Calcineurin Inhibitor Dose. Front Immunol 2021; 12:657894. [PMID: 34135891 PMCID: PMC8201497 DOI: 10.3389/fimmu.2021.657894] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022] Open
Abstract
We aimed to investigate the mechanisms of humoral immune activation in ABMR using a MHC-mismatched rat kidney transplant model. We applied low dose cyclosporine A (loCNI) to allow donor-specific antibody (DSA) formation and rejection and high dose cyclosporine A (hiCNI) for non-rejection. DSA and leukocyte subsets were measured by flow cytometry. Germinal centers (GC), T follicular helper cells (Tfh), plasma cells and interleukin-21 (IL-21) expression were analyzed by immunofluorescence microscopy. Expression of important costimulatory molecules and cytokines was measured by qRT-PCR. Allograft rejection was evaluated by a nephropathologist. We found that DSA formation correlated with GC frequency and expansion, and that GC size was linked to the number of activated Tfh. In hiCNI, GC and activated Tfh were virtually absent, resulting in fewer plasma cells and no DSA or ABMR. Expression of B cell activating T cell cytokine IL-21 was substantially inhibited in hiCNI, but not in loCNI. In addition, hiCNI showed lower expression of ICOS ligand and IL-6, which stimulate Tfh differentiation and maintenance. Overall, Tfh:B cell crosstalk was controlled only by hiCNI treatment, preventing the development of DSA and ABMR. Additional strategies targeting Tfh:B cell interactions are needed for preventing alloantibody formation and ABMR.
Collapse
Affiliation(s)
- Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Helen Poth
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Zhao Q, Dai H, Liu X, Jiang H, Liu W, Feng Z, Zhang N, Gao Y, Dong Z, Zhou X, Du J, Zhang N, Rui H, Yuan L, Liu B. Helper T Cells in Idiopathic Membranous Nephropathy. Front Immunol 2021; 12:665629. [PMID: 34093559 PMCID: PMC8173183 DOI: 10.3389/fimmu.2021.665629] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/06/2021] [Indexed: 01/09/2023] Open
Abstract
Idiopathic membranous nephropathy (IMN) is an autoimmune disease in which the immune system produces an antibody response to its own antigens due to impaired immune tolerance. Although antibodies are derived from plasma cells differentiated by B cells, the T-B cells also contribute a lot to the immune system. In particular, the subsets of helper T (Th) cells, including the dominant subsets such as Th2, Th17, and follicular helper T (Tfh) cells and the inferior subsets such as regulatory T (Treg) cells, shape the immune imbalance of IMN and promote the incidence and development of autoimmune responses. After reviewing the physiological knowledge of various subpopulations of Th cells and combining the existing studies on Th cells in IMN, the role model of Th cells in IMN was explained in this review. Finally, the existing clinical treatment regimens for IMN were reviewed, and the importance of the therapy for Th cells was highlighted.
Collapse
Affiliation(s)
- Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Haoran Dai
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| | - Xianli Liu
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| | - Hanxue Jiang
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Wenbin Liu
- Beijing University of Chinese Medicine, Beijing, China
| | - Zhendong Feng
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China.,Beijing Chinese Medicine Hospital Pinggu Hospital, Beijing, China
| | - Na Zhang
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Yu Gao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Zhaocheng Dong
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhou
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Jieli Du
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Naiqian Zhang
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Hongliang Rui
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Li Yuan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Hart AP, Laufer TM. A review of signaling and transcriptional control in T follicular helper cell differentiation. J Leukoc Biol 2021; 111:173-195. [PMID: 33866600 DOI: 10.1002/jlb.1ri0121-066r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
T follicular helper (Tfh) cells are a critical component of adaptive immunity and assist in optimal Ab-mediated defense. Multiple effector functions of Tfh support germinal center B cell survival, Ab class switching, and plasma cell maturation. In the past 2 decades, the phenotype and functional characteristics of GC Tfh have been clarified allowing for robust studies of the Th subset including activation signals and environmental cues controlling Tfh differentiation and migration during an immune response. A unique, 2-step differentiation process of Tfh has been proposed but the mechanisms underlying transition between unstable Tfh precursors and functional mature Tfh remain elusive. Likewise, newly identified transcriptional regulators of Tfh development have not yet been incorporated into our understanding of how these cells might function in disease. Here, we review the signals and downstream transcription factors that shape Tfh differentiation including what is known about the epigenetic processes that maintain Tfh identity. It is proposed that further evaluation of the stepwise differentiation pattern of Tfh will yield greater insights into how these cells become dysregulated in autoimmunity.
Collapse
Affiliation(s)
- Andrew P Hart
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Terri M Laufer
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Rheumatology, Department of Medicine, Corporal Michael C. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| |
Collapse
|
19
|
Pontarini E, Murray-Brown WJ, Croia C, Lucchesi D, Conway J, Rivellese F, Fossati-Jimack L, Astorri E, Prediletto E, Corsiero E, Romana Delvecchio F, Coleby R, Gelbhardt E, Bono A, Baldini C, Puxeddu I, Ruscitti P, Giacomelli R, Barone F, Fisher B, Bowman SJ, Colafrancesco S, Priori R, Sutcliffe N, Challacombe S, Carlesso G, Tappuni A, Pitzalis C, Bombardieri M. Unique expansion of IL-21+ Tfh and Tph cells under control of ICOS identifies Sjögren's syndrome with ectopic germinal centres and MALT lymphoma. Ann Rheum Dis 2020; 79:1588-1599. [PMID: 32963045 PMCID: PMC7677495 DOI: 10.1136/annrheumdis-2020-217646] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/28/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To explore the relevance of T-follicular-helper (Tfh) and pathogenic peripheral-helper T-cells (Tph) in promoting ectopic lymphoid structures (ELS) and B-cell mucosa-associated lymphoid tissue (MALT) lymphomas (MALT-L) in Sjögren's syndrome (SS) patients. METHODS Salivary gland (SG) biopsies with matched peripheral blood were collected from four centres across the European Union. Transcriptomic (microarray and quantitative PCR) analysis, FACS T-cell immunophenotyping with intracellular cytokine detection, multicolor immune-fluorescence microscopy and in situ hybridisation were performed to characterise lesional and circulating Tfh and Tph-cells. SG-organ cultures were used to investigate functionally the blockade of T-cell costimulatory pathways on key proinflammatory cytokine production. RESULTS Transcriptomic analysis in SG identified Tfh-signature, interleukin-21 (IL-21) and the inducible T-cell co-stimulator (ICOS) costimulatory pathway as the most upregulated genes in ELS+SS patients, with parotid MALT-L displaying a 400-folds increase in IL-21 mRNA. Peripheral CD4+CXC-motif chemokine receptor 5 (CXCR5)+programmed cell death protein 1 (PD1)+ICOS+ Tfh-like cells were significantly expanded in ELS+SS patients, were the main producers of IL-21, and closely correlated with circulating IgG and reduced complement C4. In the SG, lesional CD4+CD45RO+ICOS+PD1+ cells selectively infiltrated ELS+ tissues and were aberrantly expanded in parotid MALT-L. In ELS+SG and MALT-L parotids, conventional CXCR5+CD4+PD1+ICOS+Foxp3- Tfh-cells and a uniquely expanded population of CXCR5-CD4+PD1hiICOS+Foxp3- Tph-cells displayed frequent IL-21/interferon-γ double-production but poor IL-17 expression. Finally, ICOS blockade in ex vivo SG-organ cultures significantly reduced the production of IL-21 and inflammatory cytokines IL-6, IL-8 and tumour necrosis factor-α (TNF-α). CONCLUSIONS Overall, these findings highlight Tfh and Tph-cells, IL-21 and the ICOS costimulatory pathway as key pathogenic players in SS immunopathology and exploitable therapeutic targets in SS.
Collapse
Affiliation(s)
- Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - William James Murray-Brown
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Cristina Croia
- Immuno-Allergology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Davide Lucchesi
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - James Conway
- Oncology R&D, Astrazeneca, Gaithersburg, Maryland, USA
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Elisa Astorri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Edoardo Prediletto
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Elisa Corsiero
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | | | - Rachel Coleby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Eva Gelbhardt
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Aurora Bono
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | | | - Ilaria Puxeddu
- Immuno-Allergology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Ruscitti
- Department of Clinical Sciences and Applied Biotechnology, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Roberto Giacomelli
- Department of Clinical Sciences and Applied Biotechnology, University of L'Aquila, L'Aquila, Abruzzo, Italy
| | - Francesca Barone
- RRG, Institute of Inflamation and Ageing, University of Birmingham, Birmingham, UK, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Benjamin Fisher
- RRG, Institute of Inflamation and Ageing, University of Birmingham, Birmingham, UK, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Simon J Bowman
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Serena Colafrancesco
- Dipartimento di Medicina Interna e Specilità Mediche, UOC Reumatologia, Universita degli Studi di Roma La Sapienza Facolta di Medicina e Odontoiatria, Roma, Lazio, Italy
| | - Roberta Priori
- Dipartimento di Medicina Interna e Specilità Mediche, UOC Reumatologia, Universita degli Studi di Roma La Sapienza Facolta di Medicina e Odontoiatria, Roma, Lazio, Italy
| | | | | | - Gianluca Carlesso
- Early ICA Discovery, Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anwar Tappuni
- Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, London, England, UK
| |
Collapse
|
20
|
Song W, Craft J. T follicular helper cell heterogeneity: Time, space, and function. Immunol Rev 2019; 288:85-96. [PMID: 30874350 DOI: 10.1111/imr.12740] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
T follicular helper (Tfh) cells play a crucial role in orchestrating the humoral arm of adaptive immune responses. Mature Tfh cells localize to follicles in secondary lymphoid organs (SLOs) where they provide help to B cells in germinal centers (GCs) to facilitate immunoglobulin affinity maturation, class-switch recombination, and generation of long-lived plasma cells and memory B cells. Beyond the canonical GC Tfh cells, it has been increasingly appreciated that the Tfh phenotype is highly diverse and dynamic. As naive CD4+ T cells progressively differentiate into Tfh cells, they migrate through a variety of microanatomical locations to obtain signals from other cell types, which in turn alters their phenotypic and functional profiles. We herein review the heterogeneity of Tfh cells marked by the dynamic phenotypic changes accompanying their developmental program. Focusing on the various locations where Tfh and Tfh-like cells are found, we highlight their diverse states of differentiation. Recognition of Tfh cell heterogeneity has important implications for understanding the nature of T helper cell identity specification, especially the plasticity of the Tfh cells and their ontogeny as related to conventional T helper subsets.
Collapse
Affiliation(s)
- Wenzhi Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Joe Craft
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT.,Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
21
|
Mendoza Rojas A, Hesselink DA, van Besouw NM, Baan CC, van Gelder T. Impact of low tacrolimus exposure and high tacrolimus intra-patient variability on the development of de novo anti-HLA donor-specific antibodies in kidney transplant recipients. Expert Rev Clin Immunol 2019; 15:1323-1331. [PMID: 31721605 DOI: 10.1080/1744666x.2020.1693263] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: De novo donor-specific antibodies (dnDSA) directed against HLA are a major contributing factor to the chronic deterioration of renal allograft function. Several factors, including the degree of HLA matching, younger recipient age, and past sensitization events have been shown to increase the risk for the development of dnDSA. The development of dnDSA is also strongly associated with modifications in the immunosuppressive regimen, non-adherence, and under-immunosuppression.Areas covered: Tacrolimus is widely used after solid organ transplantation (SOT) and in recent years, both a high intra-patient variability in tacrolimus exposure and low tacrolimus exposure have been found to be associated with a higher risk of dnDSA development in kidney transplant recipients. This article provides an overview of current findings published in the recent 5 years regarding the relationship between tacrolimus exposure and variation therein and the development of dnDSA.Expert opinion: In this review, we describe how combining data on tacrolimus intra-patient variability and mean pre-dose concentration may be an effective tool to identify kidney transplant recipients who are at higher risk of developing dnDSA.
Collapse
Affiliation(s)
- Aleixandra Mendoza Rojas
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nicole M van Besouw
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Teun van Gelder
- Department of Internal Medicine, Nephrology & Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Hospital Pharmacy, Clinical Pharmacology Unit, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
22
|
Abstract
The importance of B cell and antibody-mediated immune response in the acute and long-term persistence of transplanted solid organs has become increasingly evident in recent years. A variety of therapeutic innovations target antibodies directed toward HLA or blood groups (ABO) to allow better allocation and posttransplant longevity of organs. Antibodies originate from plasma cells (PCs), which are terminally differentiated B cells. Long-term production and persistence of these antibodies is partly due to fast reactivation of previously generated memory B cells; however, there is increasing evidence that some differentiated PCs can persist independently in the bone marrow for years or even decades, producing specific antibodies or even experiencing regeneration without proliferation without need to be replaced by newly differentiating B cells. This review outlines the currently presumed pathways of differentiation, antibody, and memory generation on both B-cell and PC levels. On this background, current therapeutic concepts for antibody reduction before and after solid organ transplantation are considered, to better understand their mechanisms, possible synergisms, and specific risks. Specific differences in regards to ABO versus HLA antibodies as well as practical relevance for generation of desensitization and posttransplant antibody-directed therapy protocols are discussed.
Collapse
|
23
|
Nguyen DC, Joyner CJ, Sanz I, Lee FEH. Factors Affecting Early Antibody Secreting Cell Maturation Into Long-Lived Plasma Cells. Front Immunol 2019; 10:2138. [PMID: 31572364 PMCID: PMC6749102 DOI: 10.3389/fimmu.2019.02138] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Antibody secreting cells (ASCs) are terminally differentiated cells of the humoral immune response and must adapt morphologically, transcriptionally, and metabolically to maintain high-rates of antibody (Ab) secretion. ASCs differentiate from activated B cells in lymph nodes and transiently circulate in the blood. Most of the circulating ASCs undergo apoptosis, but a small fraction of early ASCs migrate to the bone marrow (BM) and eventually mature into long-lived plasma cells (LLPCs). LLPC survival is controlled both intrinsically and extrinsically. Their differentiation and maintenance programs are governed by many intrinsic mechanisms involving anti-apoptosis, autophagy, and metabolism. The extrinsic factors involved in LLPC generation include BM stromal cells, cytokines, and chemokines, such as APRIL, IL-6, and CXCL12. In humans, the BM CD19−CD38hiCD138+ ASC subset is the main repository of LLPCs, and our recent development of an in vitro BM mimic provides essential tools to study environmental cues that support LLPC survival and the critical molecular mechanisms of maturation from early minted blood ASCs to LLPCs. In this review, we summarize the evidence of LLPC generation and maintenance and provide novel paradigms of LLPC maturation.
Collapse
Affiliation(s)
- Doan C Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Chester J Joyner
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Iñaki Sanz
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States.,Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States.,Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States
| |
Collapse
|
24
|
Roe K, Shu GL, Draves KE, Giordano D, Pepper M, Clark EA. Targeting Antigens to CD180 but Not CD40 Programs Immature and Mature B Cell Subsets to Become Efficient APCs. THE JOURNAL OF IMMUNOLOGY 2019; 203:1715-1729. [PMID: 31484732 DOI: 10.4049/jimmunol.1900549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022]
Abstract
Targeting Ags to the CD180 receptor activates both B cells and dendritic cells (DCs) to become potent APCs. After inoculating mice with Ag conjugated to an anti-CD180 Ab, B cell receptors were rapidly internalized. Remarkably, all B cell subsets, including even transitional 1 B cells, were programed to process, present Ag, and stimulate Ag-specific CD4+ T cells. Within 24-48 hours, Ag-specific B cells were detectable at T-B borders in the spleen; there, they proliferated in a T cell-dependent manner and induced the maturation of T follicular helper (TFH) cells. Remarkably, immature B cells were sufficient for the maturation of TFH cells after CD180 targeting: TFH cells were induced in BAFFR-/- mice (with only transitional 1 B cells) and not in μMT mice (lacking all B cells) following CD180 targeting. Unlike CD180 targeting, CD40 targeting only induced DCs but not B cells to become APCs and thus failed to efficiently induce TFH cell maturation, resulting in slower and lower-affinity IgG Ab responses. CD180 targeting induces a unique program in Ag-specific B cells and to our knowledge, is a novel strategy to induce Ag presentation in both DCs and B cells, especially immature B cells and thus has the potential to produce a broad range of Ab specificities. This study highlights the ability of immature B cells to present Ag to and induce the maturation of cognate TFH cells, providing insights toward vaccination of mature B cell-deficient individuals and implications in treating autoimmune disorders.
Collapse
Affiliation(s)
- Kelsey Roe
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Geraldine L Shu
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Kevin E Draves
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Daniela Giordano
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Edward A Clark
- Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
25
|
Van DV, Bauer L, Kroczek RA, Hutloff A. ICOS Costimulation Differentially Affects T Cells in Secondary Lymphoid Organs and Inflamed Tissues. Am J Respir Cell Mol Biol 2019; 59:437-447. [PMID: 29676593 DOI: 10.1165/rcmb.2017-0309oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
B-cell interaction with follicular helper T cells and subsequent differentiation of B cells into high-affinity APCs normally takes place in secondary lymphoid organs. The costimulator ICOS plays a key role in this process and is therefore considered as an attractive target to modulate exaggerated B-cell responses in autoimmune or allergic diseases. Inflamed tissues were recently recognized as additional sites of active T-cell/B-cell interaction. To analyze whether ICOS costimulation is also important there, we employed a mouse airway inflammation model that allows direct comparison of immune reactions in the lung-draining lymph node and the lung tissue as well as assessment of the relative importance of dendritic cells versus B cells as APCs. In both organs, ICOS regulated the pool size of antigen-specific T and B cells and B-cell differentiation into germinal center(-like) cells but not into antibody-secreting cells. In the lymph node, lack of ICOS costimulation drastically reduced the frequency of T follicular helper cells but did not affect production of T-helper cell type 2 (Th2) cytokines. Vice versa in the lung tissue, ICOS did not change PD-1 expression on infiltrating T cells but regulated Th2 cytokine production, a process for which ICOS ligand expression on B cells was of particular importance. Taken together, the results of this study show that ICOS differentially regulates effector T cells in secondary lymphoid organs and inflamed tissues but that blockade of the ICOS pathway is suitable to target T cell-dependent B cell responses at both sites.
Collapse
Affiliation(s)
- Dana Vu Van
- 1 Chronic Immune Reactions, German Rheumatism Research Centre, a Leibniz Institute, Berlin, Germany; and.,2 Molecular Immunology, Robert Koch Institute, Berlin, Germany
| | - Laura Bauer
- 1 Chronic Immune Reactions, German Rheumatism Research Centre, a Leibniz Institute, Berlin, Germany; and.,2 Molecular Immunology, Robert Koch Institute, Berlin, Germany
| | | | - Andreas Hutloff
- 1 Chronic Immune Reactions, German Rheumatism Research Centre, a Leibniz Institute, Berlin, Germany; and.,2 Molecular Immunology, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
26
|
Leibler C, Thiolat A, Elsner RA, El Karoui K, Samson C, Grimbert P. Costimulatory blockade molecules and B-cell-mediated immune response: current knowledge and perspectives. Kidney Int 2019; 95:774-786. [PMID: 30711200 DOI: 10.1016/j.kint.2018.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
There is an urgent need for therapeutic agents that target humoral alloimmunity in solid organ transplantation. This includes sensitized patients with preformed donor-specific human leukocyte antigen antibodies and patients who develop de novo donor-specific antibodies, both of which are associated with acute and chronic antibody-mediated rejection and allograft loss. In the last decade, both experimental and clinical studies highlighted the major impact of costimulation molecules in the control of immune responses both in the field of transplantation and autoimmune disease. Although these molecules have been initially developed to control the early steps of T-cell activation, recent evidence also supports their influence at several steps of the humoral response. In this review, we aim to provide an overview of the current knowledge of the effects of costimulatory blockade agents on humoral responses in both autoimmune and allogeneic contexts. We first present the effects of costimulatory molecules on the different steps of alloantibody production. We then summarize mechanisms and clinical results observed using cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig molecules both in transplantation and autoimmunity. Finally, we present the potential interest and implications of other costimulatory family members as therapeutic targets, with emphasis on combinatorial approaches, for the optimal control of the alloantigen-specific humoral response.
Collapse
Affiliation(s)
- Claire Leibler
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Allan Thiolat
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Rebecca A Elsner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Chloe Samson
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Philippe Grimbert
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France.
| |
Collapse
|
27
|
Chen M, Lin X, Olsen N, He X, Zheng SG. Advances in T follicular helper and T follicular regulatory cells in transplantation immunity. Transplant Rev (Orlando) 2018; 32:187-193. [PMID: 30139705 DOI: 10.1016/j.trre.2018.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/08/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
B cells play a crucial role in alloreactivity of organ transplant rejection and graft versus host diseases (GVHD). Over the past decade, it has been well recognized that B-cell infiltration in allografts and de novo donor-specific antibodies (DSA) were strongly associated with severe graft rejection and loss, as well as glucocorticoid resistance. Emerging evidence has demonstrated that Follicular T helper (Tfh) cells are key effectors to promote the proliferation and differentiation of B cells into antibody-producing plasmablasts and memory B cells. T-follicular regulatory (Tfr) cells are a recently recognized cell population that has a negative regulatory role on Tfh cells in the follicle, preventing incessant antibody production. It is still less clear how those humoral immunoreactive cells affect transplant rejection and allograft loss. This review focuses on the production and function of Tfr/Tfh cells in the transplant environment. Better understanding of the functions and mechanisms of Tfr/Tfh cells will help to design new strategies to prevent allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Maogen Chen
- Organ transplant center, First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou 510080, PR China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou 510080, PR China
| | - Xiaohong Lin
- Division of general surgery, The Eastern Hospital of the First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Nancy Olsen
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Xiaoshun He
- Organ transplant center, First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou 510080, PR China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou 510080, PR China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA.
| |
Collapse
|
28
|
Poultsidi A, Dimopoulos Y, He TF, Chavakis T, Saloustros E, Lee PP, Petrovas C. Lymph Node Cellular Dynamics in Cancer and HIV: What Can We Learn for the Follicular CD4 (Tfh) Cells? Front Immunol 2018; 9:2233. [PMID: 30319664 PMCID: PMC6170630 DOI: 10.3389/fimmu.2018.02233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022] Open
Abstract
Lymph nodes (LNs) are central in the generation of adaptive immune responses. Follicular helper CD4 T (Tfh) cells, a highly differentiated CD4 population, provide critical help for the development of antigen-specific B cell responses within the germinal center. Throughout the past decade, numerous studies have revealed the important role of Tfh cells in Human Immunodeficiency Virus (HIV) pathogenesis as well as in the development of neutralizing antibodies post-infection and post-vaccination. It has also been established that tumors influence various immune cell subsets not only in their proximity, but also in draining lymph nodes. The role of local or tumor associated lymph node Tfh cells in disease progression is emerging. Comparative studies of Tfh cells in chronic infections and cancer could therefore provide novel information with regards to their differentiation plasticity and to the mechanisms regulating their development.
Collapse
Affiliation(s)
- Antigoni Poultsidi
- Department of Surgery, Medical School, University of Thessaly, Larissa, Greece
| | - Yiannis Dimopoulos
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Ting-Fang He
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Emmanouil Saloustros
- Department of Internal Medicine, Medical School, University of Thessaly, Larissa, Greece
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
29
|
Danelli L, Donnarumma T, Kassiotis G. Correlates of Follicular Helper Bias in the CD4 T Cell Response to a Retroviral Antigen. Front Immunol 2018; 9:1260. [PMID: 29951052 PMCID: PMC6008654 DOI: 10.3389/fimmu.2018.01260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cell differentiation is influenced by a plethora of intrinsic and extrinsic factors, providing the immune system with the ability to tailor its response according to specific stimuli. Indeed, different classes of pathogens may induce a distinct balance of CD4+ T cell differentiation programmes. Here, we report an uncommonly strong bias toward follicular helper (Tfh) differentiation of CD4+ T cells reactive with a retroviral envelope glycoprotein model antigen, presented in its natural context during retroviral infection. Conversely, the response to the same antigen, presented in different immunization regimens, elicited a response typically balanced between Tfh and T helper 1 cells. Comprehensive quantitation of variables known to influence Tfh differentiation revealed the closest correlation with the strength of T cell receptor (TCR) signaling, leading to PD-1 expression, but not with surface TCR downregulation, irrespective of TCR clonotypic avidity. In contrast, strong TCR signaling leading to TCR downregulation and induction of LAG3 expression in high TCR avidity clonotypes restrained CD4+ T cell commitment and further differentiation. Finally, stunted Th1 differentiation, correlating with limited IL-2 availability in retroviral infection, provided permissive conditions for Tfh development, suggesting that Tfh differentiation is the default program of envelope-reactive CD4+ T cells.
Collapse
Affiliation(s)
- Luca Danelli
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
| | - Tiziano Donnarumma
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
|
31
|
Sebina I, Pepper M. Humoral immune responses to infection: common mechanisms and unique strategies to combat pathogen immune evasion tactics. Curr Opin Immunol 2018; 51:46-54. [PMID: 29477969 DOI: 10.1016/j.coi.2018.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 01/08/2023]
Abstract
Humoral immune responses are crucial for protection against invading pathogens and are the underlying mechanism of protection for most successful vaccines. Our understanding of how humoral immunity develops is largely based on animal models utilizing experimental immunization systems. While these studies have made enormous progress for the field and have defined many of the fundamental principles of B cell differentiation and function, we are only now beginning to appreciate the complexities of humoral immune responses induced by infection. Co-evolution of the adaptive immune system and the pathogenic world has created a diverse array of B cell responses to infections, with both shared and unique strategies. In this review, we consider the common mechanisms that regulate the development of humoral immune responses during infection and highlight recent findings demonstrating the evolution of unique strategies used by either host or pathogen for survival.
Collapse
Affiliation(s)
- Ismail Sebina
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
32
|
Weinstein JS, Laidlaw BJ, Lu Y, Wang JK, Schulz VP, Li N, Herman EI, Kaech SM, Gallagher PG, Craft J. STAT4 and T-bet control follicular helper T cell development in viral infections. J Exp Med 2017; 215:337-355. [PMID: 29212666 PMCID: PMC5748849 DOI: 10.1084/jem.20170457] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 09/10/2017] [Accepted: 11/07/2017] [Indexed: 12/26/2022] Open
Abstract
Follicular helper T (Tfh) cells promote germinal center (GC) B cell survival and proliferation and guide their differentiation and immunoglobulin isotype switching by delivering contact-dependent and soluble factors, including IL-21, IL-4, IL-9, and IFN-γ. IL-21 and IFN-γ are coexpressed by Tfh cells during viral infections, but transcriptional regulation of these cytokines is not completely understood. In this study, we show that the T helper type 1 cell (Th1 cell) transcriptional regulators T-bet and STAT4 are coexpressed with Bcl6 in Tfh cells after acute viral infection, with a temporal decline in T-bet in the waning response. T-bet is important for Tfh cell production of IFN-γ, but not IL-21, and for a robust GC reaction. STAT4, phosphorylated in Tfh cells upon infection, is required for expression of T-bet and Bcl6 and for IFN-γ and IL-21. These data indicate that T-bet is expressed with Bcl6 in Tfh cells and is required alongside STAT4 to coordinate Tfh cell IL-21 and IFN-γ production and for promotion of the GC response after acute viral challenge.
Collapse
Affiliation(s)
- Jason S Weinstein
- Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT
| | - Brian J Laidlaw
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Yisi Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Jessica K Wang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Vincent P Schulz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Ningcheng Li
- Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT
| | - Edward I Herman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Susan M Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Patrick G Gallagher
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT.,Department of Pathology and Genetics, Yale University School of Medicine, New Haven, CT
| | - Joe Craft
- Department of Internal Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
33
|
de Graav GN, Hesselink DA, Dieterich M, Kraaijeveld R, Verschoor W, Roelen DL, Litjens NHR, Chong AS, Weimar W, Baan CC. Belatacept Does Not Inhibit Follicular T Cell-Dependent B-Cell Differentiation in Kidney Transplantation. Front Immunol 2017; 8:641. [PMID: 28620390 PMCID: PMC5450507 DOI: 10.3389/fimmu.2017.00641] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
Humoral alloreactivity has been recognized as a common cause of kidney transplant dysfunction. B-cell activation, differentiation, and antibody production are dependent on IL-21+CXCR5+follicular T-helper (Tfh) cells. Here, we studied whether belatacept, an inhibitor of the costimulatory CD28-CD80/86-pathway, interrupts the crosstalk between Tfh- and B-cells more efficiently than the calcineurin inhibitor tacrolimus. The suppressive effects of belatacept and tacrolimus on donor antigen-driven Tfh-B-cell interaction were functionally studied in peripheral blood mononuclear cells from 40 kidney transplant patients randomized to a belatacept- or tacrolimus-based immunosuppressive regimen. No significant differences in uncultured cells or donor antigen-stimulated cells were found between belatacept- and tacrolimus-treated patients in the CXCR5+Tfh cell generation and activation (upregulation of PD-1). Belatacept and tacrolimus in vitro minimally inhibited Tfh-cell generation (by ~6-7%) and partially prevented Tfh-cell activation (by ~30-50%). The proportion of IL-21+-activated Tfh-cells was partially decreased by in vitro addition of belatacept or tacrolimus (by ~60%). Baseline expressions and proportions of activated CD86+ B-cells, plasmablasts, and transitional B-cells after donor antigen stimulation did not differ between belatacept- and tacrolimus-treated patients. Donor antigen-driven CD86 upregulation on memory B-cells was not fully prevented by adding belatacept in vitro (~35%), even in supratherapeutic doses. In contrast to tacrolimus, belatacept failed to inhibit donor antigen-driven plasmablast formation (~50% inhibition vs. no inhibition, respectively, p < 0.0001). In summary, donor antigen-driven Tfh-B-cell crosstalk is similar in cells obtained from belatacept- and tacrolimus-treated patients. Belatacept is, however, less potent in vitro than tacrolimus in inhibiting Tfh-cell-dependent plasmablast formation.
Collapse
Affiliation(s)
- Gretchen N de Graav
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Rens Kraaijeveld
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Wenda Verschoor
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Dave L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Nicolle H R Litjens
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, United States
| | - Willem Weimar
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Section Transplantation and Nephrology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
34
|
Fujikura D, Ikesue M, Endo T, Chiba S, Higashi H, Uede T. Death receptor 6 contributes to autoimmunity in lupus-prone mice. Nat Commun 2017; 8:13957. [PMID: 28045014 PMCID: PMC5216082 DOI: 10.1038/ncomms13957] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 11/15/2016] [Indexed: 01/20/2023] Open
Abstract
Expansion of autoreactive follicular helper T (Tfh) cells is tightly restricted to prevent induction of autoantibody-dependent immunological diseases, such as systemic lupus erythematosus (SLE). Here we show expression of an orphan immune regulator, death receptor 6 (DR6/TNFRSF21), on a population of Tfh cells that are highly expanded in lupus-like disease progression in mice. Genome-wide screening reveals an interaction between syndecan-1 and DR6 resulting in immunosuppressive functions. Importantly, syndecan-1 is expressed specifically on autoreactive germinal centre (GC) B cells that are critical for maintenance of Tfh cells. Syndecan-1 expression level on GC B cells is associated with Tfh cell expansion and disease progression in lupus-prone mouse strains. In addition, Tfh cell suppression by DR6-specific monoclonal antibody delays disease progression in lupus-prone mice. These findings suggest that the DR6/syndecan-1 axis regulates aberrant GC reactions and could be a therapeutic target for autoimmune diseases such as SLE. Germinal centre (GC) reactions are driven by T follicular helper (Tfh) cells and their dysregulation can cause autoimmune disease. Here the authors show that the orphan receptor DR6 is a Tfh cell marker that binds syndecan-1 on GC B cells driving autoimmunity in lupus-prone mice.
Collapse
Affiliation(s)
- Daisuke Fujikura
- Division of Infection and Immunity, Hokkaido University Research Center for Zoonosis Control, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan.,Division of Molecular Immunology, Hokkaido University Institute for Genetic Medicine, North-15, West-7, Kita-ku, Sapporo 060-0815, Japan.,Division of Bioresources, Hokkaido University Research Center for Zoonosis Control, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan
| | - Masahiro Ikesue
- Division of Molecular Immunology, Hokkaido University Institute for Genetic Medicine, North-15, West-7, Kita-ku, Sapporo 060-0815, Japan
| | - Tsutomu Endo
- Division of Molecular Immunology, Hokkaido University Institute for Genetic Medicine, North-15, West-7, Kita-ku, Sapporo 060-0815, Japan
| | - Satoko Chiba
- Division of Infection and Immunity, Hokkaido University Research Center for Zoonosis Control, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan.,Division of Bioresources, Hokkaido University Research Center for Zoonosis Control, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan
| | - Hideaki Higashi
- Division of Infection and Immunity, Hokkaido University Research Center for Zoonosis Control, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan
| | - Toshimitsu Uede
- Division of Molecular Immunology, Hokkaido University Institute for Genetic Medicine, North-15, West-7, Kita-ku, Sapporo 060-0815, Japan
| |
Collapse
|
35
|
TFH cells progressively differentiate to regulate the germinal center response. Nat Immunol 2016; 17:1197-1205. [PMID: 27573866 PMCID: PMC5030190 DOI: 10.1038/ni.3554] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022]
Abstract
Germinal center (GC) B cells undergo affinity selection, which depends on interactions with CD4(+) follicular helper T cells (TFH cells). We found that TFH cells progressed through transcriptionally and functionally distinct stages and provided differential signals for GC regulation. They initially localized proximally to mutating B cells, secreted interleukin 21 (IL-21), induced expression of the transcription factor Bcl-6 and selected high-affinity B cell clones. As the GC response evolved, TFH cells extinguished IL-21 production and switched to IL-4 production, showed robust expression of the co-stimulatory molecule CD40L, and promoted the development of antibody-secreting B cells via upregulation of the transcription factor Blimp-1. Thus, TFH cells in the B cell follicle progressively differentiate through stages of localization, cytokine production and surface ligand expression to 'fine tune' the GC reaction.
Collapse
|
36
|
Wikenheiser DJ, Stumhofer JS. ICOS Co-Stimulation: Friend or Foe? Front Immunol 2016; 7:304. [PMID: 27559335 PMCID: PMC4979228 DOI: 10.3389/fimmu.2016.00304] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/27/2016] [Indexed: 12/18/2022] Open
Abstract
Over the last 15 years, the inducible T cell co-stimulator (ICOS) has been implicated in various immune outcomes, including the induction and regulation of Th1, Th2, and Th17 immunity. In addition to its role in directing effector T cell differentiation, ICOS has also been consistently linked with the induction of thymus-dependent (TD) antibody (Ab) responses and the germinal center (GC) reaction. ICOS co-stimulation, therefore, appears to play a complex role in dictating the course of adaptive immunity. In this article, we summarize the initial characterization of ICOS and its relationship with the related co-stimulatory molecule CD28. We then address the contribution of ICOS in directing an effector T cell response, and ultimately disease outcome, against various bacterial, viral, and parasitic infections. Next, we assess ICOS in the context of TD Ab responses, connecting ICOS signaling to follicular helper T cell differentiation and its role in the GC reaction. Finally, we address the link between ICOS and human autoimmune disorders and evaluate potential therapies aiming to mitigate disease progression by modulating ICOS signaling.
Collapse
Affiliation(s)
- Daniel J Wikenheiser
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences , Little Rock, AR , USA
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences , Little Rock, AR , USA
| |
Collapse
|
37
|
Abstract
The development of multiple disease-relevant autoantibodies is a hallmark of autoimmune diseases. In autoimmune type 1 diabetes (T1D), a variable time frame of autoimmunity precedes the clinically overt disease. The relevance of T follicular helper (TFH) cells for the immune system is increasingly recognized. Their pivotal contribution to antibody production by providing help to germinal center (GC) B cells facilitates the development of a long-lived humoral immunity. Their complex differentiation process, involving various stages and factors like B cell lymphoma 6 (Bcl6), is strictly controlled, as anomalous regulation of TFH cells is connected with immunopathologies. While the adverse effects of a TFH cell-related insufficient humoral immunity are obvious, the role of increased TFH frequencies in autoimmune diseases like T1D is currently highlighted. High levels of autoantigen trigger an excessive induction of TFH cells, consequently resulting in the production of autoantibodies. Therefore, TFH cells might provide promising approaches for novel therapeutic strategies.
Collapse
Affiliation(s)
- Martin G Scherm
- Institute for Diabetes Research, Independent Young Investigator Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, Munich, 80939, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), am Helmholtz Zentrum München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany
| | - Verena B Ott
- Deutsches Zentrum für Diabetesforschung (DZD), am Helmholtz Zentrum München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Technische Universität München, Parkring 13, Garching, 85748, Germany
- Institute for Advanced Study, Technische Universität München, Lichtenbergstr. 2a, Garching, 85748, Germany
| | - Carolin Daniel
- Institute for Diabetes Research, Independent Young Investigator Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, Munich, 80939, Germany.
- Deutsches Zentrum für Diabetesforschung (DZD), am Helmholtz Zentrum München, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
| |
Collapse
|
38
|
Huang J, Wu Z, Lu S, Shen J, Kong X, Shen Y. Soluble B7-H2 as a novel marker in early evaluation of the severity of acute pancreatitis. Lab Med 2016; 46:109-17. [PMID: 25918189 DOI: 10.1309/lmfsrh0v82hfxppi] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The clinical usefulness of soluble B7-H2 (sB7-H2) as an early indicator of acute pancreatitis (AP) remains unclear, so we performed the present study to investigate this issue. For our cohort, we recruited 75 patients with AP, 70 patients with other abdominal sepsis, and 20 healthy control individuals. The sB7-H2 levels of AP patients or healthy control individuals were measured by enzyme-linked immunosorbent assay (ELISA). The sB7-H2 levels in patients with AP rather than other patients with abdominal sepsis were significantly higher than those in healthy controls. Hence, we selected AP to study the clinical significance of sB7-H2 in inflammatory conditions. The sB7-H2 level was positively correlated with the white blood cell (WBC) count and the lactate dehydrogenase (LDH), high-sensitivity C-reactive protein (hs-CRP), and lipopolysaccharide LPS levels (P <.05 for each). Receiver operating characteristic (ROC) analysis revealed that sB7-H2 can distinguish moderately severe acute pancreatitis (MSAP) and severe acute pancreatitis (SAP) from mildly acute pancreatitis (MAP) with 77.8% sensitivity and 80.0% specificity; and that the levels of sB7-H2 also can distinguish SAP from MSAP and MAP with 92.0% sensitivity and 86.0% specificity. The present results indicate that sB7-H2 might be a useful marker in the clinical diagnosis of AP.
Collapse
Affiliation(s)
- Jian Huang
- Department of Emergency Internal Medicine of the First Affiliated Hospital, Suzhou University, China
| | - Zhengwang Wu
- Department of General Surgery, No. 7 Hospital of Changzhou People's Hospital, China
| | - Shiqi Lu
- Department of Emergency Internal Medicine of the First Affiliated Hospital, Suzhou University, China
| | - Jiaqing Shen
- Department of Emergency Internal Medicine of the First Affiliated Hospital, Suzhou University, China
| | - Xiaoming Kong
- Department of Emergency Internal Medicine of the First Affiliated Hospital, Suzhou University, China
| | - Yueping Shen
- School of Public Health at the Medical College of Suzhou University, China
| |
Collapse
|
39
|
Kannan S, Kurupati RK, Doyle SA, Freeman GJ, Schmader KE, Ertl HCJ. BTLA expression declines on B cells of the aged and is associated with low responsiveness to the trivalent influenza vaccine. Oncotarget 2016; 6:19445-55. [PMID: 26277622 PMCID: PMC4637297 DOI: 10.18632/oncotarget.4597] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/11/2015] [Indexed: 12/31/2022] Open
Abstract
Virus-neutralizing antibody and B cell responses to influenza A viruses were measured in 35 aged and 28 middle-aged individuals following vaccination with the 2012 and 2013 trivalent inactivated influenza vaccines. Antibody responses to the vaccine strains were lower in the aged. An analysis of B cell subsets by flow cytometry with stains for immunoregulators showed that B cells of multiple subsets from the aged as compared to younger human subjects showed differences in the expression of the co-inhibitor B and T lymphocyte attenuator (BTLA). Expression of BTLA inversely correlated with age and appears to be linked to shifting the nature of the response from IgM to IgG. High BTLA expression on mature B cells was linked to higher IgG responses to the H1N1 virus. Finally, high BTLA expression on isotype switched memory B cells was linked to better preservation of virus neutralizing antibody titers and improved recall responses to vaccination given the following year.
Collapse
Affiliation(s)
- Senthil Kannan
- Biomedical Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.,The Wistar Institute, Philadelphia, PA, USA
| | | | - Susan A Doyle
- GRECC, Durham VA Medical Center and Center for the Study of Aging and Human, Development and Division of Geriatrics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Gordon J Freeman
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth E Schmader
- GRECC, Durham VA Medical Center and Center for the Study of Aging and Human, Development and Division of Geriatrics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
40
|
A TRAF-like motif of the inducible costimulator ICOS controls development of germinal center TFH cells via the kinase TBK1. Nat Immunol 2016; 17:825-33. [PMID: 27135603 PMCID: PMC4915981 DOI: 10.1038/ni.3463] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/31/2016] [Indexed: 12/15/2022]
Abstract
Inducible costimulator (ICOS) signaling fuels the stepwise development of T follicular helper (TFH) cells. However, a signaling pathway unique to ICOS has not been identified. We show that TANK-binding kinase 1 (TBK1) associates with ICOS via a conserved motif, IProx, which shares homology with tumor necrosis factor receptor (TNFR)-associated factors, TRAF2 and TRAF3. Disruption of this motif abolishes the association with TBK1, thus identifying a TBK1-binding consensus. Mutation of this motif in ICOS, or depletion of TBK1 in T cells severely impaired the differentiation of germinal center (GC) TFH, B cell and antibody responses, but was dispensable for early TFH differentiation. These results reveal a novel ICOS-TBK1 signaling pathway that specifies GC TFH cell commitment.
Collapse
|
41
|
Hu X, Wu J, An J, Hu Y, Shen Y, Liu C, Zhang X. Development of a novel monoclonal antibody to human inducible co-stimulator ligand (ICOSL): Biological characteristics and application for enzyme-linked immunosorbent assay. Int Immunopharmacol 2016; 36:151-157. [PMID: 27138044 DOI: 10.1016/j.intimp.2016.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/28/2022]
Abstract
ICOSL (B7-H2, CD275), a co-stimulatory molecule of the B7 superfamily, functions as a positive signal in immune response. To investigate whether ICOSL could be released into sera and the possible biological function of soluble ICOS (sICOSL), we generated and characterized a functional anti-human ICOSL monoclonal antibody (mAb), 20B10, and developed a novel enzyme-linked immunosorbent assay (ELISA) based on two anti-human ICOSL antibodies with different epitope specificities. Using the ELISA system, we found that sICOSL in the serum of healthy donors increases in an age-dependent manner and that the matrix metalloproteinase inhibitor (MMPI) could suppress sICOSL production. Together, these data demonstrate that the existence of circulating sICOSL in human serum might play an important role in immunoregulation.
Collapse
Affiliation(s)
- Xiaohan Hu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Jian Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jingnan An
- Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Yumin Hu
- Department of Immunology, Medical College of Soochow University, Suzhou 215006, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Cuiping Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Immunology, Medical College of Soochow University, Suzhou 215006, China.
| |
Collapse
|
42
|
Hu X, Liu C, An J, Shen Y, Hu Y, Jiang J, Wu J, Zhang X. Development of a Novel Functional Monoclonal Antibody to Human CD275: Characterization and Biological Activity. Monoclon Antib Immunodiagn Immunother 2016; 35:18-24. [PMID: 26788906 DOI: 10.1089/mab.2015.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CD275 (B7-H2, ICOSL), a co-stimulatory molecule of the B7 superfamily, plays a critical role in immune response. In this report, a novel mouse anti-human CD275 monoclonal antibody (MAb) was prepared using hybridoma technology, and immunological characteristics of the MAb were determined. The results showed that the MAb (clone 13D11) was IgG2(κ) and bound specifically to human CD275. By mutual competition, we found that the antibody recognized different epitopes of CD275 antigen compared with commercial antibodies and could block ICOS-CD275 interaction. Crosslinking of CD275 with MAb 13D11 markedly blocked ICOS positive signal and inhibited T cell proliferation and cytokine production. In addition, the 13D11 MAb was suitable for indirect ELISA detection. Thus, the MAb against human CD275 with high specificity and different activity would be useful for further study of this molecule.
Collapse
Affiliation(s)
- Xiaohan Hu
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Cuiping Liu
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Jingnan An
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Yu Shen
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Yumin Hu
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China
| | - Juean Jiang
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Jian Wu
- 2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| | - Xueguang Zhang
- 1 Jiangsu Province Key Laboratory of Stem Cell Research, Soochow University , Suzhou, China .,2 Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University , Suzhou, China
| |
Collapse
|
43
|
Local triggering of the ICOS coreceptor by CD11c(+) myeloid cells drives organ inflammation in lupus. Immunity 2015; 42:552-65. [PMID: 25786178 DOI: 10.1016/j.immuni.2015.02.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 11/22/2014] [Accepted: 01/19/2015] [Indexed: 01/13/2023]
Abstract
The inducible T cell costimulator (ICOS) is a potent promoter of organ inflammation in murine lupus. ICOS stimulates T follicular helper cell differentiation in lymphoid tissue, suggesting that it might drive autoimmunity by enhancing autoantibody production. Yet the pathogenic relevance of this mechanism remains unclear. It is also unknown whether other ICOS-induced processes might contribute to lupus pathology. Here we show that selective ablation of ICOS ligand (ICOSL) in CD11c(+) cells, but not in B cells, dramatically ameliorates kidney and lung inflammation in lupus-prone MRL.Fas(lpr) mice. Autoantibody formation was largely unaffected by ICOSL deficiency in CD11c(+) cells. However, ICOSL display by CD11c(+) cells in inflamed organs had a nonredundant role in protecting invading T cells from apoptosis by elevating activity of the PI3K-Akt signaling pathway, thereby facilitating T cell accrual. These findings reveal a mechanism that locally sustains organ inflammation in lupus.
Collapse
|
44
|
Choi JY, Ho JHE, Pasoto SG, Bunin V, Kim S, Carrasco S, Borba EF, Gonçalves CR, Costa PR, Kallas EG, Bonfa E, Craft J. Circulating follicular helper-like T cells in systemic lupus erythematosus: association with disease activity. Arthritis Rheumatol 2015; 67:988-99. [PMID: 25581113 PMCID: PMC4450082 DOI: 10.1002/art.39020] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/30/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To assess circulating follicular helper T (Tfh)-like CD4+ T cells in patients with systemic lupus erythematosus (SLE) and determine their relationship to disease activity. METHODS Blood samples from patients with SLE, as well as blood samples from patients with Behçet's disease (BD) and healthy individuals as controls, were analyzed. In all samples, circulating Tfh-like cells were enumerated by flow cytometry, using, as markers, expression of CXCR5, inducible T cell costimulator (ICOS), and programmed death 1 (PD-1) protein, as well as secretion of interleukin-21 (IL-21). The frequency of circulating Tfh-like cells was compared to that of circulating plasmablasts (CD19+IgD-CD38+). In addition, the possible association of circulating Tfh-like cells with the SLE Disease Activity Index (SLEDAI) was evaluated. RESULTS The subset of circulating Tfh-like T cells, identified as CXCR5(high) ICOS(high) PD-1(high) , was expanded in the blood of SLE patients compared to controls. Circulating Tfh-like cells were found to produce IL-21 and had lower expression of CCR7 as compared to that in circulating CXCR5(high) central memory T cells, thereby enabling their distinction. Expression of PD-1, but not ICOS or CXCR5, was significantly elevated in circulating Tfh-like cells from SLE patients compared to controls. PD-1 expression among CXCR5(high) circulating Tfh-like cells correlated with the SLEDAI, frequency of circulating plasmablasts, and anti-double-stranded DNA antibody positivity, but not with disease duration or past organ injury; rather, this cell profile appeared to be a reflection of current active disease. CONCLUSION Circulating Tfh-like cells are associated with disease activity in SLE, suggesting that their presence indicates abnormal homeostasis of T cell-B cell collaboration, with a causal relationship that is central to disease pathogenesis. These findings also suggest that circulating Tfh-like cells provide a surrogate for aberrant germinal center activity in SLE, and that their PD-1 expression offers a tool for measuring disease activity and monitoring the response to therapies.
Collapse
Affiliation(s)
- Jin-Young Choi
- Department of Internal Medicine (Rheumatology), Yale School of Medicine, New Haven, CT USA
| | - John Hsi-en Ho
- Department of Internal Medicine (Rheumatology), Yale School of Medicine, New Haven, CT USA
| | - Sandra G Pasoto
- Division of Rheumatology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Viviane Bunin
- Department of Internal Medicine (Rheumatology), Yale School of Medicine, New Haven, CT USA
| | - Sangtaek Kim
- Department of Internal Medicine (Rheumatology), Yale School of Medicine, New Haven, CT USA
| | - Solange Carrasco
- Division of Rheumatology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo F Borba
- Division of Rheumatology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Celio R Gonçalves
- Division of Rheumatology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Priscila R Costa
- Division of Immunology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Esper G Kallas
- Division of Immunology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eloisa Bonfa
- Division of Rheumatology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Joe Craft
- Department of Internal Medicine (Rheumatology), Yale School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
45
|
Fairfax KC, Everts B, Amiel E, Smith AM, Schramm G, Haas H, Randolph GJ, Taylor JJ, Pearce EJ. IL-4-secreting secondary T follicular helper (Tfh) cells arise from memory T cells, not persisting Tfh cells, through a B cell-dependent mechanism. THE JOURNAL OF IMMUNOLOGY 2015; 194:2999-3010. [PMID: 25712216 DOI: 10.4049/jimmunol.1401225] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Humoral immunity requires cross-talk between T follicular helper (Tfh) cells and B cells. Nevertheless, a detailed understanding of this intercellular interaction during secondary immune responses is lacking. We examined this by focusing on the response to a soluble, unadjuvanted, pathogen-derived Ag (soluble extract of Schistosoma mansoni egg [SEA]) that induces type 2 immunity. We found that activated Tfh cells persisted for long periods within germinal centers following primary immunization. However, the magnitude of the secondary response did not appear to depend on pre-existing Tfh cells. Instead, Tfh cell populations expanded through a process that was dependent on memory T cells recruited into the reactive LN, as well as the participation of B cells. We found that, during the secondary response, IL-4 was critical for the expansion of a population of plasmablasts that correlated with increased SEA-specific IgG1 titers. Additionally, following immunization with SEA (but not with an Ag that induced type 1 immunity), IL-4 and IL-21 were coproduced by individual Tfh cells, revealing a potential mechanism through which appropriate class-switching can be coupled to plasmablast proliferation to enforce type 2 immunity. Our findings demonstrate a pivotal role for IL-4 in the interplay between T and B cells during a secondary Th2 response and have significant implications for vaccine design.
Collapse
Affiliation(s)
- Keke C Fairfax
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907
| | - Bart Everts
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Eyal Amiel
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT 05405
| | - Amber M Smith
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Helmut Haas
- Research Center Borstel, 23845 Borstel, Germany; and
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Edward J Pearce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
46
|
Weber JP, Fuhrmann F, Feist RK, Lahmann A, Al Baz MS, Gentz LJ, Vu Van D, Mages HW, Haftmann C, Riedel R, Grün JR, Schuh W, Kroczek RA, Radbruch A, Mashreghi MF, Hutloff A. ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2. ACTA ACUST UNITED AC 2015; 212:217-33. [PMID: 25646266 PMCID: PMC4322049 DOI: 10.1084/jem.20141432] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ICOS signaling is required for inhibition of the transcription factor Klf2, which controls expression of genes expressed by follicular T helper (Tfh) cells. When ICOS signaling is blocked, Tfh cells lose expression of characteristic Tfh genes and revert to an effector phenotype, resulting in disruption of the germinal center response. The co-stimulators ICOS (inducible T cell co-stimulator) and CD28 are both important for T follicular helper (TFH) cells, yet their individual contributions are unclear. Here, we show that each molecule plays an exclusive role at different stages of TFH cell development. While CD28 regulated early expression of the master transcription factor Bcl-6, ICOS co-stimulation was essential to maintain the phenotype by regulating the novel TFH transcription factor Klf2 via Foxo1. Klf2 directly binds to Cxcr5, Ccr7, Psgl-1, and S1pr1, and low levels of Klf2 were essential to maintain this typical TFH homing receptor pattern. Blocking ICOS resulted in relocation of fully developed TFH cells back to the T cell zone and reversion of their phenotype to non-TFH effector cells, which ultimately resulted in breakdown of the germinal center response. Our study describes for the first time the exclusive role of ICOS and its downstream signaling in the maintenance of TFH cells by controlling their anatomical localization in the B cell follicle.
Collapse
Affiliation(s)
- Jan P Weber
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Franziska Fuhrmann
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Randi K Feist
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Annette Lahmann
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Maysun S Al Baz
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Lea-Jean Gentz
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Dana Vu Van
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Hans W Mages
- Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Claudia Haftmann
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany
| | - René Riedel
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany
| | - Joachim R Grün
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Andreas Radbruch
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany
| | - Andreas Hutloff
- Chronic Immune Reactions, Cell Biology, and Bioinformatics, German Rheumatism Research Centre, a Leibniz Institute, 10117 Berlin, Germany Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| |
Collapse
|
47
|
Verstappen GM, Kroese FGM, Vissink A, Bootsma H. Pharmacotherapy for managing extraglandular symptoms of primary Sjögren’s syndrome. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1010510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
48
|
Crotty S. T follicular helper cell differentiation, function, and roles in disease. Immunity 2015; 41:529-42. [PMID: 25367570 DOI: 10.1016/j.immuni.2014.10.004] [Citation(s) in RCA: 1319] [Impact Index Per Article: 146.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Indexed: 12/22/2022]
Abstract
Follicular helper T (Tfh) cells are specialized providers of T cell help to B cells, and are essential for germinal center formation, affinity maturation, and the development of most high-affinity antibodies and memory B cells. Tfh cell differentiation is a multistage, multifactorial process involving B cell lymphoma 6 (Bcl6) and other transcription factors. This article reviews understanding of Tfh cell biology, including their differentiation, migration, transcriptional regulation, and B cell help functions. Tfh cells are critical components of many protective immune responses against pathogens. As such, there is strong interest in harnessing Tfh cells to improve vaccination strategies. Tfh cells also have roles in a range of other diseases, particularly autoimmune diseases. Overall, there have been dramatic advances in this young field, but there is much to be learned about Tfh cell biology in the interest of applying that knowledge to biomedical needs.
Collapse
Affiliation(s)
- Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
49
|
|
50
|
Liu D, Xu H, Shih C, Wan Z, Ma X, Ma W, Luo D, Qi H. T-B-cell entanglement and ICOSL-driven feed-forward regulation of germinal centre reaction. Nature 2014; 517:214-8. [PMID: 25317561 DOI: 10.1038/nature13803] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 08/27/2014] [Indexed: 01/25/2023]
Abstract
The germinal centre (GC) reaction supports affinity-based B-cell competition and generates high-affinity bone-marrow plasma cells (BMPCs). How follicular T-helper (TFH) cells regulate GC selection is not clear. Using competitive mixed chimaera, we show here that, beyond the role in promoting TFH development, ICOSL (inducible T-cell co-stimulator ligand, also known as ICOSLG) is important for individual B cells to competitively participate in the GC reaction and to develop into BMPCs. Using intravital imaging aided by a calcium reporter, we further show that ICOSL promotes an 'entangled' mode of TFH-B-cell interactions, characterized by brief but extensive surface engagement, productive T-cell calcium spikes, and B-cell acquisition of CD40 signals. Reiterated entanglement promotes outer-zone co-localization of outcompeting GC B cells together with TFH cells, affording the former increased access to T-cell help. ICOSL on GC B cells is upregulated by CD40 signals. Such an intercellular positive feedback between contact-dependent help and ICOSL-controlled entanglement promotes positive selection and BMPC development, as evidenced by observations that higher-affinity B-cell receptor variants are enriched in the ICOSL(high) fraction, that numerically disadvantaged ICOSL-deficient GC B cells or BMPCs exhibit strong affinity compensation in competitive chimaera, and that when GC competition proceeds without ICOSL, selection of high-affinity variants in otherwise normal GC reactions is impaired. By demonstrating entanglement as the basic form of GC TFH-B-cell interactions, identifying ICOSL as a molecular linkage between T-B interactional dynamics and positive selection for high-affinity BMPC formation, our study reveals a pathway by which TFH cells control the quality of long-lived humoral immunity.
Collapse
Affiliation(s)
- Dan Liu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Heping Xu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Changming Shih
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Zurong Wan
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Xiaopeng Ma
- School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Weiwei Ma
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Dan Luo
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084 Beijing, China
| |
Collapse
|