1
|
Bouteau A, Qin Z, Zurawski S, Zurawski G, Igyártó BZ. Langerhans Cells Drive Tfh and B Cell Responses Independent of Canonical Cytokine Signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632426. [PMID: 39868337 PMCID: PMC11760737 DOI: 10.1101/2025.01.10.632426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Dendritic cells (DCs) are key regulators of adaptive immunity, guiding T helper (Th) cell differentiation through antigen presentation, co-stimulation, and cytokine production. However, in steady-state conditions, certain DC subsets, such as Langerhans cells (LCs), induce T follicular helper (Tfh) cells and B cell responses without inflammatory stimuli. Using multiple mouse models and in vitro systems, we investigated the mechanisms underlying steady-state LC-induced adaptive immune responses. We found that LCs drive germinal center Tfh and B cell differentiation and antibody production independently of interleukin-6 (IL-6), type-I interferons, and ICOS ligand (ICOS-L) signaling, which are critical in inflammatory settings. Instead, these responses relied on CD80/CD86-mediated co-stimulation. Our findings challenge the conventional three-signal paradigm by demonstrating that cytokine signaling is dispensable for LC-mediated Tfh and B cell responses in steady-state. These insights provide a framework for understanding homeostatic immunity and the immune system's role in maintaining tolerance or developing autoimmunity under non-inflammatory conditions.
Collapse
Affiliation(s)
- Aurélie Bouteau
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Zhen Qin
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Sandra Zurawski
- Baylor Scott & White Research Institute, Dallas, TX 75204, United States
- Vaccine Research Institute, INSERM, Unité U955, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Gerard Zurawski
- Baylor Scott & White Research Institute, Dallas, TX 75204, United States
- Vaccine Research Institute, INSERM, Unité U955, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Botond Z. Igyártó
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
2
|
Kubo M, Harada Y, Sasaki T. The role of dendritic cells in the instruction of helper T cells in the allergic march. Int Immunol 2024; 36:559-566. [PMID: 39162776 DOI: 10.1093/intimm/dxae050] [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: 04/16/2024] [Accepted: 08/18/2024] [Indexed: 08/21/2024] Open
Abstract
Allergy is a complex array of diseases influenced by innate and adaptive immunity, genetic polymorphisms, and environmental triggers. Atopic dermatitis is a chronic inflammatory skin disease characterized by barrier defects and immune dysregulation, sometimes leading to asthma and food allergies because of the atopic march. During atopic skin inflammation, Langerhans cells and dendritic cells (DCs) in the skin capture and deliver allergen information to local lymph nodes. DCs are essential immune sensors coordinating immune reactions by capturing and presenting antigens to T cells. In the context of allergic responses, DCs play a crucial role in instructing two types of helper T cells-type 2 helper T (Th2) cells and follicular helper T (TFH) cells-in allergic responses and IgE antibody responses. In skin sensitization, the differentiation and function of Th2 cells and TFH cells are influenced by skin-derived factors, including epithelial cytokines, chemokines, and signalling pathways to modify the function of migratory DCs and conventional DCs. In this review, we aim to understand the specific mechanisms involving DCs in allergic responses to provide insights into the pathogenesis of allergic diseases and potential therapeutic strategies.
Collapse
Affiliation(s)
- Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa 230-0045, Japan
| | - Yasuyo Harada
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
| | - Takanori Sasaki
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Chiba 278-0022, Japan
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Alugupalli KR. Characterization of Turbo, a TLR Ligand-based Adjuvant for Glycoconjugate Vaccines. Immunohorizons 2024; 8:527-537. [PMID: 39093309 PMCID: PMC11374753 DOI: 10.4049/immunohorizons.2400040] [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: 04/17/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
Many bacterial polysaccharide vaccines, including the typhoid Vi polysaccharide (ViPS) and tetravalent meningococcal polysaccharide conjugate (MCV4) vaccines, do not incorporate adjuvants and are not highly immunogenic, particularly in infants. I found that endotoxin, a TLR4 ligand in ViPS, contributes to the immunogenicity of typhoid vaccines. Because endotoxin is pyrogenic, and its levels are highly variable in vaccines, I developed monophosphoryl lipid A, a nontoxic TLR4 ligand-based adjuvant named Turbo. Admixing Turbo with ViPS and MCV4 vaccines improved their immunogenicity across all ages and eliminated booster requirement. To understand the characteristics of this adjuvanticity, I compared Turbo with alum. Unlike alum, which polarizes the response toward the IgG1 isotype, Turbo promoted Ab class switching to all IgG isotypes with affinity maturation; the magnitude of this IgG response is durable and accompanied by the presence of long-lived plasma cells in the mouse bone marrow. In striking contrast with the pathways employed by alum, Turbo adjuvanticity is independent of NLPR3, pyroptotic cell death effector Gasdermin D, and canonical and noncanonical inflammasome activation mediated by Caspase-1 and Caspase-11, respectively. Turbo adjuvanticity is primarily dependent on the MyD88 axis and is lost in mice deficient in costimulatory molecules CD86 and CD40, indicating that Turbo adjuvanticity includes activation of these pathways. Because Turbo formulations containing either monophosphoryl lipid A or TLR2 ligands, Pam2CysSerLys4, and Pam3CysSerLys4 help generate Ab response of all IgG isotypes, as an adjuvant Turbo can improve the immunogenicity of glycoconjugate vaccines against a wide range of bacterial pathogens whose elimination requires appropriate IgG isotypes.
Collapse
Affiliation(s)
- Kishore R. Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
- TurboVax Inc., Philadelphia, PA
| |
Collapse
|
4
|
Li J, Liu L, Luo Q, Zhou W, Zhu Y, Jiang W. Exploring the causal relationship between immune cell and all-cause heart failure: a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1363200. [PMID: 38938655 PMCID: PMC11210391 DOI: 10.3389/fcvm.2024.1363200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
Background and objectives Heart failure (HF) is a disease with numerous genetic and environmental factors that affect it. The results of previous studies indicated that immune phenotypes are associated with HF, but there have been inconclusive studies regarding a causal relationship. Therefore, Mendelian randomization (MR) analyses were undertaken to confirm the causal connections between immune phenotypes and HF, providing genetic evidence supporting the association of immune cell factors with HF risk. Methods We selected instrumental variables that met the criteria based on data from the results of genome-wide association studies (GWAS) of immune phenotype and all-cause HF. An evaluation of the causal association between 731 immune cell factors and HF risk was carried out using the inverse variance weighted (IVW), MR-Egger regression (MR-Egger), and weighted median (WM) analysis methods. To determine the horizontal pleiotropy, heterogeneity, and stability of the genetic variants, the MR-Egger intercept test, Cochran's Q test, MR-PRESSO, and leave-one-out sensitivity analysis were performed. Results MR principal method (IVW) analysis showed that a total of 38 immune cell-related factors were significantly causally associated with HF. Further analyses combining three methods (IVW, MR-Egger and WME) showed that six exposure factors significantly associated with heart failure, as shown below. The effect of Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell% Dendritic cell, CD39+ CD8+ T cell% CD8+ T cell, CD3 on Central Memory CD4+ T cell on heart failure was positive. Whereas, a reverse effect was observed for CD14+ CD16+ monocyte% monocyte. Conclusion We investigated the causal relationship between immune phenotypes and all-cause HF. According to the results, Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell Absolute Count, CD62l- CD86+ myeloid Dendritic cell% Dendritic cell, CD39+ CD8+ T cell% CD8+ T cell, CD3 on Central Memory CD4+ T cell aggravate HF, and the risk of HF is decreased by CD14+ CD16+ monocyte% monocyte. These phenotypes may serve as new biomarkers, providing new therapeutic insights for the prevention and treatment of all-cause HF.
Collapse
Affiliation(s)
| | | | | | | | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Kim W, Kim M, Kim B. Unraveling the enigma: housekeeping gene Ugt1a7c as a universal biomarker for microglia. Front Psychiatry 2024; 15:1364201. [PMID: 38666091 PMCID: PMC11043603 DOI: 10.3389/fpsyt.2024.1364201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Background Microglia, brain resident macrophages, play multiple roles in maintaining homeostasis, including immunity, surveillance, and protecting the central nervous system through their distinct activation processes. Identifying all types of microglia-driven populations is crucial due to the presence of various phenotypes that differ based on developmental stages or activation states. During embryonic development, the E8.5 yolk sac contains erythromyeloid progenitors that go through different growth phases, eventually resulting in the formation of microglia. In addition, microglia are present in neurological diseases as a diverse population. So far, no individual biomarker for microglia has been discovered that can accurately identify and monitor their development and attributes. Summary Here, we highlight the newly defined biomarker of mouse microglia, UGT1A7C, which exhibits superior stability in expression during microglia development and activation compared to other known microglia biomarkers. The UGT1A7C sensing chemical probe labels all microglia in the 3xTG AD mouse model. The expression of Ugt1a7c is stable during development, with only a 4-fold variation, while other microglia biomarkers, such as Csf1r and Cx3cr1, exhibit at least a 10-fold difference. The UGT1A7C expression remains constant throughout its lifespan. In addition, the expression and activity of UGT1A7C are the same in response to different types of inflammatory activators' treatment in vitro. Conclusion We propose employing UGT1A7C as the representative biomarker for microglia, irrespective of their developmental state, age, or activation status. Using UGT1A7C can reduce the requirement for using multiple biomarkers, enhance the precision of microglia analysis, and even be utilized as a standard for gene/protein expression.
Collapse
Affiliation(s)
| | | | - Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
6
|
Alugupalli KR. Monophosphoryl Lipid A-based Adjuvant to Promote the Immunogenicity of Multivalent Meningococcal Polysaccharide Conjugate Vaccines. Immunohorizons 2024; 8:317-325. [PMID: 38625118 PMCID: PMC11066721 DOI: 10.4049/immunohorizons.2400013] [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: 02/09/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Activation of the adaptive immune system requires the engagement of costimulatory pathways in addition to B and T cell Ag receptor signaling, and adjuvants play a central role in this process. Many Gram-negative bacterial polysaccharide vaccines, including the tetravalent meningococcal conjugate vaccines (MCV4) and typhoid Vi polysaccharide vaccines, do not incorporate adjuvants. The immunogenicity of typhoid vaccines is due to the presence of associated TLR4 ligands in these vaccines. Because the immunogenicity of MCV4 is poor and requires boosters, I hypothesized that TLR4 ligands are absent in MCV4 and that incorporation of a TLR4 ligand-based adjuvant would improve their immunogenicity. Consistent with this hypothesis, two Food and Drug Administration-approved MCV4 vaccines, MENVEO and MenQuadfi, lack TLR4 ligands. Admixing monophosphoryl lipid A, a TLR4 ligand-based adjuvant formulation named "Turbo" with MCV4 induced significantly improved IgM and IgG responses to all four meningococcal serogroup polysaccharides in adult and aged mice after a single immunization. Furthermore, in infant mice, a single booster was sufficient to promote a robust IgG response and 100% seroconversion when MCV4 was adjuvanted with Turbo. Turbo upregulated the expression of the costimulatory molecules CD40 and CD86 on B cells, and Turbo-driven adjuvanticity is lost in mice deficient in CD40 and CD86. These data suggest that Turbo induces the required costimulatory molecules for its adjuvant activity and that incorporation of Turbo could make bacterial polysaccharide vaccines more immunogenic, minimize booster requirements, and be cost-effective, particularly for those individuals in low- and middle-income and disease-endemic countries.
Collapse
Affiliation(s)
- Kishore R. Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA; and TurboVax Inc., Philadelphia, PA
| |
Collapse
|
7
|
Lu Y, Chiang J, Zhang R, Roche PA, Hodes RJ. TRAF6 and TRAF2/3 Binding Motifs in CD40 Differentially Regulate B Cell Function in T-Dependent Antibody Responses and Dendritic Cell Function in Experimental Autoimmune Encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1814-1822. [PMID: 37921511 PMCID: PMC10694030 DOI: 10.4049/jimmunol.2300607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Expression of the costimulatory molecule CD40 on both B cells and dendritic cells (DCs) is required for induction of experimental autoimmune encephalomyelitis (EAE), and cell-autonomous CD40 expression on B cells is required for primary T-dependent (TD) Ab responses. We now ask whether the function of CD40 expressed by different cell types in these responses is mediated by the same or different cytoplasmic domains. CD40 has been reported to possess multiple cytoplasmic domains, including distinct TRAF6 and TRAF2/3 binding motifs. To elucidate the in vivo function of these motifs in B cells and DCs involved in EAE and TD germinal center responses, we have generated knock-in mice containing distinct CD40 cytoplasmic domain TRAF-binding site mutations and have used these animals, together with bone marrow chimeric mice, to assess the roles that these motifs play in CD40 function. We found that both TRAF2/3 and TRAF6 motifs of CD40 are critically involved in EAE induction and demonstrated that this is mediated by a role of both motifs for priming of pathogenic T cells by DCs. In contrast, the TRAF2/3 binding motif, but not the TRAF6 binding motif, is required for B cell CD40 function in TD high-affinity Ab responses. These data demonstrate that the requirements for expression of specific TRAF-binding CD40 motifs differ for B cells or DCs that function in specific immune responses and thus identify targets for intervention to modulate these responses.
Collapse
Affiliation(s)
- Ying Lu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jeffrey Chiang
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ray Zhang
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Paul A. Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Richard J. Hodes
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
8
|
Foster WS, Lee JL, Thakur N, Newman J, Spencer AJ, Davies S, Woods D, Godfrey L, Hay IM, Innocentin S, Yam-Puc JC, Horner EC, Sharpe HJ, Thaventhiran JE, Bailey D, Lambe T, Linterman MA. Tfh cells and the germinal center are required for memory B cell formation & humoral immunity after ChAdOx1 nCoV-19 vaccination. Cell Rep Med 2022; 3:100845. [PMID: 36455555 PMCID: PMC9663747 DOI: 10.1016/j.xcrm.2022.100845] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Emergence from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been facilitated by the rollout of effective vaccines. Successful vaccines generate high-affinity plasma blasts and long-lived protective memory B cells. Here, we show a requirement for T follicular helper (Tfh) cells and the germinal center reaction for optimal serum antibody and memory B cell formation after ChAdOx1 nCoV-19 vaccination. We found that Tfh cells play an important role in expanding antigen-specific B cells while identifying Tfh-cell-dependent and -independent memory B cell subsets. Upon secondary vaccination, germinal center B cells generated during primary immunizations can be recalled as germinal center B cells again. Likewise, primary immunization GC-Tfh cells can be recalled as either Tfh or Th1 cells, highlighting the pluripotent nature of Tfh cell memory. This study demonstrates that ChAdOx1 nCoV-19-induced germinal centers are a critical source of humoral immunity.
Collapse
Affiliation(s)
- William S Foster
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Jia Le Lee
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Nazia Thakur
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK
| | - Joseph Newman
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
| | - Alexandra J Spencer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Sophie Davies
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK
| | - Danielle Woods
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Leila Godfrey
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Iain M Hay
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Silvia Innocentin
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Juan Carlos Yam-Puc
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Emily C Horner
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Hayley J Sharpe
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Dalan Bailey
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), Oxford OX3 7BN, UK.
| | - Michelle A Linterman
- Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
9
|
Lu Y, Xu M, Dorrier CE, Zhang R, Mayer CT, Wagner D, McGavern DB, Hodes RJ. CD40 Drives Central Nervous System Autoimmune Disease by Inducing Complementary Effector Programs via B Cells and Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2083-2092. [PMID: 36426970 PMCID: PMC10065987 DOI: 10.4049/jimmunol.2200439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/19/2022] [Indexed: 01/04/2023]
Abstract
Costimulatory CD40 plays an essential role in autoimmune diseases, including experimental autoimmune encephalomyelitis (EAE), a murine model of human multiple sclerosis (MS). However, how CD40 drives autoimmune disease pathogenesis is not well defined. Here, we used a conditional knockout approach to determine how CD40 orchestrates a CNS autoimmune disease induced by recombinant human myelin oligodendrocyte glycoprotein (rhMOG). We found that deletion of CD40 in either dendritic cells (DCs) or B cells profoundly reduced EAE disease pathogenesis. Mechanistically, CD40 expression on DCs was required for priming pathogenic Th cells in peripheral draining lymph nodes and promoting their appearance in the CNS. By contrast, B cell CD40 was essential for class-switched MOG-specific Ab production, which played a crucial role in disease pathogenesis. In fact, passive transfer of MOG-immune serum or IgG into mice lacking CD40 on B cells but not DCs reconstituted autoimmune disease, which was associated with inundation of the spinal cord parenchyma by Ig and complement. These data demonstrate that CD40 supports distinct effector programs in B cells and DCs that converge to drive a CNS autoimmune disease and identify targets for intervention.
Collapse
Affiliation(s)
- Ying Lu
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Max Xu
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cayce E. Dorrier
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ray Zhang
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian T. Mayer
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Wagner
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard J. Hodes
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Antigen-presenting T cells provide critical B7 co-stimulation for thymic iNKT cell development via CD28-dependent trogocytosis. Cell Rep 2022; 41:111731. [PMID: 36450247 PMCID: PMC9805342 DOI: 10.1016/j.celrep.2022.111731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/03/2022] [Accepted: 11/04/2022] [Indexed: 12/02/2022] Open
Abstract
Invariant natural killer T (iNKT) cell development in the thymus depends on T cell receptor recognition of CD1d ligand on CD4/CD8 double-positive thymocytes. We previously reported that B7-CD28 co-stimulation is required for thymic iNKT cell development, but the cellular and molecular mechanisms underlying this co-stimulatory requirement are not understood. Here we report that CD28 expression on CD1d-expressing antigen-presenting T cells is required for thymic iNKT cell development. Mechanistically, antigen-presenting T cells provide co-stimulation through an unconventional mechanism, acquiring B7 molecules via CD28-dependent trogocytosis from B7-expressing thymic epithelial cells, dendritic cells, and B cells and providing critical B7 co-stimulation to developing iNKT cells. Thus, the present study demonstrates a mechanism of B7 co-stimulation in thymic T cell development by antigen-presenting T cells.
Collapse
|
11
|
Abboud G, Elshikha AS, Kanda N, Zeumer-Spataro L, Morel L. Contribution of Dendritic Cell Subsets to T Cell-Dependent Responses in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1066-1075. [PMID: 35140132 PMCID: PMC8881363 DOI: 10.4049/jimmunol.2100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 12/21/2021] [Indexed: 02/01/2023]
Abstract
BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.
Collapse
Affiliation(s)
- Georges Abboud
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Ahmed S. Elshikha
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Sharkia, 44519, Egypt
| | - Nathalie Kanda
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Leilani Zeumer-Spataro
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL; and
| |
Collapse
|
12
|
Cook SL, Sievert EP, Sciammas R. B Cell-Intrinsic IRF4 Haploinsufficiency Impairs Affinity Maturation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2992-3003. [PMID: 34759017 PMCID: PMC9085970 DOI: 10.4049/jimmunol.2100747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/29/2021] [Indexed: 11/19/2022]
Abstract
The germinal center (GC) reaction is a coordinated and dynamic ensemble of cells and processes that mediate the maturation and selection of high-affinity GC B cells (GCBs) from lower-affinity precursors and ultimately results in plasma cell and memory cell fates that exit the GC. It is of great interest to identify intrinsic and extrinsic factors that control the selection process. The transcription factor IRF4, induced upon BCR and CD40 signaling, is essential for the acquisition of plasma cell and GCB cell fates. We hypothesized that beyond this early requirement, IRF4 continuously operates at later phases of the B cell response. We show that IRF4 is expressed in GCBs at levels greater than seen in resting cells and plays a role in efficient selection of high-affinity GCBs. Halving Irf4 gene copy number in an Ag-specific murine B cell model, we found that Ag presentation, isotype switching, GC formation and zonation, somatic hypermutation rates, and proliferation were comparable with cells with a full Irf4 allelic complement. In contrast, Irf4 haploinsufficient GCBs exhibited impaired generation of high-affinity cells. Mechanistically, we demonstrate suboptimal Blimp-1 regulation among high-affinity Irf4 haploinsufficient GCBs. Furthermore, in cotransfer settings, we observed a marked disadvantage of Irf4 haploinsufficient cells for GC entry, evidential of ineffective recruitment of T cell help. We propose that, analogous to its role in early GC entry, IRF4 continues to function in the late phase of the Ab response to promote productive T follicular helper cell interactions and to activate optimal Blimp-1 expression during GC selection and affinity maturation.
Collapse
Affiliation(s)
- Sarah L Cook
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA
| | - Evelyn P Sievert
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA
| | - Roger Sciammas
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA
| |
Collapse
|
13
|
Bala N, McGurk AI, Zilch T, Rup AN, Carter EM, Leddon SA, Fowell DJ. T cell activation niches-Optimizing T cell effector function in inflamed and infected tissues. Immunol Rev 2021; 306:164-180. [PMID: 34859453 PMCID: PMC9218983 DOI: 10.1111/imr.13047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 12/29/2022]
Abstract
Successful immunity to infection, malignancy, and tissue damage requires the coordinated recruitment of numerous immune cell subsets to target tissues. Once within the target tissue, effector T cells rely on local chemotactic cues and structural cues from the tissue matrix to navigate the tissue, interact with antigen-presenting cells, and release effector cytokines. This highly dynamic process has been "caught on camera" in situ by intravital multiphoton imaging. Initial studies revealed a surprising randomness to the pattern of T cell migration through inflamed tissues, behavior thought to facilitate chance encounters with rare antigen-bearing cells. Subsequent tissue-wide visualization has uncovered a high degree of spatial preference when it comes to T cell activation. Here, we discuss the basic tenants of a successful effector T cell activation niche, taking cues from the dynamics of Tfh positioning in the lymph node germinal center. In peripheral tissues, steady-state microanatomical organization may direct the location of "pop-up" de novo activation niches, often observed as perivascular clusters, that support early effector T cell activation. These perivascular activation niches appear to be regulated by site-specific chemokines that coordinate the recruitment of dendritic cells and other innate cells for local T cell activation, survival, and optimized effector function.
Collapse
Affiliation(s)
- Noor Bala
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Alexander I McGurk
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Tiago Zilch
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Anastasia N Rup
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Evan M Carter
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Scott A Leddon
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Deborah J Fowell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
14
|
CD40-CD154: A perspective from type 2 immunity. Semin Immunol 2021; 53:101528. [PMID: 34810089 DOI: 10.1016/j.smim.2021.101528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
The interaction between CD40 and CD154 (CD40 ligand) is central in immunology, participating in CD4+ T cell priming by dendritic cells (DC), CD4+ T cell help to B cells and classical macrophage activation by CD4+ T cells. However, its role in the Th2 side of immunology including helminth infection remains incompletely understood. Contrary to viral and bacterial stimuli, helminth products usually do not cause CD40 up-regulation in DC, and exogenous CD40 ligation drives Th2-biased systems towards Th1. On the other hand, CD40 and CD154 are necessary for induction of most Th2 responses. We attempt to reconcile these observations, mainly by proposing that (i) CD40 up-regulation in DC in Th2 systems is mostly induced by alarmins, (ii) the Th2 to Th1 shift induced by exogenous CD40 ligation is related to the capacity of such ligation to enhance IL-12 production by myeloid cells, and (iii) signals elicited by endogenous CD154 available in Th2 contexts and by exogenous CD40 ligation are probably different. We stress that CD40-CD154 is important beyond cognate cellular interactions. In such a context, we argue that the proliferation response of B-cells to IL-4 plus CD154 reflects a Th2-specific mechanism for polyclonal B-cell amplification and IgE production at infection sites. Finally, we argue that CD154 is a general immune activation signal across immune polarization including Th2, and propose that competition for CD154 at tissue sites may provide negative feedback on response induction at each site.
Collapse
|
15
|
Chiang K, Largent AD, Arkatkar T, Thouvenel CD, Du SW, Shumlak N, Woods J, Li QZ, Liu Y, Hou B, Rawlings DJ, Jackson SW. Cutting Edge: A Threshold of B Cell Costimulatory Signals Is Required for Spontaneous Germinal Center Formation in Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2021; 207:2217-2222. [PMID: 34588220 DOI: 10.4049/jimmunol.2100548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022]
Abstract
Cognate interactions between autoreactive B and T cells promote systemic lupus erythematosus pathogenesis by inter alia facilitating spontaneous germinal center (GC) formation. Whereas both myeloid and B cell APCs express B7 ligands (CD80 and CD86), the prevailing model holds that dendritic cell costimulation is sufficient for CD28-dependent T cell activation. In this study, we report that B cell-intrinsic CD80/CD86 deletion unexpectedly abrogates GCs in murine lupus. Interestingly, absent GCs differentially impacted serum autoantibodies. In keeping with distinct extrafollicular and GC activation pathways driving lupus autoantibodies, lack of GCs correlated with loss of RNA-associated autoantibodies but preserved anti-dsDNA and connective tissue autoantibody titers. Strikingly, even heterozygous B cell CD80/CD86 deletion was sufficient to prevent autoimmune GCs and RNA-associated autoantibodies. Together, these findings identify a key mechanism whereby B cells promote lupus pathogenesis by providing a threshold of costimulatory signals required for autoreactive T cell activation.
Collapse
Affiliation(s)
- Kristy Chiang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Andrea D Largent
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Tanvi Arkatkar
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | | | - Samuel W Du
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Natali Shumlak
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Jonathan Woods
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yifan Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Baidong Hou
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA; and.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Shaun W Jackson
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA; .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
16
|
Wang Z, Sun Y, Yao W, Ba Q, Wang H. Effects of Cadmium Exposure on the Immune System and Immunoregulation. Front Immunol 2021; 12:695484. [PMID: 34354707 PMCID: PMC8330548 DOI: 10.3389/fimmu.2021.695484] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Cadmium (Cd), a biologically non-essential heavy metal, is widespread in the environment, including the air, water, and soil, and is widely present in foods and quantum dot preparations. Cd enters the body primarily through inhalation and ingestion. Its biological half-life in humans is 10-35 years; therefore, Cd poses long-term health risks. While most studies on Cd toxicity have focused on organ and tissue damage, the immunotoxicity of Cd has drawn increasing attention recently. Cd accumulates in immune cells, modulates the function of the immune system, triggers immunological responses, and leads to diverse health problems. Cd acts as an immunotoxic agent by regulating the activity and apoptosis of immune cells, altering the secretion of immune cytokines, inducing reactive oxygen species (ROS) production and oxidative stress, changing the frequency of T lymphocyte subsets, and altering the production of selective antibodies in immune cells. This review summarizes the immunological toxicity of Cd, elucidates the mechanisms underlying Cd toxicity in terms of innate immunity and adaptive immunity, and discusses potential strategies to alleviate the adverse effects of Cd on the immune system.
Collapse
Affiliation(s)
- Zhineng Wang
- School of Food and Biotechnological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Ying Sun
- School of Food and Biotechnological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Wenbo Yao
- School of Food and Biotechnological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Qian Ba
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Wieland A, Ahmed R. Immunological lessons from CD28 deficiency in humans. Cell 2021; 184:3595-3597. [PMID: 34242561 DOI: 10.1016/j.cell.2021.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this issue of Cell, Casanova and colleagues examine three family members with a mutation that results in deficiency of the T cell co-stimulatory molecule CD28. These patients exhibit clinical symptoms due to human papillomavirus-2 and -4 infections, show increased levels of Epstein-Barr virus and cytomegalovirus in the blood, and respond poorly to vaccines.
Collapse
Affiliation(s)
- Andreas Wieland
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
18
|
Involvement of Dendritic Cells and Th17 Cells in Induced Tertiary Lymphoid Structures in a Chronic Beryllium Disease Mouse Model. Mediators Inflamm 2021; 2021:8845966. [PMID: 34054347 PMCID: PMC8123089 DOI: 10.1155/2021/8845966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/27/2021] [Accepted: 04/20/2021] [Indexed: 11/18/2022] Open
Abstract
Objective To study airway pathophysiology and the role of dendritic cells (DCs) and IL-17 receptor (IL-17R) signals in a mouse model for CBD. Methods Here, we present a CBD mouse model in which mice were exposed to beryllium during three weeks. We also exposed IL-17R-deficient mice and mice in which DCs were depleted. Results Eight weeks after the initial beryllium exposure, an inflammatory response was detected in the lungs. Mice displayed inflammation of the lower airways that included focal dense infiltrates, granuloma-like foci, and tertiary lymphoid structure (TLS) containing T cells, B cells, and germinal centers. Alveolar cell analysis showed significantly increased numbers of CD4+ T cells expressing IFNγ, IL-17, or both cytokines. The pathogenic role of IL-17R signals was demonstrated in IL-17R-deficient mice, which had strongly reduced lung inflammation and TLS development following beryllium exposure. In CBD mice, pulmonary DC subsets including CD103+ conventional DCs (cDCs), CD11b+ cDCs, and monocyte-derived DCs (moDCs) were also prominently increased. We used diphtheria toxin receptor-mediated targeted cell ablation to conditionally deplete DCs and found that DCs are essential for the maintenance of TLS in CBD. Furthermore, the presence of antinuclear autoantibodies in the serum of CBD mice showed that CBD had characteristics of autoimmune disease. Conclusions We generated a translational model of sarcoidosis driven by beryllium and show that DCs and IL-17R signals play a pathophysiological role in CBD development as well as in established CBD in vivo.
Collapse
|
19
|
Marschall P, Wei R, Segaud J, Yao W, Hener P, German BF, Meyer P, Hugel C, Ada Da Silva G, Braun R, Kaplan DH, Li M. Dual function of Langerhans cells in skin TSLP-promoted T FH differentiation in mouse atopic dermatitis. J Allergy Clin Immunol 2021; 147:1778-1794. [PMID: 33068561 DOI: 10.1016/j.jaci.2020.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is among the most common chronic inflammatory skin diseases, usually occurring early in life, and often preceding other atopic diseases such as asthma. TH2 has been believed to play a crucial role in cellular and humoral response in AD, but accumulating evidence has shown that follicular helper T cell (TFH), a critical player in humoral immunity, is associated with disease severity and plays an important role in AD pathogenesis. OBJECTIVES This study aimed at investigating how TFHs are generated during the pathogenesis of AD, particularly what is the role of keratinocyte-derived cytokine TSLP and Langerhans cells (LCs). METHODS Two experimental AD mouse models were employed: (1) triggered by the overproduction of TSLP through topical application of MC903, and (2) induced by epicutaneous allergen ovalbumin (OVA) sensitization. RESULTS This study demonstrated that the development of TFHs and germinal center (GC) response were crucially dependent on TSLP in both the MC903 model and the OVA sensitization model. Moreover, we found that LCs promoted TFH differentiation and GC response in the MC903 model, and the depletion of Langerin+ dendritic cells (DCs) or selective depletion of LCs diminished the TFH/GC response. By contrast, in the model with OVA sensitization, LCs inhibited TFH/GC response and suppressed TH2 skin inflammation and the subsequent asthma. Transcriptomic analysis of Langerin+ and Langerin- migratory DCs revealed that Langerin+ DCs became activated in the MC903 model, whereas these cells remained inactivated in OVA sensitization model. CONCLUSIONS Together, these studies revealed a dual functionality of LCs in TSLP-promoted TFH and TH2 differentiation in AD pathogenesis.
Collapse
Affiliation(s)
- Pierre Marschall
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Ruicheng Wei
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Justine Segaud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Wenjin Yao
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Pierre Hener
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Beatriz Falcon German
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Pierre Meyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Cecile Hugel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Grace Ada Da Silva
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France
| | | | - Daniel H Kaplan
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Mei Li
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7104, Institut National de la Santé et de la Recherch Médicale U1258, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
20
|
Lee ST, Georgiev H, Breed ER, Ruscher R, Hogquist KA. MHC Class I on murine hematopoietic APC selects Type A IEL precursors in the thymus. Eur J Immunol 2021; 51:1080-1088. [PMID: 33521937 PMCID: PMC9846822 DOI: 10.1002/eji.202048996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 01/29/2021] [Indexed: 01/21/2023]
Abstract
TCRαβ+ CD8α+ CD8β- intestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely β2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+ . However, selective depletion of β2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.
Collapse
Affiliation(s)
| | | | | | - Roland Ruscher
- Corresponding authors: Kristin Hogquist, , Roland Ruscher,
| | | |
Collapse
|
21
|
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: 1.0] [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
|
22
|
Abstract
As the professional antigen-presenting cells of the immune system, dendritic cells (DCs) sense the microenvironment and shape the ensuing adaptive immune response. DCs can induce both immune activation and immune tolerance according to the peripheral cues. Recent work has established that DCs comprise several phenotypically and functionally heterogeneous subsets that differentially regulate T lymphocyte differentiation. This review summarizes both mouse and human DC subset phenotypes, development, diversification, and function. We focus on advances in our understanding of how different DC subsets regulate distinct CD4+ T helper (Th) cell differentiation outcomes, including Th1, Th2, Th17, T follicular helper, and T regulatory cells. We review DC subset intrinsic properties, local tissue microenvironments, and other immune cells that together determine Th cell differentiation during homeostasis and inflammation.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Shuting Chen
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
23
|
Li X, Gong L, Meli AP, Karo-Atar D, Sun W, Zou Y, King IL, Gu H. Cbl and Cbl-b control the germinal center reaction by facilitating naive B cell antigen processing. J Exp Med 2021; 217:151892. [PMID: 32584413 PMCID: PMC7478728 DOI: 10.1084/jem.20191537] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/19/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022] Open
Abstract
Antigen uptake and presentation by naive and germinal center (GC) B cells are different, with the former expressing even low-affinity BCRs efficiently capture and present sufficient antigen to T cells, whereas the latter do so more efficiently after acquiring high-affinity BCRs. We show here that antigen uptake and processing by naive but not GC B cells depend on Cbl and Cbl-b (Cbls), which consequently control naive B and cognate T follicular helper (Tfh) cell interaction and initiation of the GC reaction. Cbls mediate CD79A and CD79B ubiquitination, which is required for BCR-mediated antigen endocytosis and postendocytic sorting to lysosomes, respectively. Blockade of CD79A or CD79B ubiquitination or Cbls ligase activity is sufficient to impede BCR-mediated antigen processing and GC development. Thus, Cbls act at the entry checkpoint of the GC reaction by promoting naive B cell antigen presentation. This regulation may facilitate recruitment of naive B cells with a low-affinity BCR into GCs to initiate the process of affinity maturation.
Collapse
Affiliation(s)
- Xin Li
- Montreal Clinical Research Institute, Montreal, Quebec, Canada.,Department of Microbiology and Immunology, Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Liying Gong
- Montreal Clinical Research Institute, Montreal, Quebec, Canada.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Alexandre P Meli
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, McGill University Health Center, Montreal, Quebec, Canada
| | - Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, McGill University Health Center, Montreal, Quebec, Canada
| | - Weili Sun
- Montreal Clinical Research Institute, Montreal, Quebec, Canada.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Yongrui Zou
- The Feinstein Institute for Medical Research, Manhasset, New York, NY
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, McGill University Health Center, Montreal, Quebec, Canada
| | - Hua Gu
- Montreal Clinical Research Institute, Montreal, Quebec, Canada.,Department of Microbiology and Immunology, Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, Canada.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
24
|
Cao Y, Dong L, He Y, Hu X, Hou Y, Dong Y, Yang Q, Bi Y, Liu G. The direct and indirect regulation of follicular T helper cell differentiation in inflammation and cancer. J Cell Physiol 2021; 236:5466-5480. [PMID: 33421124 DOI: 10.1002/jcp.30263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/03/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Follicular T helper (Tfh) cells play important roles in facilitating B-cell differentiation and inducing the antibody response in humoral immunity and immune-associated inflammatory diseases, including infections, autoimmune diseases, and cancers. However, Tfh cell differentiation is mainly achieved through self-directed differentiation regulation and the indirect regulation mechanism of antigen-presenting cells (APCs). During the direct intrinsic differentiation of naïve CD4+ T cells into Tfh cells, Bcl-6, as the characteristic transcription factor, plays the core role of transcriptional regulation. APCs indirectly drive Tfh cell differentiation mainly by changing cytokine secretion mechanisms. Altered metabolic signaling is also critically involved in Tfh cell differentiation. This review summarizes the recent progress in understanding the direct and indirect regulatory signals and metabolic mechanisms of Tfh cell differentiation and function in immune-associated diseases.
Collapse
Affiliation(s)
- Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Ying He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Xuelian Hu
- Immunochina Pharmaceuticals Co., Ltd., No. 80, Xingshikou Road, Haidian District, Beijing, China
| | - Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, No. 20, East Street, Fengtai District, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| |
Collapse
|
25
|
La Muraglia GM, Zeng S, Crichton ES, Wagener ME, Ford ML, Badell IR. Superior inhibition of alloantibody responses with selective CD28 blockade is CTLA-4 dependent and T follicular helper cell specific. Am J Transplant 2021; 21:73-86. [PMID: 32406182 PMCID: PMC7665991 DOI: 10.1111/ajt.16004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/15/2020] [Accepted: 05/07/2020] [Indexed: 01/25/2023]
Abstract
Anti-donor antibodies cause immunologic injury in transplantation. CD28 blockade with CTLA-4-Ig has the ability to reduce the incidence of these donor-specific antibodies (DSA), but its mechanism is suboptimal for the inhibition of alloimmunity in that CTLA-4-Ig blocks both CD28 costimulation and CTLA-4 coinhibition. Thus selective CD28 blockade that spares CTLA-4 has potential to result in improved inhibition of humoral alloimmunity. To test this possibility, we utilized a full allogeneic mismatch murine transplant model and T follicular helper (Tfh):B cell co-culture system. We observed that selective blockade with an anti-CD28 domain antibody (dAb) compared to CTLA-4-Ig led to superior inhibition of Tfh cell, germinal center, and DSA responses in vivo and better control of B cell responses in vitro. CTLA-4 blockade enhanced the humoral alloresponse and, in combination with anti-CD28 dAb, abrogated the effects of selective blockade. This CTLA-4-dependent inhibition was Tfh cell specific in that CTLA-4 expression by Tfh cells was necessary and sufficient for the improved humoral inhibition observed with selective CD28 blockade. As CD28 blockade attracts interest for control of alloantibodies in the clinic, these data support selective CD28 blockade as a superior strategy to address DSA via the sparing of CTLA-4 and more potent targeting of Tfh cells.
Collapse
Affiliation(s)
| | - Susan Zeng
- Emory Transplant Center, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
26
|
Watanabe M, Lu Y, Breen M, Hodes RJ. B7-CD28 co-stimulation modulates central tolerance via thymic clonal deletion and Treg generation through distinct mechanisms. Nat Commun 2020; 11:6264. [PMID: 33293517 PMCID: PMC7722925 DOI: 10.1038/s41467-020-20070-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
The molecular and cellular mechanisms mediating thymic central tolerance and prevention of autoimmunity are not fully understood. Here we show that B7-CD28 co-stimulation and B7 expression by specific antigen-presenting cell (APC) types are required for clonal deletion and for regulatory T (Treg) cell generation from endogenous tissue-restricted antigen (TRA)-specific thymocytes. While B7-CD28 interaction is required for both clonal deletion and Treg induction, these two processes differ in their CD28 signaling requirements and in their dependence on B7-expressing dendritic cells, B cells, and thymic epithelial cells. Meanwhile, defective thymic clonal deletion due to altered B7-CD28 signaling results in the accumulation of mature, peripheral TRA-specific T cells capable of mediating destructive autoimmunity. Our findings thus reveal a function of B7-CD28 co-stimulation in shaping the T cell repertoire and limiting autoimmunity through both thymic clonal deletion and Treg cell generation.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/metabolism
- Autoimmunity/physiology
- B7-1 Antigen/metabolism
- CD28 Antigens/genetics
- CD28 Antigens/metabolism
- Cell Differentiation/immunology
- Central Tolerance
- Clonal Deletion
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Flow Cytometry
- Gene Knock-In Techniques
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thymocytes/physiology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Masashi Watanabe
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ying Lu
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Michael Breen
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Richard J Hodes
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
27
|
Lutckii A, Strunz B, Zhirkov A, Filipovich O, Rukoiatkina E, Gusev D, Lobzin Y, Fischler B, Aleman S, Sällberg M, Björkström NK. Evidence for B cell maturation but not trained immunity in uninfected infants exposed to hepatitis C virus. Gut 2020; 69:2203-2213. [PMID: 32341018 DOI: 10.1136/gutjnl-2019-320269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/11/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Vertical transmission of hepatitis C virus (HCV) is rare compared with other chronic viral infections, despite that newborns have an immature, and possibly more susceptible, immune system. It further remains unclear to what extent prenatal and perinatal exposure to HCV affects immune system development in neonates. DESIGN To address this, we studied B cells, innate immune cells and soluble factors in a cohort of 62 children that were either unexposed, exposed uninfected or infected with HCV. Forty of these infants were followed longitudinally from birth up until 18 months of age. RESULTS As expected, evidence for B cell maturation was observed with increased age in children, whereas few age-related changes were noticed among innate immune cells. HCV-infected children had a high frequency of HCV-specific IgG-secreting B cells. Such a response was also detected in some exposed but uninfected children but not in uninfected controls. Consistent with this, both HCV-exposed uninfected and HCV-infected infants had evidence of early B cell immune maturation with an increased proportion of IgA-positive plasma cells and upregulated CD40 expression. In contrast, actual HCV viraemia, but not mere exposure, led to alterations within myeloid immune cell populations, natural killer (NK) cells and a distinct soluble factor profile with increased levels of inflammatory cytokines and chemokines. CONCLUSION Our data reveal that exposure to, and infection with, HCV causes disparate effects on adaptive B cells and innate immune cell such as myeloid cells and NK cells in infants.
Collapse
Affiliation(s)
- Anton Lutckii
- Department of Laboratory Medicine, Karolinska institutet, Stockholm, Sweden.,Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russian Federation
| | - Benedikt Strunz
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anton Zhirkov
- Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russian Federation
| | - Olga Filipovich
- North-Western State Medical University named after I.I.Mechnikov, Saint Petersburg, Russian Federation
| | - Elena Rukoiatkina
- Maternity Hospital No 16, Saint Petersburg, Russian Federation.,Department of Pediatrics, Gynecology and Female Reproductology, Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russian Federation
| | - Denis Gusev
- Center for Prevention and Control of AIDS and Infectious Diseases, Saint Petersburg, Russian Federation
| | - Yuriy Lobzin
- Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russian Federation
| | - Björn Fischler
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Department of Pediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Matti Sällberg
- Department of Laboratory Medicine, Karolinska institutet, Stockholm, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination. Int J Mol Sci 2020; 21:ijms21228524. [PMID: 33198297 PMCID: PMC7696930 DOI: 10.3390/ijms21228524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
T follicular helper (Tfh) cells are a specialised subset of CD4+ T cells that play a significant role in the adaptive immune response, providing critical help to B cells within the germinal centres (GC) of secondary lymphoid organs. The B cell receptors of GC B cells undergo multiple rounds of somatic hypermutation and affinity maturation within the GC response, a process dependent on cognate interactions with Tfh cells. B cells that receive sufficient help from Tfh cells form antibody-producing long-lived plasma and memory B cells that provide the basis of decades of effective and efficient protection and are considered the gold standard in correlates of protection post-vaccination. However, the T cell response to vaccination has been understudied, and over the last 10 years, exponential improvements in the technological underpinnings of sampling techniques, experimental and analytical tools have allowed multidisciplinary characterisation of the role of T cells and the immune system as a whole. Of particular interest to the field of vaccinology are GCs and Tfh cells, representing a unique target for improving immunisation strategies. Here, we discuss recent insights into the unique journey of Tfh cells from thymus to lymph node during differentiation and their role in the production of high-quality antibody responses as well as their journey back to the periphery as a population of memory cells. Further, we explore their function in health and disease and the power of next-generation sequencing techniques to uncover their potential as modulators of vaccine-induced immunity.
Collapse
|
29
|
Sun L, Wang X, Saredy J, Yuan Z, Yang X, Wang H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol 2020; 37:101759. [PMID: 33086106 PMCID: PMC7575795 DOI: 10.1016/j.redox.2020.101759] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Innate and adaptive immune cell activation and infiltration is the key characteristic of tissue inflammation. The innate immune system is the front line of host defense in which innate immune cells are activated by danger signals, including pathogen- and danger-associated molecular pattern, and metabolite-associated danger signal. Innate immunity activation can directly contribute to tissue inflammation or immune resolution by phagocytosis and secretion of biologically active molecules, or indirectly via antigen-presenting cell (APC) activation-mediated adaptive immune responses. This review article describes the cellular and molecular interplay of innate-adaptive immune systems. Three major mechanisms are emphasized in this article for their role in facilitating innate-adaptive immunity interplay. 1) APC can be formed from classical and conditional innate immune cells to bridge innate-adaptive immune response. 2) Immune checkpoint molecular pairs connect innate and adaptive immune cells to direct one-way and two-way immune checkpoint reactions. 3) Metabolic reprogramming during immune responses leads to excessive cytosolic and mitochondrial reactive oxygen species (ROS) production. Increased NADPH oxidase-derived extracellular and intracellular ROS are mostly responsible for oxidative stress, which contributes to functional changes in immune cells. Further understanding of innate-adaptive immunity interplay and its underlying molecular basis would lead to the identification of therapeutic targets for immunological and inflammatory disease.
Collapse
Affiliation(s)
- Lizhe Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China; Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xianwei Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jason Saredy
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Arroyo EN, Pepper M. B cells are sufficient to prime the dominant CD4+ Tfh response to Plasmodium infection. J Exp Med 2020; 217:jem.20190849. [PMID: 31748243 PMCID: PMC7041722 DOI: 10.1084/jem.20190849] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Arroyo and Pepper demonstrate that interactions with B cells, not dendritic cells, are required for the priming of the CD4+ T cell response during Plasmodium infection. This results in a Tfh-biased response as reported by others in both mice and humans. CD4+ T follicular helper (Tfh) cells dominate the acute response to a blood-stage Plasmodium infection and provide signals to direct B cell differentiation and protective antibody expression. We studied antigen-specific CD4+ Tfh cells responding to Plasmodium infection in order to understand the generation and maintenance of the Tfh response. We discovered that a dominant, phenotypically stable, CXCR5+ Tfh population emerges within the first 4 d of infection and results in a CXCR5+ CCR7+ Tfh/central memory T cell response that persists well after parasite clearance. We also found that CD4+ T cell priming by B cells was both necessary and sufficient to generate this Tfh-dominant response, whereas priming by conventional dendritic cells was dispensable. This study provides important insights into the development of CD4+ Tfh cells during Plasmodium infection and highlights the heterogeneity of antigen-presenting cells involved in CD4+ T cell priming.
Collapse
Affiliation(s)
- E Nicole Arroyo
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
31
|
Biram A, Shulman Z. T cell help to B cells: Cognate and atypical interactions in peripheral and intestinal lymphoid tissues. Immunol Rev 2020; 296:36-47. [PMID: 32557712 DOI: 10.1111/imr.12890] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Enduring immunity against harmful pathogens depends on the generation of immunological memory. Serum immunoglobulins are constantly secreted by long-lived antibody-producing cells, which provide extended protection from recurrent exposures. These cells originate mainly from germinal center structures, wherein B cells introduce mutations to their immunoglobulin genes followed by affinity-based selection. Generation of high-affinity antibodies relies on physical contacts between T and B cells, a process that facilitates the delivery of fate decision signals. T-B cellular engagements are mediated through interactions between the T cell receptor and its cognate peptide presented on B cell major histocompatibility class II molecules. Here, we describe the cellular and molecular aspects of these cognate T-B interactions, and highlight exceptional cases, especially those arising at intestinal lymphoid organs, at which T cells provide help to B cells in an atypical manner, independent of T cell specificity.
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
32
|
Chulpanova DS, Kitaeva KV, Green AR, Rizvanov AA, Solovyeva VV. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front Cell Dev Biol 2020; 8:402. [PMID: 32582698 PMCID: PMC7283917 DOI: 10.3389/fcell.2020.00402] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Cytokine-based immunotherapy is a promising field in the cancer treatment, since cytokines, as proteins of the immune system, are able to modulate the host immune response toward cancer cell, as well as directly induce tumor cell death. Since a low dose monotherapy with some cytokines has no significant therapeutic results and a high dose treatment leads to a number of side effects caused by the pleiotropic effect of cytokines, the problem of understanding the influence of cytokines on the immune cells involved in the pro- and anti-tumor immune response remains a pressing one. Immune system cells carry CD makers on their surface which can be used to identify various populations of cells of the immune system that play different roles in pro- and anti-tumor immune responses. This review discusses the functions and specific CD markers of various immune cell populations which are reported to participate in the regulation of the immune response against the tumor. The results of research studies and clinical trials investigating the effect of cytokine therapy on the regulation of immune cell populations and their surface markers are also discussed. Current trends in the development of cancer immunotherapy, as well as the role of cytokines in combination with other therapeutic agents, are also discussed.
Collapse
Affiliation(s)
- Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
33
|
Khan S, Liu Y, Ernst LM, Leung LYT, Budylowski P, Dong S, Campisi P, Propst EJ, Wolter NE, Grunebaum E, Ostrowski M, Ehrhardt GRA. Detection of Human CD38 Using Variable Lymphocyte Receptor (VLR) Tetramers. Cells 2020; 9:E950. [PMID: 32290546 PMCID: PMC7226959 DOI: 10.3390/cells9040950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/03/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022] Open
Abstract
CD38 is a multifunctional cell surface receptor expressed on multiple cell lineages of hematopoietic origin with high levels of expression on human plasma cells. Previously, we isolated the monoclonal variable lymphocyte receptor B (VLRB) MM3 antibody from the evolutionarily distant sea lamprey, which recognized the CD38 ectoenzyme exclusively on human plasma cells in a manner that correlated with CD38 enzymatic activity. The plasma cell-specific binding of VLRB MM3 contrasts with the broad pattern of expression of CD38-determined conventional antibodies specific for this antigen. In an effort to facilitate the application of this unique reagent in combination with conventional antibody panels, we explored a strategy to generate VLRB MM3 tetramers. The resulting reagent maintained the threshold-based recognition of CD38. Increased sensitivity achieved with VLRB MM3 tetramers also showed preferential recognition of germinal center centroblasts over centrocytes. VLRB MM3 tetramers thus provided a unique and versatile single-step staining reagent for the detection of human CD38 that is readily incorporated into multi-color flow cytometry panels.
Collapse
Affiliation(s)
- Srijit Khan
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Yanling Liu
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Laura M. Ernst
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Leslie Y. T. Leung
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Patrick Budylowski
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Shilan Dong
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Paolo Campisi
- Department of Otolaryngology—Head and Neck Surgery, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada; (P.C.); (E.J.P.); (N.E.W.)
| | - Evan J. Propst
- Department of Otolaryngology—Head and Neck Surgery, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada; (P.C.); (E.J.P.); (N.E.W.)
| | - Nikolaus E. Wolter
- Department of Otolaryngology—Head and Neck Surgery, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada; (P.C.); (E.J.P.); (N.E.W.)
| | - Eyal Grunebaum
- Division of Immunology and Allergy, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada;
| | - Mario Ostrowski
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| | - Götz R. A. Ehrhardt
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada; (S.K.); (Y.L.); (L.M.E.); (L.Y.T.L.); (S.D.); (M.O.)
| |
Collapse
|
34
|
Grant SM, Lou M, Yao L, Germain RN, Radtke AJ. The lymph node at a glance - how spatial organization optimizes the immune response. J Cell Sci 2020; 133:133/5/jcs241828. [PMID: 32144196 DOI: 10.1242/jcs.241828] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A hallmark of the mammalian immune system is its ability to respond efficiently to foreign antigens without eliciting an inappropriate response to self-antigens. Furthermore, a robust immune response requires the coordination of a diverse range of cells present at low frequencies within the host. This problem is solved, in part, by concentrating antigens, antigen-presenting cells and antigen-responsive cells in lymph nodes (LNs). Beyond housing these cell types in one location, LNs are highly organized structures consisting of pre-positioned cells within well-defined microanatomical niches. In this Cell Science at a Glance article and accompanying poster, we outline the key cellular populations and components of the LN microenvironment that are present at steady state and chronicle the dynamic changes in these elements following an immune response. This review highlights the LN as a staging ground for both innate and adaptive immune responses, while providing an elegant example of how structure informs function.
Collapse
Affiliation(s)
- Spencer M Grant
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Memorial Dr, Bethesda, MD 20892, USA
| | - Meng Lou
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Memorial Dr, Bethesda, MD 20892, USA
| | - Li Yao
- Science Education Department, Howard Hughes Medical Institute, 4000 Jones Bridge Rd, Chevy Chase, MD 20815, USA
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Memorial Dr, Bethesda, MD 20892, USA
| | - Andrea J Radtke
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Memorial Dr, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Esteves de Oliveira E, de Castro E Silva FM, Caçador Ayupe M, Gomes Evangelista Ambrósio M, Passos de Souza V, Costa Macedo G, Ferreira AP. Obesity affects peripheral lymphoid organs immune response in murine asthma model. Immunology 2019; 157:268-279. [PMID: 31112301 DOI: 10.1111/imm.13081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 01/12/2023] Open
Abstract
Asthma and obesity present rising incidence, and their concomitance is a reason for concern, as obese individuals are usually resistant to conventional asthma treatments and have more exacerbation episodes. Obesity affects several features in the lungs during asthma onset, shifting the T helper type 2 (Th2)/eosinophilic response towards a Th17/neutrophilic profile. Moreover, those individuals can present reduced atopy and delayed cytokine production. However, the impact of obesity on follicular helper T (Tfh) cells and B cells that could potentially result in antibody production disturbances are still unclear. Therefore, we aimed to assess the peripheral response to ovalbumin (OVA) in a concomitant model of obesity and asthma. Pulmonary allergy was induced, in both lean and obese female BALB/c mice, through OVA sensitizations and challenges. Mediastinal lymph nodes (MLNs) and spleen were processed for immunophenotyping. Lung was used for standard allergy analysis. Obese-allergic mice produced less anti-OVA IgE and more IgG2a than lean-allergic mice. Dendritic cells (CD11c+ MHCIIhigh ) expressed less CD86 and more PDL1 in obese-allergic mice compared with lean-allergic mice, in the MLNs. Meanwhile, B cells (CD19+ CD40+ ) were more frequent and the amount of PDL1/PD1+ cells was diminished by obesity, with the opposite effects in the spleen. Tfh cells (CD3+ CD4+ CXCR5+ PD1+ ) expressing FoxP3 were more frequent in obese mice, associated with the predominance of Th (CD3+ CD4+ ) cells expressing interleukin-4/GATA3 in the MLNs and interleukin-17A/RORγT in the spleen. Those modifications to the main components of the germinal centers could be resulting in the increased IgG2a production, which - associated with the Th17/neutrophilic profile - contributes to asthma worsening and represents an important target for future treatment strategies.
Collapse
Affiliation(s)
- Erick Esteves de Oliveira
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Flávia Márcia de Castro E Silva
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Marina Caçador Ayupe
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Marcilene Gomes Evangelista Ambrósio
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Viviane Passos de Souza
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Gilson Costa Macedo
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Ana Paula Ferreira
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| |
Collapse
|
36
|
Vacchio MS, Ciucci T, Gao Y, Watanabe M, Balmaceno-Criss M, McGinty MT, Huang A, Xiao Q, McConkey C, Zhao Y, Shetty J, Tran B, Pepper M, Vahedi G, Jenkins MK, McGavern DB, Bosselut R. A Thpok-Directed Transcriptional Circuitry Promotes Bcl6 and Maf Expression to Orchestrate T Follicular Helper Differentiation. Immunity 2019; 51:465-478.e6. [PMID: 31422869 DOI: 10.1016/j.immuni.2019.06.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/08/2019] [Accepted: 06/21/2019] [Indexed: 01/19/2023]
Abstract
The generation of high-affinity neutralizing antibodies, the objective of most vaccine strategies, occurs in B cells within germinal centers (GCs) and requires rate-limiting "help" from follicular helper CD4+ T (Tfh) cells. Although Tfh differentiation is an attribute of MHC II-restricted CD4+ T cells, the transcription factors driving Tfh differentiation, notably Bcl6, are not restricted to CD4+ T cells. Here, we identified a requirement for the CD4+-specific transcription factor Thpok during Tfh cell differentiation, GC formation, and antibody maturation. Thpok promoted Bcl6 expression and bound to a Thpok-responsive region in the first intron of Bcl6. Thpok also promoted the expression of Bcl6-independent genes, including the transcription factor Maf, which cooperated with Bcl6 to mediate the effect of Thpok on Tfh cell differentiation. Our findings identify a transcriptional program that links the CD4+ lineage with Tfh differentiation, a limiting factor for efficient B cell responses, and suggest avenues to optimize vaccine generation.
Collapse
Affiliation(s)
- Melanie S Vacchio
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yayi Gao
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Masashi Watanabe
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Mariah Balmaceno-Criss
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Mitchell T McGinty
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Allan Huang
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Qi Xiao
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Cameron McConkey
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yongmei Zhao
- Center for Cancer Research Sequencing Facility, Advanced Technology Research Facility, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Jyoti Shetty
- Center for Cancer Research Sequencing Facility, Advanced Technology Research Facility, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Bao Tran
- Center for Cancer Research Sequencing Facility, Advanced Technology Research Facility, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Golnaz Vahedi
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
37
|
Breed ER, Watanabe M, Hogquist KA. Measuring Thymic Clonal Deletion at the Population Level. THE JOURNAL OF IMMUNOLOGY 2019; 202:3226-3233. [PMID: 31010850 DOI: 10.4049/jimmunol.1900191] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/25/2019] [Indexed: 12/18/2022]
Abstract
Clonal deletion of T cells specific for self-antigens in the thymus has been widely studied, primarily by approaches that focus on a single receptor (using TCR transgenes) or a single specificity (using peptide-MHC tetramers). However, less is known about clonal deletion at the population level. In this article, we report an assay that measures cleaved caspase 3 to define clonal deletion at the population level. This assay distinguishes clonal deletion from apoptotic events caused by neglect and approximates the anatomic site of deletion using CCR7. This approach showed that 78% of clonal deletion events occur in the cortex in mice. Medullary deletion events were detected at both the semimature and mature stages, although mature events were associated with failed regulatory T cell induction. Using this assay, we showed that bone marrow-derived APC drive approximately half of deletion events at both stages. We also found that both cortical and medullary deletion rely heavily on CD28 costimulation. These findings demonstrate a useful strategy for studying clonal deletion within the polyclonal repertoire.
Collapse
Affiliation(s)
- Elise R Breed
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Masashi Watanabe
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
38
|
Karbalaei Zadeh Babaki M, Taghiabadi M, Soleimanpour S, Saleh Moghadam M, Mosavat A, Amini AA, Mohammadi A, Rezaee SA. Mycobacterium tuberculosis Ag85b:hfcγ1 recombinant fusion protein as a selective receptor-dependent delivery system for antigen presentation. Microb Pathog 2019; 129:68-73. [DOI: 10.1016/j.micpath.2019.01.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 02/03/2023]
|
39
|
Maul J, Alterauge D, Baumjohann D. Micro
RNA
‐mediated regulation of T follicular helper and T follicular regulatory cell identity. Immunol Rev 2019; 288:97-111. [DOI: 10.1111/imr.12735] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Julia Maul
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| | - Dominik Alterauge
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| | - Dirk Baumjohann
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| |
Collapse
|
40
|
Abstract
Dendritic cells (DCs) can be viewed as translators between innate and adaptive immunity. They integrate signals derived from tissue infection or damage and present processed antigen from these sites to naive T cells in secondary lymphoid organs while also providing multiple soluble and surface-bound signals that help to guide T cell differentiation. DC-mediated tailoring of the appropriate T cell programme ensures a proper cascade of immune responses that adequately targets the insult. Recent advances in our understanding of the different types of DC subsets along with the cellular organization and orchestration of DC and lymphocyte positioning in secondary lymphoid organs over time has led to a clearer understanding of how the nature of the T cell response is shaped. This Review discusses how geographical organization and ordered sequences of cellular interactions in lymph nodes and the spleen regulate immunity.
Collapse
Affiliation(s)
- S C Eisenbarth
- Department of Laboratory Medicine, Immunobiology, Section of Allergy & Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
41
|
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: 1.0] [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
|
42
|
Pedros C, Altman A, Kong KF. Role of TRAFs in Signaling Pathways Controlling T Follicular Helper Cell Differentiation and T Cell-Dependent Antibody Responses. Front Immunol 2018; 9:2412. [PMID: 30405612 PMCID: PMC6204373 DOI: 10.3389/fimmu.2018.02412] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/28/2018] [Indexed: 01/02/2023] Open
Abstract
Follicular helper T (TFH) cells represent a highly specialized CD4+ T cell subpopulation that supports the generation of germinal centers (GC) and provides B cells with critical signals promoting antibody class switching, generation of high affinity antibodies, and memory formation. TFH cells are characterized by the expression of the chemokine receptor CXCR5, the transcription factor Bcl-6, costimulatory molecules ICOS, and PD-1, and the production of cytokine IL-21. The acquisition of a TFH phenotype is a complex and multistep process that involves signals received through engagement of the TCR along with a multitude of costimulatory molecules and cytokines receptors. Members of the Tumor necrosis factor Receptor Associated Factors (TRAF) represent one of the major classes of signaling mediators involved in the differentiation and functions of TFH cells. TRAF molecules are the canonical adaptor molecules that physically interact with members of the Tumor Necrosis Factor Receptor Superfamily (TNFRSF) and actively modulate their downstream signaling cascades through their adaptor function and/or E3 ubiquitin ligase activity. OX-40, GITR, and 4-1BB are the TRAF-dependent TNFRSF members that have been implicated in the differentiation and functions of TFH cells. On the other hand, emerging data demonstrate that TRAF proteins also participate in signaling from the TCR and CD28, which deliver critical signals leading to the differentiation of TFH cells. More intriguingly, we recently showed that the cytoplasmic tail of ICOS contains a conserved TANK-binding kinase 1 (TBK1)-binding motif that is shared with TBK1-binding TRAF proteins. The presence of this TRAF-mimicking signaling module downstream of ICOS is required to mediate the maturation step during TFH differentiation. In addition, JAK-STAT pathways emanating from IL-2, IL-6, IL-21, and IL-27 cytokine receptors affect TFH development, and crosstalk between TRAF-mediated pathways and the JAK-STAT pathways can contribute to generate integrated signals required to drive and sustain TFH differentiation. In this review, we will introduce the molecular interactions and the major signaling pathways controlling the differentiation of TFH cells. In each case, we will highlight the contributions of TRAF proteins to these signaling pathways. Finally, we will discuss the role of individual TRAF proteins in the regulation of T cell-dependent humoral responses.
Collapse
Affiliation(s)
- Christophe Pedros
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| |
Collapse
|
43
|
Krishnaswamy JK, Alsén S, Yrlid U, Eisenbarth SC, Williams A. Determination of T Follicular Helper Cell Fate by Dendritic Cells. Front Immunol 2018; 9:2169. [PMID: 30319629 PMCID: PMC6170619 DOI: 10.3389/fimmu.2018.02169] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/03/2018] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T cells that collaborate with B cells to promote and regulate humoral responses. Unlike other CD4+ effector lineages, Tfh cells require interactions with both dendritic cells (DCs) and B cells to complete their differentiation. While numerous studies have assessed the potential of different DC subsets to support Tfh priming, the conclusions of these studies depend heavily on the model and method of immunization used. We propose that the location of different DC subsets within the lymph node (LN) and their access to antigen determine their potency in Tfh priming. Finally, we provide a three-step model that accounts for the ability of multiple DC subsets and related lineages to support the Tfh differentiation program.
Collapse
Affiliation(s)
| | - Samuel Alsén
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States.,Department of Genetics and Genomic Sciences, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
44
|
Muller J, Baeyens A, Dustin ML. Tumor Necrosis Factor Receptor Superfamily in T Cell Priming and Effector Function. Adv Immunol 2018; 140:21-57. [PMID: 30366518 DOI: 10.1016/bs.ai.2018.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor necrosis factor receptor superfamily (TNFRSF) and their ligands mediate lymphoid tissue development and homeostasis in addition to key aspects of innate and adaptive immune responses. T cells of the adaptive immune system express a number of TNFRSF members that are used to receive signals at different instructive stages and produce several tumor necrosis factor superfamily (TNFSF) members as effector molecules. There is also one example of a TNFRSF member serving as a ligand for negative regulatory checkpoint receptors. In most cases, the ligands in afferent and efferent phases are membrane proteins and thus the interaction with TNFRSF members must take place in immunological synapses and other modes of cell-cell interaction. A particular feature of the TNFRSF-mediated signaling is the prominent use of linear ubiquitin chains as scaffolds for signaling complexes that activate nuclear factor κ-B and Fos/Jun transcriptional regulators. This review will focus on the signaling mechanisms triggered by TNFRSF members in their role as costimulators of early and late phases of T cell instruction and the delivery mechanism of TNFSF members through the immunological synapses of helper and cytotoxic effector cells.
Collapse
Affiliation(s)
- James Muller
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Audrey Baeyens
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States; Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
45
|
Wing JB, Tekgüç M, Sakaguchi S. Control of Germinal Center Responses by T-Follicular Regulatory Cells. Front Immunol 2018; 9:1910. [PMID: 30197643 PMCID: PMC6117393 DOI: 10.3389/fimmu.2018.01910] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Regulatory T-cells (Treg cells), expressing the transcription factor Foxp3, have an essential role in the control of immune homeostasis. In order to control diverse types of immune responses Treg cells must themselves show functional heterogeneity to control different types of immune responses. Recent advances have made it clear that Treg cells are able to mirror the homing capabilities of known T-helper subtypes such as Th1, Th2, Th17, and T-follicular helper cells (Tfh), allowing them to travel to the sites of inflammation and deliver suppression in situ. One of the more recent discoveries in this category is the description of T-follicular regulatory (Tfr) cells, a specialized subset of Treg cells that control Tfh and resulting antibody responses. In this review we will discuss recent advances in our understanding of Tfr biology and the role of both Tfr and activated extra-follicular Tregs (eTreg) in the control of humoral immunity.
Collapse
Affiliation(s)
- James B Wing
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Murat Tekgüç
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Fähnrich A, Klein S, Sergé A, Nyhoegen C, Kombrink S, Möller S, Keller K, Westermann J, Kalies K. CD154 Costimulation Shifts the Local T-Cell Receptor Repertoire Not Only During Thymic Selection but Also During Peripheral T-Dependent Humoral Immune Responses. Front Immunol 2018; 9:1019. [PMID: 29867987 PMCID: PMC5966529 DOI: 10.3389/fimmu.2018.01019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
CD154 is a transmembrane cytokine expressed transiently on activated CD4 T cells upon T-cell receptor (TCR) stimulation that interacts with CD40 on antigen-presenting cells. The signaling via CD154:CD40 is essential for B-cell maturation and germinal center formation and also for the final differentiation of CD4 T cells during T-dependent humoral immune responses. Recent data demonstrate that CD154 is critically involved in the selection of T-cell clones during the negative selection process in the thymus. Whether CD154 signaling influences the TCR repertoire during peripheral T-dependent humoral immune responses has not yet been elucidated. To find out, we used CD154-deficient mice and assessed the global TCRβ repertoire in T-cell zones (TCZ) of spleens by high-throughput sequencing after induction of a Th2 response to the multiepitopic antigen sheep red blood cells. Qualitative and quantitative comparison of the splenic TCZ-specific TCRβ repertoires revealed that CD154 deficiency shifts the distribution of Vβ-Jβ genes after antigen exposure. This data led to the conclusion that costimulation via CD154:CD40 during the interaction of T cells with CD40-matured B cells contributes to the recruitment of T-cell clones into the immune response and thereby shapes the peripheral TCR repertoire.
Collapse
Affiliation(s)
- Anke Fähnrich
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Sebastian Klein
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Arnauld Sergé
- Centre de Recherche en Cancérologie de Marseille (CRCM) U1068 INSERM - UMR7258 CNRS - Institut Paoli Calmette, Aix-Marseille University, UM105, Marseille, France
| | | | - Sabrina Kombrink
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Karsten Keller
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | | | - Kathrin Kalies
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| |
Collapse
|
47
|
Hos BJ, Tondini E, van Kasteren SI, Ossendorp F. Approaches to Improve Chemically Defined Synthetic Peptide Vaccines. Front Immunol 2018; 9:884. [PMID: 29755468 PMCID: PMC5932164 DOI: 10.3389/fimmu.2018.00884] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022] Open
Abstract
Progress made in peptide-based vaccinations to induce T-cell-dependent immune responses against cancer has invigorated the search for optimal vaccine modalities. Design of new vaccine strategies intrinsically depends on the knowledge of antigen handling and optimal epitope presentation in both major histocompatibility complex class I and -II molecules by professional antigen-presenting cells to induce robust CD8 and CD4 T-cell responses. Although there is a steady increase in the understanding of the underlying mechanisms that bridges innate and adaptive immunology, many questions remain to be answered. Moreover, we are in the early stage of exploiting this knowledge to clinical advantage. Several adaptations of peptide-based vaccines like peptide-adjuvant conjugates have been explored and showed beneficial outcomes in preclinical models; but in the clinical trials conducted so far, mixed results were obtained. A major limiting factor to unravel antigen handling mechanistically is the lack of tools to efficiently track peptide vaccines at the molecular and (sub)cellular level. In this mini-review, we will discuss options to develop molecular tools for improving, as well as studying, peptide-based vaccines.
Collapse
Affiliation(s)
- Brett J Hos
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sander I van Kasteren
- Leiden Institute of Chemistry, The Institute for Chemical Immunology, Leiden University, Leiden, Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|