1
|
Ke F, Benet ZL, Shelyakin P, Britanova OV, Gupta N, Dent AL, Moore BB, Grigorova IL. Targeted checkpoint control of B cells undergoing positive selection in germinal centers by follicular regulatory T cells. Proc Natl Acad Sci U S A 2024; 121:e2304020121. [PMID: 38261619 PMCID: PMC10835130 DOI: 10.1073/pnas.2304020121] [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: 03/10/2023] [Accepted: 11/20/2023] [Indexed: 01/25/2024] Open
Abstract
Follicular regulatory T cells (Tfr) can play opposite roles in the regulation of germinal center (GC) responses. Depending on the studies, Tfr suppress or support GC and B cell affinity maturation. However, which factors determine positive vs. negative effects of Tfr on the GC B cell is unclear. In this study, we show that GC centrocytes that express MYC up-regulate expression of CCL3 chemokine that is needed for both the positive and negative regulation of GC B cells by Tfr. B cell-intrinsic expression of CCL3 contributes to Tfr-dependent positive selection of foreign Ag-specific GC B cells. At the same time, expression of CCL3 is critical for direct Tfr-mediated suppression of GC B cells that acquire cognate to Tfr nuclear proteins. Our study suggests that CCR5 and CCR1 receptors promote Tfr migration to CCL3 and highlights Ccr5 expression on the Tfr subset that expresses Il10. Based on our findings and previous studies, we suggest a model of chemotactically targeted checkpoint control of B cells undergoing positive selection in GCs by Tfr, where Tfr directly probe and license foreign antigen-specific B cells to complete their positive selection in GCs but, at the same time, suppress GC B cells that present self-antigens cognate to Tfr.
Collapse
Affiliation(s)
- Fang Ke
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Zachary L. Benet
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Pavel Shelyakin
- Abu Dhabi Stem Cells Center, Abu Dhabi4600, United Arab Emirates
- Molecular Technologies Division, Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow117997, Russian Federation
| | - Olga V. Britanova
- Molecular Technologies Division, Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow117997, Russian Federation
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russian Federation
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel24105, Germany
| | - Neetu Gupta
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Alexander L. Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46123
| | - Bethany B. Moore
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
- Department of Internal Medicine, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Irina L. Grigorova
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
2
|
Sokolova S, Grigorova IL. Follicular regulatory T cell subsets in mice and humans: origins, antigen specificity and function. Int Immunol 2023; 35:583-594. [PMID: 37549239 DOI: 10.1093/intimm/dxad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/05/2023] [Indexed: 08/09/2023] Open
Abstract
Follicular regulatory T (Tfr) cells play various roles in immune responses, contributing to both positive and negative regulation of foreign antigen-specific B cell responses, control over autoreactive antibody responses and autoimmunity, and B cell class-switching to IgE and allergy development. Studies conducted on mice uncovered various subsets of CXCR5+FoxP3+CD4+ Tfr cells that could differently contribute to immune regulation. Moreover, recent studies of human Tfr cells revealed similar complexity with various subsets of follicular T cells of different origins and immunosuppressive and/or immunostimulatory characteristics. In this review we will overview and compare Tfr subsets currently identified in mice and humans and will discuss their origins and antigen specificity, as well as potential modes of action and contribution to the control of the autoimmune and allergic reactions.
Collapse
Affiliation(s)
- Sophia Sokolova
- Division of Molecular Technology, Institute of Translational Medicine, Pirogov National Research Medical University, Moscow, 117513, Russia
| | - Irina L Grigorova
- Division of Molecular Technology, Institute of Translational Medicine, Pirogov National Research Medical University, Moscow, 117513, Russia
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Schips M, Mitra T, Bandyopadhyay A, Meyer-Hermann M. Suppressive might of a few: T follicular regulatory cells impede auto-reactivity despite being outnumbered in the germinal centres. Front Immunol 2023; 14:1253704. [PMID: 37818361 PMCID: PMC10561256 DOI: 10.3389/fimmu.2023.1253704] [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: 07/06/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
The selection of high-affinity B cells and the production of high-affinity antibodies are mediated by T follicular helper cells (Tfhs) within germinal centres (GCs). Therein, somatic hypermutation and selection enhance B cell affinity but risk the emergence of self-reactive B cell clones. Despite being outnumbered compared to their helper counterpart, the ablation of T follicular regulatory cells (Tfrs) results in enhanced dissemination of self-reactive antibody-secreting cells (ASCs). The specific mechanisms by which Tfrs exert their regulatory action on self-reactive B cells are largely unknown. We developed computer simulations to investigate how Tfrs regulate either selection or differentiation of B cells to prevent auto-reactivity. We observed that Tfr-induced apoptosis of self-reactive B cells during the selection phase impedes self-reactivity with physiological Tfr numbers, especially when Tfrs can access centrocyte-enriched GC areas. While this aided in selecting non-self-reactive B cells by restraining competition, higher Tfr numbers distracted non-self-reactive B cells from receiving survival signals from Tfhs. Thus, the location and number of Tfrs must be regulated to circumvent such Tfr distraction and avoid disrupting GC evolution. In contrast, when Tfrs regulate differentiation of selected centrocytes by promoting recycling to the dark zone phenotype of self-reactive GC resident pre-plasma cells (GCPCs), higher Tfr numbers were required to impede the circulation of self-reactive ASCs (s-ASCs). On the other hand, Tfr-engagement with GCPCs and subsequent apoptosis of s-ASCs can control self-reactivity with low Tfr numbers, but does not confer selection advantage to non-self-reactive B cells. The simulations predict that to restrict auto-reactivity, natural redemption of self-reactive B cells is insufficient and that Tfrs should increase the mutation probability of self-reactive B cells.
Collapse
Affiliation(s)
- Marta Schips
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Tanmay Mitra
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Arnab Bandyopadhyay
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
- Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universitat Braunschweig, Braunschweig, Germany
| |
Collapse
|
4
|
Matz HC, McIntire KM, Ellebedy AH. 'Persistent germinal center responses: slow-growing trees bear the best fruits'. Curr Opin Immunol 2023; 83:102332. [PMID: 37150126 PMCID: PMC10829534 DOI: 10.1016/j.coi.2023.102332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/09/2023]
Abstract
Germinal centers (GCs) are key microanatomical sites in lymphoid organs where responding B cells mature and undergo affinity-based selection. The duration of the GC reaction has long been assumed to be relatively brief, but recent studies in humans, nonhuman primates, and mice indicate that GCs can last for weeks to months after initial antigen exposure. This review examines recent studies investigating the factors that influence GC duration, including antigen persistence, T-follicular helper cells, and mode of immunization. Potential mechanisms for how persistent GCs influence the B-cell repertoire are considered. Overall, these studies provide a blueprint for how to design better vaccines that elicit persistent GC responses.
Collapse
Affiliation(s)
- Hanover C Matz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, USA.
| |
Collapse
|
5
|
Chen Q, Dent AL. Nonbinary Roles for T Follicular Helper Cells and T Follicular Regulatory Cells in the Germinal Center Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:15-22. [PMID: 37339403 DOI: 10.4049/jimmunol.2200953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/28/2023] [Indexed: 06/22/2023]
Abstract
Development of high-affinity Abs in the germinal center (GC) is dependent on a specialized subset of T cells called "T follicular helper" (TFH) cells that help select Ag-specific B cells. A second T cell subset, T follicular regulatory (TFR) cells, can act as repressors of the GC and Ab response but can also provide a helper function for GC B cells in some contexts. Recent studies showed that, apart from their traditional helper role, TFH cells can also act as repressors of the Ab response, particularly for IgE responses. We review how both TFH and TFR cells express helper and repressor factors that coordinately regulate the Ab response and how the line between these two subsets is less clear than initially thought. Thus, TFH and TFR cells are interconnected and have "nonbinary" functions. However, many questions remain about how these critical cells control the Ab response.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
6
|
Liu X, Liu B, Qi H. Germinal center reaction and output: recent advances. Curr Opin Immunol 2023; 82:102308. [PMID: 37018876 DOI: 10.1016/j.coi.2023.102308] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 04/05/2023]
Abstract
The germinal center (GC) reaction is unique in that it incorporates clonal expansion, somatic mutagenesis, affinity-based selection, and differentiation events all in one tightly packed but highly dynamic microenvironment to produce affinity-matured plasma cells (PCs) or memory B cells (MBCs). Here, we review recent advances in our understanding of how cyclic expansion and selection are orchestrated, how stringency and efficiency of selection are maintained, and how external signals are integrated in B cells to promote post-GC development of PCs and MBCs.
Collapse
Affiliation(s)
- Xin Liu
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Changping Laboratory, Beijing, China
| | - Bo Liu
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Changping Laboratory, Beijing, China
| | - Hai Qi
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Changping Laboratory, Beijing, China; Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
7
|
Ke F, Benet ZL, Maz MP, Liu J, Dent AL, Kahlenberg JM, Grigorova IL. Germinal center B cells that acquire nuclear proteins are specifically suppressed by follicular regulatory T cells. eLife 2023; 12:e83908. [PMID: 36862132 PMCID: PMC9981149 DOI: 10.7554/elife.83908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
Follicular regulatory T cells (Tfr) restrict development of autoantibodies and autoimmunity while supporting high-affinity foreign antigen-specific humoral response. However, whether Tfr can directly repress germinal center (GC) B cells that acquire autoantigens is unclear. Moreover, TCR specificity of Tfr to self-antigens is not known. Our study suggests that nuclear proteins contain antigens specific to Tfr. Targeting of these proteins to antigen-specific B cells in mice triggers rapid accumulation of Tfr with immunosuppressive characteristics. Tfr then exert negative regulation of GC B cells with predominant inhibition of the nuclear protein-acquiring GC B cells, suggesting an important role of direct cognate Tfr-GC B cells interactions for the control of effector B cell response.
Collapse
Affiliation(s)
- Fang Ke
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Zachary L Benet
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Mitra P Maz
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Jianhua Liu
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of MedicineIndianapolisUnited States
| | - Joanne Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Irina L Grigorova
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| |
Collapse
|
8
|
Islam M, Sevak JK, Sharma MK, Jindal A, Vyas AK, Bajpai M, Ramakrishna G, Sarin SK, Trehanpati N. Immune predictors of hepatitis B surface antigen seroconversion in patients with hepatitis B reactivation. Aliment Pharmacol Ther 2023; 57:689-708. [PMID: 36411952 DOI: 10.1111/apt.17306] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/16/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Hepatitis B surface antigen (HBsAg) seroconversion is sometimes observed in hepatitis B reactivation (rHBV), probably due to immune resetting and differentiation. AIMS To investigate sequential immune differentiation and abrogation of tolerance in patients with rHBV who achieved HBsAg seroconversion. METHODS We included 19 patients with chronic hepatitis B (CHBV; HBV DNA log103-8 ), 67 with rHBV (raised ALT [>5XULN], HBV DNAlog104-8 ) and 10 healthy controls. Immune differentiation, tolerance and functional status of CD4, CD8, T regulatory cells (Tregs), B cells and follicular T helper (Tfh) cells were assessed at baseline and 24 weeks. RESULTS At 24 weeks, 81% rHBV (n = 67) lost HBV DNA and HBeAg (41%), and 12 (19%) lost HBsAg and made anti-HBs titers >10 IU/ml. rHBV patients had higher Th1/17, TEM , Tfh, Tfh1/17, plasma and ATM B cells, and lower Tregs, Th2, Th17 and TEMRA expression. rHBV showed lower PD1, TIM3, LAG3, SLAM and TOX compared to CHBV. There was a significant increase in CD8, CD8EM, Tfh, Tfh1/17 and plasma B cells in seroconverters than non-seroconverters. At 24 weeks, we also observed increased plasma B cell frequency in seroconverters. While non-seroconverters showed higher expression of PD1, TIM3, LAG3, SLAM and TOX on CD4/CD8 T cells, blockade of PD1, TIM3, LAG3 and CTLA4 significantly enhanced IFN-γ, TNF-α, IL-4 and IL-21 expression on CD4/CD8 and Tfh cells in non-seroconverters. CONCLUSIONS Non-seroconverters have increased inhibitory markers on CD4/CD8 T cells. There is a critical play of CD8, Tfh and B cells and subsets in seroclearance, along with checkpoint molecules as a potential therapy for non-seroconverters in HBV infection.
Collapse
Affiliation(s)
- Mojahidul Islam
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Jayesh Kumar Sevak
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Manoj Kumar Sharma
- Department of Hepatology, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Ankur Jindal
- Department of Hepatology, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Ashish Kumar Vyas
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Meenu Bajpai
- Department of Transfusion Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Gayatri Ramakrishna
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Nirupma Trehanpati
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| |
Collapse
|
9
|
Acosta F, Fernández PL, Goodridge A. Do B-1 cells play a role in response to Mycobacterium tuberculosis Beijing lineages? Virulence 2022; 13:1-4. [PMID: 34753390 PMCID: PMC8741279 DOI: 10.1080/21505594.2021.2003116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We highlight the need to include an analysis of the B-1 B cell subset to complement the characterization of the cell-mediated immune response to the Mycobacterium tuberculosis Beijing lineage. The literature describes the B-1 cell repertoire's involvement in the cell-mediated response within granulomas, which is different from the classic antibody response B cells are generally associated with. Specifically, the B-1 B cell subset migrates from other compartments along with other cells to the infection site. We provide details to complement the reported results from Cerezo-Cortes et al.
Collapse
Affiliation(s)
- Fermín Acosta
- Centro de Biología Molecular y Celular de las Enfermedades (CBCME) del Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama City, Panamá
| | - Patricia L. Fernández
- Centro de Biología Molecular y Celular de las Enfermedades (CBCME) del Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama City, Panamá
| | - Amador Goodridge
- Centro de Biología Molecular y Celular de las Enfermedades (CBCME) del Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), City of Knowledge, Panama City, Panamá,CONTACT Amador Goodridge
| |
Collapse
|
10
|
Yang R, Avery DT, Jackson KJL, Ogishi M, Benhsaien I, Du L, Ye X, Han J, Rosain J, Peel JN, Alyanakian MA, Neven B, Winter S, Puel A, Boisson B, Payne KJ, Wong M, Russell AJ, Mizoguchi Y, Okada S, Uzel G, Goodnow CC, Latour S, Bakkouri JE, Bousfiha A, Preece K, Gray PE, Keller B, Warnatz K, Boisson-Dupuis S, Abel L, Pan-Hammarström Q, Bustamante J, Ma CS, Casanova JL, Tangye SG. Human T-bet governs the generation of a distinct subset of CD11c highCD21 low B cells. Sci Immunol 2022; 7:eabq3277. [PMID: 35867801 PMCID: PMC9413977 DOI: 10.1126/sciimmunol.abq3277] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
High-level expression of the transcription factor T-bet characterizes a phenotypically distinct murine B cell population known as "age-associated B cells" (ABCs). T-bet-deficient mice have reduced ABCs and impaired humoral immunity. We describe a patient with inherited T-bet deficiency and largely normal humoral immunity including intact somatic hypermutation, affinity maturation and memory B cell formation in vivo, and B cell differentiation into Ig-producing plasmablasts in vitro. Nevertheless, the patient exhibited skewed class switching to IgG1, IgG4, and IgE, along with reduced IgG2, both in vivo and in vitro. Moreover, T-bet was required for the in vivo and in vitro development of a distinct subset of human B cells characterized by reduced expression of CD21 and the concomitantly high expression of CD19, CD20, CD11c, FCRL5, and T-bet, a phenotype that shares many features with murine ABCs. Mechanistically, human T-bet governed CD21loCD11chi B cell differentiation by controlling the chromatin accessibility of lineage-defining genes in these cells: FAS, IL21R, SEC61B, DUSP4, DAPP1, SOX5, CD79B, and CXCR4. Thus, human T-bet is largely redundant for long-lived protective humoral immunity but is essential for the development of a distinct subset of human CD11chiCD21lo B cells.
Collapse
Affiliation(s)
- Rui Yang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Department of Pediatrics, Weill Cornell Medicine, New York, NY, 10065, USA,Corresponding authors: Rui Yang (); Jean-Laurent Casanova (); Stuart Tangye ()
| | - Danielle T. Avery
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia
| | | | - Masato Ogishi
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Ibtihal Benhsaien
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, 20460 Casablanca, Morocco,Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, CHU Averroes, 20460 Casablanca, Morocco
| | - Likun Du
- Department of Biosciences and Nutrition, Karolinska Institutet, 17177 Stockholm, Sweden, EU
| | - Xiaofei Ye
- Department of Biosciences and Nutrition, Karolinska Institutet, 17177 Stockholm, Sweden, EU
| | - Jing Han
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Jessica N. Peel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Marie-Alexandra Alyanakian
- Immunology Laboratory, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France, EU
| | - Bénédicte Neven
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Sarah Winter
- Paris Cité University, Imagine Institute, 75015 Paris, France,Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine Institute, 75015 Paris, France
| | - Anne Puel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Bertrand Boisson
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Kathryn J. Payne
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia
| | - Melanie Wong
- Children’s Hospital at Westmead, NSW, Australia,Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Amanda J. Russell
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia
| | - Yoko Mizoguchi
- Department of Pediatrics, Hiroshima University, Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University, Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Christopher C. Goodnow
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia,St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst 2010, Australia
| | - Sylvain Latour
- Paris Cité University, Imagine Institute, 75015 Paris, France,Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine Institute, 75015 Paris, France
| | - Jalila El Bakkouri
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, 20460 Casablanca, Morocco,Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, CHU Averroes, 20460 Casablanca, Morocco
| | - Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, 20460 Casablanca, Morocco,Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, CHU Averroes, 20460 Casablanca, Morocco
| | - Kahn Preece
- John Hunter Children's Hospital, Newcastle, New South Wales, Australia
| | - Paul E. Gray
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia.,Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stéphanie Boisson-Dupuis
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Laurent Abel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, 17177 Stockholm, Sweden, EU
| | - Jacinta Bustamante
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France,Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia,St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst 2010, Australia
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, 75015 Paris, France,Paris Cité University, Imagine Institute, 75015 Paris, France,Howard Hughes Medical Institute, New York, NY, USA,Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France,Corresponding authors: Rui Yang (); Jean-Laurent Casanova (); Stuart Tangye ()
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst 2010, NSW Australia,St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst 2010, Australia,Corresponding authors: Rui Yang (); Jean-Laurent Casanova (); Stuart Tangye ()
| |
Collapse
|
11
|
Bacteria reduce flagellin synthesis to evade microglia-astrocyte-driven immunity in the brain. Cell Rep 2022; 40:111033. [PMID: 35793624 DOI: 10.1016/j.celrep.2022.111033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The immune response of brain cells to invading bacteria in vivo and the mechanism used by pathogenic bacteria to escape brain immune surveillance remain largely unknown. It is believed that microglia eliminate bacteria by phagocytosis based on in vitro data. Here we find that a small percentage of microglia in the brain engulf neonatal meningitis-causing Escherichia coli (NMEC), but more microglia are activated to produce tumor necrosis factor alpha (TNFα), which activates astrocytes to secrete complement component 3 (C3) involved in anti-bacterial activity. To evade anti-bacterial activity of the immune system, NMEC senses low concentration of threonine in cerebrospinal fluid (CSF) to down-modulate the expression of flagellin and reduce microglial TNFα and astrocyte C3 production. Our findings may help develop strategies for bacterial meningitis treatment.
Collapse
|
12
|
James Bates RE, Browne E, Schalks R, Jacobs H, Tan L, Parekh P, Magliozzi R, Calabrese M, Mazarakis ND, Reynolds R. Lymphotoxin-alpha expression in the meninges causes lymphoid tissue formation and neurodegeneration. Brain 2022; 145:4287-4307. [PMID: 35776111 DOI: 10.1093/brain/awac232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 05/24/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Organised meningeal immune cell infiltrates are suggested to play an important role in cortical grey matter pathology in the multiple sclerosis brain, but the mechanisms involved are as yet unresolved. Lymphotoxin-alpha plays a key role in lymphoid organ development and cellular cytotoxicity in the immune system and its expression is increased in the cerebrospinal fluid of naïve and progressive multiple sclerosis patients and post-mortem meningeal tissue. Here we show that persistently increased levels of lymphotoxin alpha in the cerebral meninges can give rise to lymphoid-like structures and underlying multiple sclerosis-like cortical pathology. Stereotaxic injections of recombinant lymphotoxin-alpha into the rat meninges led to acute meningeal inflammation and subpial demyelination that resolved after 28 days, with demyelination being dependent on prior sub-clinical immunisation with myelin oligodendrocyte glycoprotein. Injection of a lymphotoxin-alpha lentiviral vector into the cortical meningeal space, to produce chronic localised over-expression of the cytokine, induced extensive lymphoid-like immune cell aggregates, maintained over 3 months, including T-cell rich zones containing podoplanin+ fibroblastic reticular stromal cells and B-cell rich zones with a network of follicular dendritic cells, together with expression of lymphoid chemokines and their receptors. Extensive microglial and astroglial activation, subpial demyelination and marked neuronal loss occurred in the underlying cortical parenchyma. Whereas subpial demyelination was partially dependent on prior myelin oligodendrocyte glycoprotein immunisation, the neuronal loss was present irrespective of immunisation. Conditioned medium from LTα treated microglia was able to induce a reactive phenotype in astrocytes. Our results show that chronic lymphotoxin-alpha overexpression alone is sufficient to induce formation of meningeal lymphoid-like structures and subsequent neurodegeneration, similar to that seen in the progressive multiple sclerosis brain.
Collapse
Affiliation(s)
- Rachel E James Bates
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Eleanor Browne
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Renee Schalks
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Heather Jacobs
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Li Tan
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Puja Parekh
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Roberta Magliozzi
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK.,Neurology Section, Department of Neurological and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Massimiliano Calabrese
- Neurology Section, Department of Neurological and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Nicholas D Mazarakis
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
13
|
Hiwa R, Brooks JF, Mueller JL, Nielsen HV, Zikherman J. NR4A nuclear receptors in T and B lymphocytes: Gatekeepers of immune tolerance . Immunol Rev 2022; 307:116-133. [PMID: 35174510 DOI: 10.1111/imr.13072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 12/21/2022]
Abstract
Random VDJ recombination early in T and B cell development enables the adaptive immune system to recognize a vast array of evolving pathogens via antigen receptors. However, the potential of such randomly generated TCRs and BCRs to recognize and respond to self-antigens requires layers of tolerance mechanisms to mitigate the risk of life-threatening autoimmunity. Since they were originally cloned more than three decades ago, the NR4A family of nuclear hormone receptors have been implicated in many critical aspects of immune tolerance, including negative selection of thymocytes, peripheral T cell tolerance, regulatory T cells (Treg), and most recently in peripheral B cell tolerance. In this review, we discuss important insights from many laboratories as well as our own group into the function and mechanisms by which this small class of primary response genes promotes self-tolerance and immune homeostasis to balance the need for host defense against the inherent risks posed by the adaptive immune system.
Collapse
Affiliation(s)
- Ryosuke Hiwa
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA.,Department of Rheumatology and Clinical Immunology, Kyoto University Hospital, Kyoto, Japan
| | - Jeremy F Brooks
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - James L Mueller
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Hailyn V Nielsen
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Julie Zikherman
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| |
Collapse
|
14
|
Jacobsen JT, Hu W, R Castro TB, Solem S, Galante A, Lin Z, Allon SJ, Mesin L, Bilate AM, Schiepers A, Shalek AK, Rudensky AY, Victora GD. Expression of Foxp3 by T follicular helper cells in end-stage germinal centers. Science 2021; 373:373/6552/eabe5146. [PMID: 34437125 DOI: 10.1126/science.abe5146] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 05/19/2021] [Indexed: 12/23/2022]
Abstract
Germinal centers (GCs) are the site of immunoglobulin somatic hypermutation and affinity maturation, processes essential to an effective antibody response. The formation of GCs has been studied in detail, but less is known about what leads to their regression and eventual termination, factors that ultimately limit the extent to which antibodies mature within a single reaction. We show that contraction of immunization-induced GCs is immediately preceded by an acute surge in GC-resident Foxp3+ T cells, attributed at least partly to up-regulation of the transcription factor Foxp3 by T follicular helper (TFH) cells. Ectopic expression of Foxp3 in TFH cells is sufficient to decrease GC size, implicating the natural up-regulation of Foxp3 by TFH cells as a potential regulator of GC lifetimes.
Collapse
Affiliation(s)
- Johanne T Jacobsen
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.
| | - Wei Hu
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Ludwig Center for Cancer Immunotherapy, New York, NY, USA
| | - Tiago B R Castro
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.,Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Sigrid Solem
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Alice Galante
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Zeran Lin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Samuel J Allon
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA.,Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Angelina M Bilate
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Alex K Shalek
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA.,Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Program in Immunology Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Alexander Y Rudensky
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Ludwig Center for Cancer Immunotherapy, New York, NY, USA.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
15
|
Lu Y, Craft J. T Follicular Regulatory Cells: Choreographers of Productive Germinal Center Responses. Front Immunol 2021; 12:679909. [PMID: 34177925 PMCID: PMC8222975 DOI: 10.3389/fimmu.2021.679909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/27/2021] [Indexed: 12/22/2022] Open
Abstract
T follicular regulatory cells, or Tfr cells, are a discernable population of regulatory T (Treg) cells that migrate to the B cell follicle and germinal center (GC) upon immune challenge. These cells express the transcription factor Bcl6, the master regulator required for development and differentiation of T follicular helper cells, and are among a group of previously described Treg cells that use T helper cell–associated transcription factors to adapt their regulatory function to diverse milieus for maintenance of immune homeostasis. While there is consensus that Tfr cells control B-cell autoreactivity, it has been unclear whether they regulate productive, antigen-specific GC responses. Accordingly, understanding the regulatory balancing that Tfr cells play in maintenance of B-cell tolerance while optimizing productive humoral immunity is crucial for vaccine-design strategies. To this end, we discuss recent evidence that Tfr cells promote humoral immunity and memory following viral infections, fitting with the accepted role of Treg cells in maintaining homeostasis with promotion of productive immunity, while mitigating that which is potentially pathological. We also propose models in which Tfr cells regulate antigen-specific B cell responses.
Collapse
Affiliation(s)
- Yisi Lu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Joe Craft
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States.,Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
16
|
CXCL13 plasma levels function as a biomarker for disease activity in patients with chronic lymphocytic leukemia. Leukemia 2020; 35:1610-1620. [PMID: 33087831 DOI: 10.1038/s41375-020-01063-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
The chemoattractant CXCL13 organizes the cellular architecture of B-cell follicles and germinal centers. During adaptive immune responses, CXCL13 plasma concentrations transiently increase and function as a biomarker for normal germinal center activity. Chronic lymphocytic leukemia (CLL) cells express high levels of CXCR5, the receptor for CXCL13, and proliferate in pseudofollicles within secondary lymphoid organs (SLO). Given the morphologic and functional similarities between normal and CLL B-cell expansion in SLO, we hypothesized that CXCL13 plasma concentrations would correlate with CLL disease activity and progression. We analyzed CXCL13 plasma concentrations in 400 CLL patients and correlated the findings with other prognostic markers, time to treatment (TTT), CCL3 and CCL4 plasma concentrations, and in vivo CLL cell proliferation. We found that CXCL13 plasma concentrations were higher in CLL patients with active and advanced stage disease, resulting in a significantly shorter TTT. Accordingly, high CXCL13 levels correlated with other markers of disease activity and CCL3 levels. Higher CLL cell birth rates in vivo also associated with higher CXCL13 plasma concentrations. Interestingly, elevated CXCL13 plasma levels normalized during ibrutinib therapy, and increased in ibrutinib resistance patients. Collectively, these studies emphasize the importance of CXCL13 in crosstalk between CLL cells and the SLO microenvironment.
Collapse
|
17
|
Feng B, Zhu J, Xu Y, Chen W, Sheng X, Feng X, Shi X, Liu J, Pan Q, Yang J, Yu J, Li L, Cao H. Immunosuppressive effects of mesenchymal stem cells on lung B cell gene expression in LPS-induced acute lung injury. Stem Cell Res Ther 2020; 11:418. [PMID: 32977837 PMCID: PMC7517809 DOI: 10.1186/s13287-020-01934-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Background Immune system disorders play important roles in acute lung injury (ALI), and mesenchymal stem cell (MSC) treatment can reduce inflammation during ALI. In this study, we compared the changes in lung B cells during MSC treatment. Methods We investigated the effects of MSCs on lung B cells in a mouse model of lipopolysaccharide (LPS)-induced ALI. MSCs were administered intratracheally 4 h after LPS. As vehicle-treated controls, mice were treated with phosphate-buffered saline (PBS) containing 2% C57BL/6 (PBS group). Histopathological changes, survival rate, inflammatory factor levels, and the number of neutrophils in bronchoalveolar lavage fluid (BALF) were determined. Single-cell RNA sequencing (scRNA-Seq) analysis was performed to evaluate the transcriptional changes in lung B cells between the PBS, LPS, and LPS/MSC groups on days 3 and 7. Results MSC treatment ameliorated LPS-induced ALI, as indicated by the reductions in mortality, the levels of chemokines and cytokines in BALF, and the severity of lung tissue histopathology in ALI mice. Lung B cells in the PBS group remained undifferentiated and had an inhibitory phenotype. Based on our scRNA-Seq results, the differentially expressed genes (DEGs) in lung B cells in both the PBS group and LPS group were involved in chemotaxis processes and some proinflammatory pathways. MSC treatment inhibited the expression of chemokine genes that were upregulated by LPS and were related to the recruitment of neutrophils into lung tissues. Immunoglobulin-related gene expression was decreased in lung B cells of mice treated with LPS/MSC for 7 days. The DEGs regulated by MSCs were enriched in biological processes, including humoral immune response and apoptotic signaling. Conclusions Lung B cells played an important role in the effects of treatment of ALI with MSCs. These observations provide new insights into the mechanisms underlying the effects of MSC treatment for ALI.
Collapse
Affiliation(s)
- Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Jiaqi Zhu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Yanping Xu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Wenyi Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Xinyu Sheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Xiaowei Shi
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Jingqi Liu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Jinfeng Yang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China.,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd, Hangzhou City, 310003, China. .,National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China. .,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, 79 Qingchun Rd, Hangzhou City, 310003, China.
| |
Collapse
|
18
|
Kiripolsky J, Romano RA, Kasperek EM, Yu G, Kramer JM. Activation of Myd88-Dependent TLRs Mediates Local and Systemic Inflammation in a Mouse Model of Primary Sjögren's Syndrome. Front Immunol 2020; 10:2963. [PMID: 31993047 PMCID: PMC6964703 DOI: 10.3389/fimmu.2019.02963] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) are important mediators of chronic inflammation in numerous autoimmune diseases, although the role of these receptors in primary Sjögren's syndrome (pSS) remains incompletely understood. Previous studies in our laboratory established Myd88 as a crucial mediator of pSS, although the disease-relevant ligands and the upstream signaling events that culminate in Myd88 activation have yet to be established. The objective of this study was to identify specific Myd88-dependent TLR-related pathways that are dysregulated both locally and systemically in a mouse model of pSS [NOD.B10Sn-H2b/J (NOD.B10)]. We performed RNA-sequencing on spleens derived from NOD.B10 mice. We then harvested salivary tissue and spleens from Myd88-sufficient and deficient C57BL/10 (BL/10) and NOD.B10 mice and performed flow cytometry to determine expression of Myd88-dependent TLRs. We cultured splenocytes with TLR2 and TLR4 agonists and measured production of inflammatory mediators by ELISA. Next, we evaluated spontaneous and TLR4-mediated inflammatory cytokine secretion in NOD.B10 salivary tissue. Finally, we assessed spontaneous Myd88-dependent cytokine secretion by NOD.B10 salivary cells. We identified dysregulation of numerous TLR-related networks in pSS splenocytes, particularly those employed by TLR2 and TLR4. We found upregulation of TLRs in both the splenic and salivary tissue from pSS mice. In NOD.B10 splenic tissue, robust expression of B cell TLR1 and TLR2 required Myd88. Splenocytes from NOD.B10 mice were hyper-responsive to TLR2 ligation and the endogenous molecule decorin modulated inflammation via TLR4. Finally, we observed spontaneous secretion of numerous inflammatory cytokines and this was enhanced following TLR4 ligation in female NOD.B10 salivary tissue as compared to males. The spontaneous production of salivary IL-6, MCP-1 and TNFα required Myd88 in pSS salivary tissue. Thus, our data demonstrate that Myd88-dependent TLR pathways contribute to the inflammatory landscape in pSS, and inhibition of such will likely have therapeutic utility.
Collapse
Affiliation(s)
- Jeremy Kiripolsky
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Eileen M Kasperek
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Guan Yu
- Department of Biostatistics, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| | - Jill M Kramer
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States.,Department of Oral Diagnostic Sciences, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|