101
|
Vu MN, Kelly HG, Tan H, Juno JA, Esterbauer R, Davis TP, Truong NP, Wheatley AK, Kent SJ. Hemagglutinin Functionalized Liposomal Vaccines Enhance Germinal Center and Follicular Helper T Cell Immunity. Adv Healthc Mater 2021; 10:e2002142. [PMID: 33690985 PMCID: PMC8206650 DOI: 10.1002/adhm.202002142] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Despite remarkable successes of immunization in protecting public health, safe and effective vaccines against a number of life-threatening pathogens such as HIV, ebola, influenza, and SARS-CoV-2 remain urgently needed. Subunit vaccines can avoid potential toxicity associated with traditional whole virion-inactivated and live-attenuated vaccines; however, the immunogenicity of subunit vaccines is often poor. A facile method is here reported to produce lipid nanoparticle subunit vaccines that exhibit high immunogenicity and elicit protection against influenza virus. Influenza hemagglutinin (HA) immunogens are functionalized on the surface of liposomes via stable metal chelation chemistry, using a scalable advanced microfluidic mixing technology (NanoAssemblr). Immunization of mice with HA-liposomes elicits increased serum antibody titers and superior protection against highly pathogenic virus challenge compared with free HA protein. HA-liposomal vaccines display enhanced antigen deposition into germinal centers within the draining lymph nodes, driving increased HA-specific B cell, and follicular helper T cell responses. This work provides mechanistic insights into highly protective HA-liposome vaccines and informs the rational design and rapid production of next generation nanoparticle subunit vaccines.
Collapse
Affiliation(s)
- Mai N. Vu
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
- Department of PharmaceuticsHanoi University of PharmacyHanoi10000Vietnam
| | - Hannah G. Kelly
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
| | - Hyon‐Xhi Tan
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
| | - Jennifer A. Juno
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
| | - Robyn Esterbauer
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
| | - Thomas P. Davis
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Australia Institute of Bioengineering & NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
| | - Nghia P. Truong
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Adam K. Wheatley
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
| | - Stephen J. Kent
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVIC3000Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVIC3004Australia
| |
Collapse
|
102
|
Liu Y, Yang CL, Yang B, Du T, Li XL, Zhang P, Ge MR, Lian Y, Li H, Liu YD, Duan RS. Prophylactic administration of fingolimod (FTY720) ameliorated experimental autoimmune myasthenia gravis by reducing the number of dendritic cells, follicular T helper cells and antibody-secreting cells. Int Immunopharmacol 2021; 96:107511. [PMID: 33915521 DOI: 10.1016/j.intimp.2021.107511] [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: 12/21/2020] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 11/28/2022]
Abstract
Fingolimod (FTY720), a sphingosine 1-phosphate (S1P) receptor antagonist, possesses potent immunomodulatory activity via lymphocyte homing. The effects of FTY720 have been widely studied in various T-cell-mediated autoimmune diseases, while the immunomodulatory effects on experimental autoimmune myasthenia gravis (EAMG), a typical disease model for antibody-mediated autoimmunity, remain elusive. In the present study, FTY720 was administered to EAMG rats as prophylaxis. The clinical scores were recorded every other day, and serum antibodies at different time points were measured by enzyme-linked immunosorbent assay (ELISA). The immune cell subsets in the spleen, bone marrow, circulation, and thymus were determined by flow cytometry. The prophylactic administration alleviated EAMG symptoms by reducing the level of serum antibodies IgG and its isotype IgG2b on days 30 and 46 post immunization, as well as IgG and Ig kappa antibody-secreting cells in the spleen and bone marrow. The mitigated humoral immune response can be attributed to the decreased dendritic cells, follicular T help cells (Tfh) and Tfh subsets (Tfh1, Tfh2, and Tfh17), and T helper cell subsets (Th1, Th2, and Th17) in the spleen. The promotion of lymphocyte homing and inhibition of thymocyte egress contribute to the effects of FTY720 on these effector T cell subsets. Overall, the prophylactic administration of FTY720 ameliorated EAMG partially by regulating humoral immune response,suggesting that FTY720 could be part of a pharmacological strategy for managing myasthenia gravis.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China.
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Bing Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Meng-Ru Ge
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China.
| | - Ying Lian
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Yu-Dong Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Rui-Sheng Duan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China; Shandong Institute of Neuroimmunology, China.
| |
Collapse
|
103
|
Baumjohann D, Fazilleau N. Antigen-dependent multistep differentiation of T follicular helper cells and its role in SARS-CoV-2 infection and vaccination. Eur J Immunol 2021; 51:1325-1333. [PMID: 33788271 PMCID: PMC8250352 DOI: 10.1002/eji.202049148] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 01/20/2023]
Abstract
T follicular helper (Tfh) cells play an essential role in regulating the GC reaction and, consequently, the generation of high‐affinity antibodies and memory B cells. Therefore, Tfh cells are critical for potent humoral immune responses against various pathogens and their dysregulation has been linked to autoimmunity and cancer. Tfh cell differentiation is a multistep process, in which cognate interactions with different APC types, costimulatory and coinhibitory pathways, as well as cytokines are involved. However, it is still not fully understood how a subset of activated CD4+ T cells begins to express the Tfh cell‐defining chemokine receptor CXCR5 during the early stage of the immune response, how some CXCR5+ pre‐Tfh cells enter the B‐cell follicles and mature further into GC Tfh cells, and how Tfh cells are maintained in the memory compartment. In this review, we discuss recent advances on how antigen and cognate interactions are important for Tfh cell differentiation and long‐term persistence of Tfh cell memory, and how this is relevant to the current understanding of COVID‐19 pathogenesis and the development of potent SARS‐CoV‐2 vaccines.
Collapse
Affiliation(s)
- Dirk Baumjohann
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, Inserm, Toulouse, U1291, France.,French Germinal Center Club, French Society for Immunology (SFI), Paris, France
| |
Collapse
|
104
|
Scholz J, Kuhrau J, Heinrich F, Heinz GA, Hutloff A, Worm M, Heine G. Vitamin A controls the allergic response through T follicular helper cell as well as plasmablast differentiation. Allergy 2021; 76:1109-1122. [PMID: 32895937 DOI: 10.1111/all.14581] [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: 03/24/2020] [Revised: 07/16/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Vitamin A regulates the adaptive immune response and a modulatory impact on type I allergy is discussed. The cellular mechanisms are largely unknown. OBJECTIVE To determine the vitamin A-responding specific lymphocyte reaction in vivo. METHODS Antigen-specific B and T lymphocytes were analyzed in an adoptive transfer airway inflammation mouse model in response to 9-cis retinoic acid (9cRA) and after lymphocyte-specific genetic targeting of the receptor RARα. Flow cytometry, quantitative PCR, next-generation sequencing, and specific Ig-ELISA were used to characterize the cells functionally. RESULTS Systemic 9cRA profoundly enhanced the specific IgA-secreting B-cell frequencies in the lung tissue and serum IgA while reducing serum IgE concentrations. RARα overexpression in antigen-specific B cells promoted differentiation into plasmablasts at the expense of germinal center B cells. In antigen-specific T cells, RARα strongly promoted the differentiation of T follicular helper cells followed by an enhanced germinal center response. CONCLUSIONS 9cRA signaling via RARα impacts the allergen-specific immunoglobulin response directly by the differentiation of B cells and indirectly by promoting T follicular helper cells.
Collapse
Affiliation(s)
- Josephine Scholz
- Division of Allergy and Immunology Department of Dermatology, Venereology and Allergy Charité – Universitätsmedizin Berlin Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
| | - Julia Kuhrau
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
- Institute of Immunology University Hospital Schleswig‐HolsteinCampus Kiel Kiel Germany
| | - Frederik Heinrich
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
| | - Gitta Anne Heinz
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
| | - Andreas Hutloff
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
- Institute of Immunology University Hospital Schleswig‐HolsteinCampus Kiel Kiel Germany
- Institute of Clinical Molecular Biology University Hospital Schleswig‐HolsteinCampus Kiel Kiel Germany
| | - Margitta Worm
- Division of Allergy and Immunology Department of Dermatology, Venereology and Allergy Charité – Universitätsmedizin Berlin Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
| | - Guido Heine
- Division of Allergy and Immunology Department of Dermatology, Venereology and Allergy Charité – Universitätsmedizin Berlin Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Deutsches Rheuma‐Forschungszentrum, A Leibniz Institute Berlin Germany
- Department of Dermatology and Allergy University Hospital Schleswig‐HolsteinCampus Kiel Kiel Germany
| |
Collapse
|
105
|
Yin M, Xiong Y, Liang D, Tang H, Hong Q, Liu G, Zeng J, Lian T, Huang J, Ni J. Circulating Tfh cell and subsets distribution are associated with low-responsiveness to hepatitis B vaccination. Mol Med 2021; 27:32. [PMID: 33794763 PMCID: PMC8015036 DOI: 10.1186/s10020-021-00290-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/09/2021] [Indexed: 12/22/2022] Open
Abstract
Background An estimated 5–10 % of healthy vaccinees lack adequate antibody response following receipt of a standard three-dose hepatitis B vaccination regimen. The cellular mechanisms responsible for poor immunological responses to hepatitis B vaccine have not been fully elucidated to date. Methods There were 61 low responders and 56 hyper responders involved in our study. Peripheral blood samples were mainly collected at D7, D14 and D28 after revaccinated with a further dose of 20 µg of recombinant hepatitis B vaccine. Results We found low responders to the hepatitis B vaccine presented lower frequencies of circulating follicular helper T (cTfh) cells, plasmablasts and a profound skewing away from cTfh2 and cTfh17 cells both toward cTfh1 cells. Importantly, the skewing of Tfh cell subsets correlated with IL-21 and protective antibody titers. Based on the key role of microRNAs involved in Tfh cell differentiation, we revealed miR-19b-1 and miR-92a-1 correlated with the cTfh cell subsets distribution and antibody production. Conclusions Our findings highlighted a decrease in cTfh cells and specific subset skewing contribute to reduced antibody responses in low responders. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00290-7.
Collapse
Affiliation(s)
- Mingjuan Yin
- Department of Preventive Medicine, Guangdong Medical University, Dongguan, China
| | - Yongzhen Xiong
- School Clinic, Guangdong Medical University, Dongguan, China
| | - Dongmei Liang
- Department of Epidemiology and Biostatistics, Guangdong Medical University, No.1 Xincheng Road, 523808, Dongguan, China
| | - Hao Tang
- Teaching&Research Department, Dongguan Guancheng Hospital, Dongguan, China
| | - Qian Hong
- Dongguan Guancheng Hospital, Dongguan, China
| | - Gang Liu
- Department of Immunization Program, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jinmei Zeng
- Department of Epidemiology and Biostatistics, Guangdong Medical University, No.1 Xincheng Road, 523808, Dongguan, China
| | - Tingyu Lian
- Department of Epidemiology and Biostatistics, Guangdong Medical University, No.1 Xincheng Road, 523808, Dongguan, China
| | - Jingxiao Huang
- Department of Epidemiology and Biostatistics, Guangdong Medical University, No.1 Xincheng Road, 523808, Dongguan, China
| | - Jindong Ni
- Department of Epidemiology and Biostatistics, Guangdong Medical University, No.1 Xincheng Road, 523808, Dongguan, China.
| |
Collapse
|
106
|
Sharma J, Collins TD, Roach T, Mishra S, Lam BK, Mohamed ZS, Veal AE, Polk TB, Jones A, Cornaby C, Haider MI, Zeumer-Spataro L, Johnson HM, Morel LM, Larkin J. Suppressor of cytokine signaling-1 mimetic peptides attenuate lymphocyte activation in the MRL/lpr mouse autoimmune model. Sci Rep 2021; 11:6354. [PMID: 33737712 PMCID: PMC7973732 DOI: 10.1038/s41598-021-86017-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/09/2021] [Indexed: 12/30/2022] Open
Abstract
Autoimmune diseases are driven largely by a pathogenic cytokine milieu produced by aberrantly activated lymphocytes. Many cytokines, including interferon gamma (IFN-γ), utilize the JAK/STAT pathway for signal propagation. Suppressor of Cytokine Signaling-1 (SOCS1) is an inducible, intracellular protein that regulates IFN-γ signaling by dampening JAK/STAT signaling. Using Fas deficient, MRL/MpJ-Faslpr/J (MRL/lpr) mice, which develop lupus-like disease spontaneously, we tested the hypothesis that a peptide mimic of the SOCS1 kinase inhibitory region (SOCS1-KIR) would inhibit lymphocyte activation and modulate lupus-associated pathologies. Consistent with in vitro studies, SOCS1-KIR intraperitoneal administration reduced the frequency, activation, and cytokine production of memory CD8+ and CD4+ T lymphocytes within the peripheral blood, spleen, and lymph nodes. In addition, SOCS1-KIR administration reduced lymphadenopathy, severity of skin lesions, autoantibody production, and modestly reduced kidney pathology. On a cellular level, peritoneal SOCS1-KIR administration enhanced Foxp3 expression in total splenic and follicular regulatory T cells, reduced the effector memory/naïve T lymphocyte ratio for both CD4+ and CD8+ cells, and reduced the frequency of GL7+ germinal center enriched B cells. Together, these data show that SOCS1-KIR treatment reduced auto-reactive lymphocyte effector functions and suggest that therapeutic targeting of the SOCS1 pathway through peptide administration may have efficacy in mitigating autoimmune pathologies.
Collapse
Affiliation(s)
- Jatin Sharma
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Teresa D Collins
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Tracoyia Roach
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Shiwangi Mishra
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Brandon K Lam
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Zaynab Sidi Mohamed
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Antia E Veal
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Timothy B Polk
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Amari Jones
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Caleb Cornaby
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mohammed I Haider
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Leilani Zeumer-Spataro
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Howard M Johnson
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Laurence M Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Joseph Larkin
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA.
| |
Collapse
|
107
|
Abstract
For over 35 years since Mosmann and Coffman proposed the seminal “type 1 T helper (Th1)/type 2 T helper (Th2)” hypothesis in 1986, the immunological community has appreciated that naïve CD4 T cells need to make important decisions upon their activation, namely to differentiate towards a Th1, Th2, Th17 (interleukin-17-producing T helper), follicular T helper (Tfh), or regulatory T cell (Treg) fate to orchestrate a variety of adaptive immune responses. The major molecular underpinnings of the Th1/Th2 effector fate choice had been initially characterized using excellent reductionist in vitro culture systems, through which the transcription factors T-bet and GATA3 were identified as the master regulators for the differentiation of Th1 and Th2 cells, respectively. However, Th1/Th2 cell differentiation and their cellular heterogeneity are usually determined by a combinatorial expression of multiple transcription factors, particularly in vivo, where dendritic cell (DC) and innate lymphoid cell (ILC) subsets can also influence T helper lineage choices. In addition, inflammatory cytokines that are capable of inducing Th17 cell differentiation are also found to be induced during typical Th1- or Th2-related immune responses, resulting in an alternative differentiation pathway, transiting from a Th17 cell phenotype towards Th1 or Th2 cells. In this review, we will discuss the recent advances in the field, focusing on some new players in the transcriptional network, contributions of DCs and ILCs, and alternative differentiation pathways towards understanding the Th1/Th2 effector choice in vivo.
Collapse
Affiliation(s)
- Matthew J Butcher
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
108
|
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
|
109
|
Alonso GT, Fomin DS, Rizzo LV. Human follicular helper T lymphocytes critical players in antibody responses. EINSTEIN-SAO PAULO 2021; 19:eRB6077. [PMID: 33681888 PMCID: PMC7909002 DOI: 10.31744/einstein_journal/2021rb6077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/30/2020] [Indexed: 11/05/2022] Open
Abstract
Follicular helper T lymphocytes are a subpopulation of CD4+ T lymphocytes initially identified in germinal centers of follicles found in secondary lymphoid organs. The primary function of follicular helper T lymphocytes is to help B lymphocytes' antibody production. Changing of antibody class and affinity, B cell differentiation and memory generation depend on cooperation between follicular helper T lymphocytes and B cells. In blood, follicular helper T lymphocytes are called circulating follicular helper T lymphocytes. They are considered to have specificities similar to those developed in the secondary lymphoid organs. The phenotype of human follicular helper T lymphocytes is given by simultaneous expression of the markers CXCR5, Bcl-6, CD40L, PD-1, and ICOS. In germinal centers, follicular helper T lymphocytes synthesize interleukin 21 as predominant cytokine. In blood, subpopulations of circulating follicular helper T lymphocytes can be recognized, with different expressions of the classical follicular helper T lymphocytes markers and, in addition, can express other markers such as CXCR3 and CCR6. Presently, there is great interest in follicular helper T lymphocytes and circulating follicular helper T lymphocytes in vaccination studies as indicators of immunization efficacy. In addition, follicular helper T lymphocytes are investigated as possible markers of activity in many diseases and potential therapeutic intervention. This short review describes aspects of immunobiology and quantification of follicular helper T lymphocytes and circulating follicular helper T lymphocytes, and presents a few examples of related findings in systemic lupus erythematosus, rheumatoid arthritis, HIV infection and vaccination.
Collapse
|
110
|
Ma X, Nakayamada S. Multi-Source Pathways of T Follicular Helper Cell Differentiation. Front Immunol 2021; 12:621105. [PMID: 33717120 PMCID: PMC7947315 DOI: 10.3389/fimmu.2021.621105] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/14/2021] [Indexed: 12/31/2022] Open
Abstract
T follicular helper (Tfh) cells participate in humoral immune by promoting inflammation and aiding B cells survival, proliferation, maturation, and generation autoantibodies. The plasticity of Tfh cells enables the immune system to adjust the direction of differentiation according to the degree of the immune response, regulate the germinal center (GC) response and maintain homeostasis. Tfh differentiation involves several signaling factors, including multiple cytokines, receptors, transcription factors and genes. The signal transducer and activator of transcription (STAT) family signaling pathways are crucial for Tfh formation. However, because of the multi-factorial and multi-stage features of Tfh differentiation, every STAT member plays a role in Tfh differentiation, but is not completely depended on. With the gradual recognition of different Tfh subsets (Tfh1, Tfh2, Tfh17), the process of Tfh differentiation can no longer be explained by straight-line derivation models. In this review, we summarize the roles of different STATs in mediating Tfh subsets, analyze the contributions of mutual restraint and cooperation among cytokine-STAT signals to terminal Tfh differentiation, and clarify the multi-source pathways of Tfh differentiation with a three-dimensional illustration.
Collapse
Affiliation(s)
- Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China.,Department of Microbiology & Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
111
|
Bugya Z, Prechl J, Szénási T, Nemes É, Bácsi A, Koncz G. Multiple Levels of Immunological Memory and Their Association with Vaccination. Vaccines (Basel) 2021; 9:174. [PMID: 33669597 PMCID: PMC7922266 DOI: 10.3390/vaccines9020174] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022] Open
Abstract
Immunological memory is divided into many levels to counteract the provocations of diverse and ever-changing infections. Fast functions of effector memory and the superposition of both quantitatively and qualitatively plastic anticipatory memory responses together form the walls of protection against pathogens. Here we provide an overview of the role of different B and T cell subsets and their interplay, the parallel and independent functions of the B1, marginal zone B cells, T-independent- and T-dependent B cell responses, as well as functions of central and effector memory T cells, tissue-resident and follicular helper T cells in the memory responses. Age-related limitations in the immunological memory of these cell types in neonates and the elderly are also discussed. We review how certain aspects of immunological memory and the interactions of components can affect the efficacy of vaccines, in order to link our knowledge of immunological memory with the practical application of vaccination.
Collapse
Affiliation(s)
- Zsófia Bugya
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (Z.B.); (T.S.); (A.B.)
| | - József Prechl
- R&D Laboratory, Diagnosticum Zrt, H-1047 Budapest, Hungary;
| | - Tibor Szénási
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (Z.B.); (T.S.); (A.B.)
| | - Éva Nemes
- Clinical Center, Department of Pediatrics, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (Z.B.); (T.S.); (A.B.)
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (Z.B.); (T.S.); (A.B.)
| |
Collapse
|
112
|
Ioannidou K, Ndiaye DR, Noto A, Fenwick C, Fortis SP, Pantaleo G, Petrovas C, de Leval L. In Situ Characterization of Follicular Helper CD4 T Cells Using Multiplexed Imaging. Front Immunol 2021; 11:607626. [PMID: 33633728 PMCID: PMC7901994 DOI: 10.3389/fimmu.2020.607626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/24/2020] [Indexed: 12/19/2022] Open
Abstract
Follicular helper CD4 T (Tfh) cells play an essential role in the formation of germinal centers (GCs), where mature B cells proliferate, differentiate, and provide long-term protective humoral responses. Despite the extensive phenotypic characterization and identification of human Tfh cell subsets, their spatial positioning at tissue level is not well understood. Here, we describe a quantitative multiplexed immunofluorescence approach allowing for the comprehensive in situ characterization of Tfh cells in human tonsils and lymph nodes (LNs) from individuals with angioimmunoblastic T-cell lymphoma (AITL). We have developed eight multiplexed panels comprising a spectrum of Tfh cell markers, like PD-1, CXCR5, and ICOS, along with transcription factors (Bcl6, Tbet, GATA3), to assess their expression, frequencies, spatial distribution and co-localization in a quantitative manner. Combined analysis of relevant markers revealed the presence of several Tfh cell subsets at tissue level based on the differential expression of surface receptors, nuclear factors as well as their distinct localization within the follicular areas. Interestingly, we found a considerable amount of tonsillar Tfh cells expressing high levels of the Th2 regulator GATA3. The co-expression of GATA3, CXCR5, and BCL6, points to an important role of GATA3 for the generation of effector human Tfh cells. Furthermore, our data revealed significantly different Tfh cell profile signatures between health and disease. Therefore, our imaging platform generates meaningful information for the in situ characterization of human Tfh cells and could provide the base for future studies aiming to a comprehensive understanding of Tfh cell tissue heterogeneity.
Collapse
Affiliation(s)
- Kalliopi Ioannidou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Daba-Rokhya Ndiaye
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sotirios P Fortis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.,Tissue Analysis Core, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| |
Collapse
|
113
|
Richardson JR, Schöllhorn A, Gouttefangeas C, Schuhmacher J. CD4+ T Cells: Multitasking Cells in the Duty of Cancer Immunotherapy. Cancers (Basel) 2021; 13:596. [PMID: 33546283 PMCID: PMC7913359 DOI: 10.3390/cancers13040596] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy activates the immune system to specifically target malignant cells. Research has often focused on CD8+ cytotoxic T cells, as those have the capacity to eliminate tumor cells after specific recognition upon TCR-MHC class I interaction. However, CD4+ T cells have gained attention in the field, as they are not only essential to promote help to CD8+ T cells, but are also able to kill tumor cells directly (via MHC-class II dependent recognition) or indirectly (e.g., via the activation of other immune cells like macrophages). Therefore, immunotherapy approaches have shifted from only stimulating CD8+ T cells to targeting and assessing both, CD4+ and CD8+ T cell subsets. Here, we discuss the various subsets of CD4+ T cells, their plasticity and functionality, their relevance in the antitumor immune response in patients affected by cancer, and their ever-growing role in therapeutic approaches for human cancer.
Collapse
Affiliation(s)
- Jennifer R. Richardson
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
| | - Anna Schöllhorn
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany
| | - Juliane Schuhmacher
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
114
|
Webb LMC, Fra‐Bido S, Innocentin S, Matheson LS, Attaf N, Bignon A, Novarino J, Fazilleau N, Linterman MA. Ageing promotes early T follicular helper cell differentiation by modulating expression of RBPJ. Aging Cell 2021; 20:e13295. [PMID: 33387451 PMCID: PMC7811847 DOI: 10.1111/acel.13295] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/17/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Ageing profoundly changes our immune system and is thought to be a driving factor in the morbidity and mortality associated with infectious disease in older people. We have previously shown that the impaired immunity to vaccination that occurs in aged individuals is partly attributed to the effect of age on T follicular helper (Tfh) cell formation. In this study, we examined how age intrinsically affects Tfh cell formation in both mice and humans. We show increased formation of Tfh precursors (pre-Tfh) but no associated increase in germinal centre (GC)-Tfh cells in aged mice, suggesting age-driven promotion of only early Tfh cell differentiation. Mechanistically, we show that ageing alters TCR signalling which drives expression of the Notch-associated transcription factor, RBPJ. Genetic or chemical modulation of RBPJ or Notch rescues this age-associated early Tfh cell differentiation, and increased intrinsic Notch activity recapitulates this phenomenon in younger mice. Our data offer mechanistic insight into the age-induced changes in T-cell activation that affects the differentiation and ultimately the function of effector T cells.
Collapse
Affiliation(s)
- Louise M. C. Webb
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteBabrahamUK
| | - Sigrid Fra‐Bido
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteBabrahamUK
| | - Silvia Innocentin
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteBabrahamUK
| | - Louise S. Matheson
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteBabrahamUK
| | - Noudjoud Attaf
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity)Inserm U1291University of ToulouseToulouse, FCNRS U5282France
| | - Alexandre Bignon
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteBabrahamUK
| | - Julien Novarino
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity)Inserm U1291University of ToulouseToulouse, FCNRS U5282France
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity)Inserm U1291University of ToulouseToulouse, FCNRS U5282France
| | | |
Collapse
|
115
|
Abstract
Follicular helper T (Tfh) cells play a key role in B cell activation and differentiation. Within recent years, distinct subsets of follicular T cells, including regulatory and cytotoxic T cells, have been identified. Apart from classical Tfh cells in secondary lymphoid organs, Tfh-like cells are found in chronically inflamed nonlymphoid tissues. Here, we provide protocols to identify different follicular T cell subsets in murine and human tissues by flow cytometry. This chapter also contains an immunization protocol for the induction of large numbers of Tfh cells in mice.
Collapse
|
116
|
邵宽 芙, 关 凤, 董 晨. [Clinical effect and mechanism of total glucosides of paeony in the adjuvant therapy for children with Henoch-Schönlein purpura nephritis: a prospective randomized controlled study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:49-54. [PMID: 33476537 PMCID: PMC7818153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/17/2020] [Indexed: 09/20/2023]
Abstract
OBJECTIVE To study the clinical effect and mechanism of total glucosides of paeony (TGP) in the adjuvant therapy for children with Henoch-Schönlein purpura nephritis (HSPN). METHODS Sixty-four HSPN children with moderate proteinuria were divided into a TGP treatment group (n=34) and a routine treatment group (n=30) using a random number table. Thirty healthy children who underwent physical examination were enrolled as the healthy control group. The children in the routine treatment group were given conventional treatment alone, and those in the observation group were given TGP in addition to the conventional treatment. The two groups were compared in the clinical outcome after 4 weeks of treatment. The proportion of follicular helper T (Tfh) cells in peripheral blood and the plasma levels of interleukin-21 (IL-21) and interleukin-4 (IL-4) were measured in the healthy control group and the two HSPN groups. The changes in serum cystatin C (CysC) level and urinary alpha 1-microglobulin (A1M) concentration were compared before and after treatment in the two HSPN groups. RESULTS Compared with the healthy children before treatment, the children with HSPN had higher proportion of Tfh cells and expression levels of IL-21 and IL-4 (P < 0.01). The TGP treatment group had a higher overall response rate to treatment than the routine treatment group (94% vs 67%, P < 0.05). After treatment, both groups had reductions in the proportion of Tfh cells in peripheral blood, the expression levels of IL-21, IL-4, serum CysC, and urinary A1M concentration. The TGP treatment group had greater reductions in these indices than the routine treatment group (P < 0.01). CONCLUSIONS TGP has a marked clinical effect in the treatment of HSPN and can reduce the inflammatory response of the kidney and exert a protective effect on the kidney by inhibiting the proliferation of Tfh cells and downregulating the expression of IL-21 and IL-4 in plasma.
Collapse
Affiliation(s)
- 芙蓉 邵宽
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - 凤军 关
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - 晨 董
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| |
Collapse
|
117
|
邵宽 芙, 关 凤, 董 晨. [Clinical effect and mechanism of total glucosides of paeony in the adjuvant therapy for children with Henoch-Schönlein purpura nephritis: a prospective randomized controlled study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:49-54. [PMID: 33476537 PMCID: PMC7818153 DOI: 10.7499/j.issn.1008-8830.2008135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/17/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To study the clinical effect and mechanism of total glucosides of paeony (TGP) in the adjuvant therapy for children with Henoch-Schönlein purpura nephritis (HSPN). METHODS Sixty-four HSPN children with moderate proteinuria were divided into a TGP treatment group (n=34) and a routine treatment group (n=30) using a random number table. Thirty healthy children who underwent physical examination were enrolled as the healthy control group. The children in the routine treatment group were given conventional treatment alone, and those in the observation group were given TGP in addition to the conventional treatment. The two groups were compared in the clinical outcome after 4 weeks of treatment. The proportion of follicular helper T (Tfh) cells in peripheral blood and the plasma levels of interleukin-21 (IL-21) and interleukin-4 (IL-4) were measured in the healthy control group and the two HSPN groups. The changes in serum cystatin C (CysC) level and urinary alpha 1-microglobulin (A1M) concentration were compared before and after treatment in the two HSPN groups. RESULTS Compared with the healthy children before treatment, the children with HSPN had higher proportion of Tfh cells and expression levels of IL-21 and IL-4 (P < 0.01). The TGP treatment group had a higher overall response rate to treatment than the routine treatment group (94% vs 67%, P < 0.05). After treatment, both groups had reductions in the proportion of Tfh cells in peripheral blood, the expression levels of IL-21, IL-4, serum CysC, and urinary A1M concentration. The TGP treatment group had greater reductions in these indices than the routine treatment group (P < 0.01). CONCLUSIONS TGP has a marked clinical effect in the treatment of HSPN and can reduce the inflammatory response of the kidney and exert a protective effect on the kidney by inhibiting the proliferation of Tfh cells and downregulating the expression of IL-21 and IL-4 in plasma.
Collapse
Affiliation(s)
- 芙蓉 邵宽
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - 凤军 关
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - 晨 董
- />徐州医科大学附属医院儿科, 江苏徐州 221000Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| |
Collapse
|
118
|
Negron A, Stüve O, Forsthuber TG. Ectopic Lymphoid Follicles in Multiple Sclerosis: Centers for Disease Control? Front Neurol 2020; 11:607766. [PMID: 33363512 PMCID: PMC7753025 DOI: 10.3389/fneur.2020.607766] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
While the contribution of autoreactive CD4+ T cells to the pathogenesis of Multiple Sclerosis (MS) is widely accepted, the advent of B cell-depleting monoclonal antibody (mAb) therapies has shed new light on the complex cellular mechanisms underlying MS pathogenesis. Evidence supports the involvement of B cells in both antibody-dependent and -independent capacities. T cell-dependent B cell responses originate and take shape in germinal centers (GCs), specialized microenvironments that regulate B cell activation and subsequent differentiation into antibody-secreting cells (ASCs) or memory B cells, a process for which CD4+ T cells, namely follicular T helper (TFH) cells, are indispensable. ASCs carry out their effector function primarily via secreted Ig but also through the secretion of both pro- and anti-inflammatory cytokines. Memory B cells, in addition to being capable of rapidly differentiating into ASCs, can function as potent antigen-presenting cells (APCs) to cognate memory CD4+ T cells. Aberrant B cell responses are prevented, at least in part, by follicular regulatory T (TFR) cells, which are key suppressors of GC-derived autoreactive B cell responses through the expression of inhibitory receptors and cytokines, such as CTLA4 and IL-10, respectively. Therefore, GCs represent a critical site of peripheral B cell tolerance, and their dysregulation has been implicated in the pathogenesis of several autoimmune diseases. In MS patients, the presence of GC-like leptomeningeal ectopic lymphoid follicles (eLFs) has prompted their investigation as potential sources of pathogenic B and T cell responses. This hypothesis is supported by elevated levels of CXCL13 and circulating TFH cells in the cerebrospinal fluid (CSF) of MS patients, both of which are required to initiate and maintain GC reactions. Additionally, eLFs in post-mortem MS patient samples are notably devoid of TFR cells. The ability of GCs to generate and perpetuate, but also regulate autoreactive B and T cell responses driving MS pathology makes them an attractive target for therapeutic intervention. In this review, we will summarize the evidence from both humans and animal models supporting B cells as drivers of MS, the role of GC-like eLFs in the pathogenesis of MS, and mechanisms controlling GC-derived autoreactive B cell responses in MS.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Neurology Section, Veterans Affairs North Texas Health Care System, Medical Service, Dallas, TX, United States
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
119
|
Holstein J, Solimani F, Baum C, Meier K, Pollmann R, Didona D, Tekath T, Dugas M, Casadei N, Hudemann C, Polakova A, Matthes J, Schäfer I, Yazdi AS, Eming R, Hertl M, Pfützner W, Ghoreschi K, Möbs C. Immunophenotyping in pemphigus reveals a T H17/T FH17 cell-dominated immune response promoting desmoglein1/3-specific autoantibody production. J Allergy Clin Immunol 2020; 147:2358-2369. [PMID: 33221382 DOI: 10.1016/j.jaci.2020.11.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 10/19/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND TH2 cells were thought to be a pivotal factor for initiation of the autoimmune blistering disease pemphigus. However, the role of other T-cell subsets in pemphigus pathogenesis remained unclear. OBJECTIVE We aimed to characterize the exact phenotype of T cells responsible for the development of pemphigus. METHODS Whole transcriptome shotgun sequencing was performed to determine differential gene expression in pemphigus lesions and skin of healthy individuals. The cutaneous cytokine signature was further evaluated by real-time quantitative PCR. In peripheral blood, the distribution of TH cell and folliclular helper (TFH) cell subsets was analyzed by flow cytometry. Finally, the capacity of TH and TFH cell subsets to induce desmoglein (Dsg)-specific autoantibodies by memory B cells was evaluated in coculture experiments. RESULTS Transcriptome analysis of skin samples identified an IL-17A-dominated immune signature in patients with pemphigus, and Kyoto Encyclopedia of Genes and Genomes pathway analysis confirmed the dominance of the IL-17A signaling pathway. Increased expression of IL17A and associated cytokines was also detected by real-time quantitative PCR comparing lesional with perilesional or healthy skin. Interestingly, utilization of flow cytometry showed that patients with active pemphigus had elevated levels of circulating IL-17+, TH17, TFH17, and TFH17.1 cells. Notably, levels of TH17 and TFH17 cells correlated with levels of Dsg-specific CD19+CD27+ memory B cells, and patients with acute pemphigus showed higher levels of Dsg3-autoreactive TFH17 cells. Coculture experiments revealed TFH17 cells as primarily responsible for inducing Dsg-specific autoantibody production by B cells. CONCLUSION Our findings show that TFH17 cells are critically involved in the pathogenesis of pemphigus and offer novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Julia Holstein
- Department of Dermatology, University Medical Center, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Carolin Baum
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Katharina Meier
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Pollmann
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Dario Didona
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Tobias Tekath
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Christoph Hudemann
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Alexandra Polakova
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Jakob Matthes
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Iris Schäfer
- Department of Dermatology, University Medical Center, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Amir S Yazdi
- Department of Dermatology and Allergology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Wolfgang Pfützner
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| | - Christian Möbs
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
120
|
Scarsella L, Pollmann R, Amber KT. Autoreactive T cells in pemphigus: perpetrator and target. Ital J Dermatol Venerol 2020; 156:124-133. [PMID: 33179878 DOI: 10.23736/s2784-8671.20.06706-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pemphigus vulgaris (PV) is an autoimmune blistering disease, in which autoantibodies against epidermal cadherins, such as desmoglein (Dsg)1 and Dsg3, lead to the development of blisters and erosions on the skin and mucous membranes. Autoreactive CD4+ T cells are essential for the induction and perpetuation of the disease by interaction with B cells producing autoantibodies. PV has a strong genetic association with certain human leucocyte antigen (HLA) alleles with HLA-DRB1*04:02 and LA-DQB1*05:03 being the most prevalent in patients. Recently, genome-wide association studies have provided a new approach to identify single nucleotide polymorphisms, alongside the known association with HLA alleles. Loss of tolerance against Dsgs and other autoantigens is a critical event in the pathogenesis of PV. Epitope spreading contributes to the progression of PV, leading to an extension of the Dsg-specific autoimmune response to other molecular epitopes of autoantigens, such as desmocollins or muscarinic receptors. Alterations in CD4+CD25+ FoxP3+ regulatory T cells are thought to contribute to the development of PV representing a suitable target for therapeutic interventions. Several CD4+ T-cell subsets and cytokines are involved in the pathogenesis of PV, while Th2 cells are the extensively studied population. Recently, other T cell subsets like T follicular helper cells and Th17 have gained attention as new potential players in PV pathogenesis. The involvement of local autoantibody production in the lesional skin of PV patients in tertiary lymphoid organs is currently discussed but not yet clarified. In this study, we reviewed the current knowledge about the development, characteristics and function of autoreactive T cells in pemphigus and present current new T cell-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Luca Scarsella
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - Robert Pollmann
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany -
| | - Kyle T Amber
- Department of Dermatology, University of Illinois, Chicago, IL, USA
| |
Collapse
|
121
|
Abstract
PURPOSE OF REVIEW To summarize recent studies elucidating the roles of follicular T cells in controlling allospecific antibody responses and antibody-mediated rejection (AbMR). RECENT FINDINGS The field of antibody regulation has provided an in depth identification of the T-cell subsets involved in regulation of antibody responses. In addition, tools have been developed to study these cells during disease. Over the past few years, these strategies have been implemented in the field of transplantation to study the roles of T cells in mediating pathogenic antibody responses. SUMMARY AbMR is largely responsible for long-term graft failure after solid organ transplantation and is induced by allospecific antibodies. In vaccination and infection, antiboody responses are controlled by humoral immunoregulation in which T follicular helper (Tfh) cells promote, and T follicular regulatory (Tfr) cells inhibit, antibody responses. Recent studies have suggested multifaceted roles for follicular T-cell subsets in regulating allospecific antibody responses and AbMR during organ transplantation. In addition, we discuss research priorities for the field to help elucidate mechanisms used by these cells so that new targeted therapeutics can be developed to prevent AbMR in human organ transplantation.
Collapse
|
122
|
DeGottardi Q, Gates TJ, Yang J, James EA, Malhotra U, Chow IT, Simoni Y, Fehlings M, Newell EW, DeBerg HA, Kwok WW. Ontogeny of different subsets of yellow fever virus-specific circulatory CXCR5 + CD4 + T cells after yellow fever vaccination. Sci Rep 2020; 10:15686. [PMID: 32973217 PMCID: PMC7519049 DOI: 10.1038/s41598-020-72610-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/02/2020] [Indexed: 11/09/2022] Open
Abstract
Monitoring the frequency of circulatory CXCR5+ (cCXCR5+) CD4+ T cells in periphery blood provides a potential biomarker to draw inferences about T follicular helper (TFH) activity within germinal center. However, cCXCR5+ T cells are highly heterogeneous in their expression of ICOS, PD1 and CD38 and the relationship between different cCXCR5 subsets as delineated by these markers remains unclear. We applied class II tetramer reagents and mass cytometry to investigate the ontogeny of different subsets of cCXCR5+ T cell following yellow fever immunization. Through unsupervised analyses of mass cytometry data, we show yellow fever virus-specific cCXCR5 T cells elicited by vaccination were initially CD38+ICOS+PD1+, but then transitioned to become CD38+ICOS-PD1+ and CD38-ICOS-PD1+ before coming to rest as a CD38-ICOS-PD1- subset. These results imply that most antigen-specific cCXCR5+ T cells, including the CD38-ICOS-PD1- CXCR5+ T cells are derived from the CXCR5+CD38+ICOS+PD1+ subset, the subset that most resembles preTFH/TFH in the germinal center.
Collapse
Affiliation(s)
- Quinn DeGottardi
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA.,Adaptive Biotechnologies, Seattle, WA, USA
| | - Theresa J Gates
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA
| | - Junbao Yang
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA.,Cs-Bay Therapeutics, Newark, CA, USA
| | - Eddie A James
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA
| | - Uma Malhotra
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA.,Virginia Mason Hospital, Seattle, WA, USA
| | - I-Ting Chow
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA
| | - Yannick Simoni
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael Fehlings
- Singapore Immunology Network, Agency for Science Research and Technology, Singapore, Singapore
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hannah A DeBerg
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA
| | - William W Kwok
- Benaroya Research Institute At Virginia Mason Medical Center, 1201, 9th Ave, Seattle, WA, 98101, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
123
|
Liu H, Li R, Liu T, Yang L, Yin G, Xie Q. Immunomodulatory Effects of Mesenchymal Stem Cells and Mesenchymal Stem Cell-Derived Extracellular Vesicles in Rheumatoid Arthritis. Front Immunol 2020; 11:1912. [PMID: 32973792 PMCID: PMC7468450 DOI: 10.3389/fimmu.2020.01912] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/16/2020] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints and other organs for which there is currently no effective treatment. Mesenchymal stem cells (MSCs) have therapeutic potential due to their immunomodulatory and differentiation effects. While extensive experimental studies and clinical trials have demonstrated the effects of MSCs in various diseases, MSCs have been found to cause abnormal differentiation and tumor formation. Therefore, extracellular vesicles derived from MSCs (MSC-EVs) are more effective, less toxic, and more stable than the parental cells. MSC-EVs transfer various nucleic acids, proteins, and lipids from parent cells to recipient cells, and thus participate in chronic inflammatory and immune processes. In this review, we summarize the properties and biological functions of MSCs and MSC-EVs in RA. Improvement in our understanding of the mechanisms underlying MSC and MSC-EVs in RA provides an insight into potential biomarkers and therapeutic strategies for RA.
Collapse
Affiliation(s)
- Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruicen Li
- Health Management Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Leiyi Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
124
|
Janssen E, Tohme M, Butts J, Giguere S, Sage PT, Velázquez FE, Kam C, Milin E, Das M, Sobh A, Al-Tamemi S, Luscinskas FW, Batista F, Geha RS. DOCK8 is essential for LFA-1-dependent positioning of T follicular helper cells in germinal centers. JCI Insight 2020; 5:134508. [PMID: 32573493 DOI: 10.1172/jci.insight.134508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 06/18/2020] [Indexed: 01/07/2023] Open
Abstract
T follicular helper (Tfh) cell migration into germinal centers (GCs) is essential for the generation of GC B cells and antibody responses to T cell-dependent (TD) antigens. This process requires interactions between lymphocyte function-associated antigen 1 (LFA-1) on Tfh cells and ICAMs on B cells. The mechanisms underlying defective antibody responses to TD antigens in DOCK8 deficiency are incompletely understood. We show that mice selectively lacking DOCK8 in T cells had impaired IgG antibody responses to TD antigens, decreased GC size, and reduced numbers of GC B cells. However, they developed normal numbers of Tfh cells with intact capacity for driving B cell differentiation into a GC phenotype in vitro. Notably, migration of DOCK8-deficient T cells into GCs was defective. Following T cell receptor (TCR)/CD3 ligation, DOCK8-deficient T cells had impaired LFA-1 activation and reduced binding to ICAM-1. Our results therefore indicate that DOCK8 is important for LFA-1-dependent positioning of Tfh cells in GCs, and thereby the generation of GC B cells and IgG antibody responses to TD antigen.
Collapse
Affiliation(s)
- Erin Janssen
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mira Tohme
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jordan Butts
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sophie Giguere
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Francisco E Velázquez
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Departments of Pathology and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Christy Kam
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Elena Milin
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mrinmoy Das
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ali Sobh
- Department of Pediatrics, Mansoura University Children's Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Francis W Luscinskas
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Departments of Pathology and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Facundo Batista
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
125
|
Donnadieu E, Reisinger KB, Scharf S, Michel Y, Bein J, Hansen S, Loth AG, Flinner N, Hartmann S, Hansmann ML. Landscape of T Follicular Helper Cell Dynamics in Human Germinal Centers. THE JOURNAL OF IMMUNOLOGY 2020; 205:1248-1255. [PMID: 32699157 DOI: 10.4049/jimmunol.1901475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/28/2020] [Indexed: 11/19/2022]
Abstract
T follicular helper (Tfh) cells play a very important role in mounting a humoral response. Studies conducted in mouse models have revealed with good kinetic and spatial resolution the dynamics of these cells in germinal centers (GC) and their cross-talk with B cells upon an immune response. However, whether a similar migratory behavior is performed by human Tfh cells is unclear, as technology to track them in situ has been lacking. In this study, we combined traditional immunohistochemistry and real-time fluorescent imaging approaches on fresh human adenoid slices to provide static and dynamic information on Tfh cells. Our data indicate that GC light zones are composed of two distinct areas in terms of Tfh cell distribution and migration. In the outer GC light zones, Tfh cells migrate actively and with a high ability to form dynamic clusters showing intense and rapid reorganization. In these outer regions, Tfh cells demonstrate multiple interactions between each other. Conversely, in central regions of GC light zones, Tfh cells are much more static, forming long-lasting conjugates. These findings reveal for the first time, to our knowledge, the dynamic behavior whereby Tfh cells migrate in human GC and highlight the heterogeneity of GC for Tfh cell motility.
Collapse
Affiliation(s)
- Emmanuel Donnadieu
- Département Immunologie, Inflammation, et Infection, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Université de Paris, F-75014 Paris, France;
| | | | - Sonja Scharf
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Yvonne Michel
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Julia Bein
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany
| | - Susanne Hansen
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Andreas G Loth
- Department of Otolaryngology, Head and Neck Surgery, University Hospital, 60590 Frankfurt am Main, Germany; and
| | - Nadine Flinner
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt, Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany
| | - Martin-Leo Hansmann
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany.,Frankfurt Institute for Advanced Studies, 60438 Frankfurt, Germany
| |
Collapse
|
126
|
Verstegen RHJ, Kusters MAA. Inborn Errors of Adaptive Immunity in Down Syndrome. J Clin Immunol 2020; 40:791-806. [PMID: 32638194 DOI: 10.1007/s10875-020-00805-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
Down syndrome fits an immunophenotype of combined immunodeficiency with immunodysregulation, manifesting with increased susceptibility to infections, autoimmunity, autoinflammatory diseases, and hematologic malignancies. Qualitative and quantitative alterations in innate and adaptive immunity are found in most individuals with Down syndrome. However, there is substantial heterogeneity and no correlation between immunophenotype and clinical presentation. Previously, it was thought that the immunological changes in Down syndrome were caused by precocious aging. We emphasize in this review that the immune system in Down syndrome is intrinsically different from the very beginning. The overexpression of specific genes located on chromosome 21 contributes to immunodeficiency and immunodysregulation, but gene expression differs between genes located on chromosome 21 and depends on tissue and cell type. In addition, trisomy 21 results in gene dysregulation of the whole genome, reflecting the complex nature of this syndrome in comparison to well-known inborn errors of immunity that result from monogenic germline mutations. In this review, we provide an updated overview focusing on inborn errors of adaptive immunity in Down syndrome.
Collapse
Affiliation(s)
- Ruud H J Verstegen
- Division of Clinical Pharmacology and Toxicology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada. .,Division of Rheumatology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Maaike A A Kusters
- Department of Immunology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,University College London Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
127
|
Su C, Johnson ME, Torres A, Thomas RM, Manduchi E, Sharma P, Mehra P, Le Coz C, Leonard ME, Lu S, Hodge KM, Chesi A, Pippin J, Romberg N, Grant SFA, Wells AD. Mapping effector genes at lupus GWAS loci using promoter Capture-C in follicular helper T cells. Nat Commun 2020; 11:3294. [PMID: 32620744 PMCID: PMC7335045 DOI: 10.1038/s41467-020-17089-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 06/02/2020] [Indexed: 01/14/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is mediated by autoreactive antibodies that damage multiple tissues. Genome-wide association studies (GWAS) link >60 loci with SLE risk, but the causal variants and effector genes are largely unknown. We generated high-resolution spatial maps of SLE variant accessibility and gene connectivity in human follicular helper T cells (TFH), a cell type required for anti-nuclear antibodies characteristic of SLE. Of the ~400 potential regulatory variants identified, 90% exhibit spatial proximity to genes distant in the 1D genome sequence, including variants that loop to regulate the canonical TFH genes BCL6 and CXCR5 as confirmed by genome editing. SLE 'variant-to-gene' maps also implicate genes with no known role in TFH/SLE disease biology, including the kinases HIPK1 and MINK1. Targeting these kinases in TFH inhibits production of IL-21, a cytokine crucial for class-switched B cell antibodies. These studies offer mechanistic insight into the SLE-associated regulatory architecture of the human genome.
Collapse
Affiliation(s)
- Chun Su
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Matthew E Johnson
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Annabel Torres
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Rajan M Thomas
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Elisabetta Manduchi
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania, 3700 Hamilton Walk, Philadelphia, PA, USA
| | - Prabhat Sharma
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Parul Mehra
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Carole Le Coz
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Michelle E Leonard
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Sumei Lu
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Kenyaita M Hodge
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Alessandra Chesi
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - James Pippin
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Neil Romberg
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Struan F A Grant
- Division of Human Genetics, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Division of Diabetes and Endocrinology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA
| | - Andrew D Wells
- Department of Pathology, The Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, USA.
| |
Collapse
|
128
|
Yin M, Xiong Y, Huang L, Liu G, Yu Z, Zhao Y, Zhao J, Zhang Y, Lian T, Huang J, Liang D, Zeng J, Ni J. Circulating follicular helper T cells and subsets are associated with immune response to hepatitis B vaccination. Hum Vaccin Immunother 2020; 17:566-574. [PMID: 32614645 DOI: 10.1080/21645515.2020.1775457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Around 5-10% of healthy vaccinees lack or produce an inadequate antibody response following receipt of a standard hepatitis B vaccination regimen. Studying immune response to hepatitis B vaccination could promote researches of immunological events contributing to this poor response. To address this, we investigated follicular helper T (Tfh) cells and firstly demonstrated similar kinetics between circulating Tfh (cTfh) cells and Tfh cells derived from mice spleen after hepatitis B vaccination. And cTfh cells were positively associated with anti-HBs at one week after vaccination (D7). Furthermore, we found PBMCs stimulated by HBsAg showed preferential activation of CXCR3- Tfh cells subsets in vitro. The expression of transcription factor BCL6 in CD4+ T cell significantly differed between D7 and four weeks after vaccination (D28). However, dynamic curve of CD19+ B cells tended to rise then fall but no significant trends were observed. Our findings revealed a decrease in cTfh cells and subset skewing contribute to reduced antibody responses in immune response to hepatitis B vaccination, which indicated the importance of Tfh cell in facilitating the optimization of vaccine efficacy.
Collapse
Affiliation(s)
- Mingjuan Yin
- Department of Preventive Medicine, Guangdong Medical University , Dongguan, China.,Maternal and Child Research Institute, Women and Children's Hospital Affiliated to Guangdong Medical University (Shunde District Maternal and Child Health Hospital) , Foshan, China
| | - Yongzhen Xiong
- School Clinic, Guangdong Medical University , Dongguan, China
| | - Lingfeng Huang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Gang Liu
- Department of Immunization Program, Shenzhen Center for Disease Control and Prevention , Shenzhen, China
| | - Zuwei Yu
- Public Health Office, Dalang Town Community Health Service Center , Dongguan, China
| | - Yi Zhao
- Microbiology and Immunology, Guangdong Medical University , Dongguan, China
| | - Jie Zhao
- Neonatal Department, Shenzhen Maternal and Child Health Hospital Affiliated to Southern Medical University , Shenzhen, China
| | - Yan Zhang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Tingyu Lian
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Jingxiao Huang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - DongMei Liang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - JinMei Zeng
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Jindong Ni
- Maternal and Child Research Institute, Women and Children's Hospital Affiliated to Guangdong Medical University (Shunde District Maternal and Child Health Hospital) , Foshan, China.,Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| |
Collapse
|
129
|
Schijns V, Fernández-Tejada A, Barjaktarović Ž, Bouzalas I, Brimnes J, Chernysh S, Gizurarson S, Gursel I, Jakopin Ž, Lawrenz M, Nativi C, Paul S, Pedersen GK, Rosano C, Ruiz-de-Angulo A, Slütter B, Thakur A, Christensen D, Lavelle EC. Modulation of immune responses using adjuvants to facilitate therapeutic vaccination. Immunol Rev 2020; 296:169-190. [PMID: 32594569 PMCID: PMC7497245 DOI: 10.1111/imr.12889] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/30/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022]
Abstract
Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.
Collapse
Affiliation(s)
- Virgil Schijns
- Wageningen University, Cell Biology & Immunology and, ERC-The Netherlands, Schaijk, Landerd campus, The Netherlands
| | - Alberto Fernández-Tejada
- Chemical Immunology Lab, Center for Cooperative Research in Biosciences, CIC bioGUNE, Biscay, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Žarko Barjaktarović
- Agency for Medicines and Medical Devices of Montenegro, Podgorica, Montenegro
| | - Ilias Bouzalas
- Hellenic Agricultural Organization-DEMETER, Veterinary Research Institute, Thessaloniki, Greece
| | | | - Sergey Chernysh
- Laboratory of Insect Biopharmacology and Immunology, Department of Entomology, Saint-Petersburg State University, Saint-Petersburg, Russia
| | | | | | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Maria Lawrenz
- Vaccine Formulation Institute (CH), Geneva, Switzerland
| | - Cristina Nativi
- Department of Chemistry, University of Florence, Florence, Italy
| | | | | | | | - Ane Ruiz-de-Angulo
- Chemical Immunology Lab, Center for Cooperative Research in Biosciences, CIC bioGUNE, Biscay, Spain
| | - Bram Slütter
- Div. BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
130
|
Gryzik S, Hoang Y, Lischke T, Mohr E, Venzke M, Kadner I, Poetzsch J, Groth D, Radbruch A, Hutloff A, Baumgrass R. Identification of a super-functional Tfh-like subpopulation in murine lupus by pattern perception. eLife 2020; 9:53226. [PMID: 32441253 PMCID: PMC7274784 DOI: 10.7554/elife.53226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/20/2020] [Indexed: 01/20/2023] Open
Abstract
Dysregulated cytokine expression by T cells plays a pivotal role in the pathogenesis of autoimmune diseases. However, the identification of the corresponding pathogenic subpopulations is a challenge, since a distinction between physiological variation and a new quality in the expression of protein markers requires combinatorial evaluation. Here, we were able to identify a super-functional follicular helper T cell (Tfh)-like subpopulation in lupus-prone NZBxW mice with our binning approach "pattern recognition of immune cells (PRI)". PRI uncovered a subpopulation of IL-21+ IFN-γhigh PD-1low CD40Lhigh CXCR5- Bcl-6- T cells specifically expanded in diseased mice. In addition, these cells express high levels of TNF-α and IL-2, and provide B cell help for IgG production in an IL-21 and CD40L dependent manner. This super-functional T cell subset might be a superior driver of autoimmune processes due to a polyfunctional and high cytokine expression combined with Tfh-like properties.
Collapse
Affiliation(s)
- Stefanie Gryzik
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Yen Hoang
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Timo Lischke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Elodie Mohr
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Melanie Venzke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Isabelle Kadner
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Josephine Poetzsch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | | | - Andreas Radbruch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,Charité, Campus Mitte, Berlin, Germany
| | - Andreas Hutloff
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Ria Baumgrass
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| |
Collapse
|
131
|
Bartsch YC, Eschweiler S, Leliavski A, Lunding HB, Wagt S, Petry J, Lilienthal GM, Rahmöller J, de Haan N, Hölscher A, Erapaneedi R, Giannou AD, Aly L, Sato R, de Neef LA, Winkler A, Braumann D, Hobusch J, Kuhnigk K, Krémer V, Steinhaus M, Blanchard V, Gemoll T, Habermann JK, Collin M, Salinas G, Manz RA, Fukuyama H, Korn T, Waisman A, Yogev N, Huber S, Rabe B, Rose-John S, Busch H, Berberich-Siebelt F, Hölscher C, Wuhrer M, Ehlers M. IgG Fc sialylation is regulated during the germinal center reaction following immunization with different adjuvants. J Allergy Clin Immunol 2020; 146:652-666.e11. [PMID: 32445838 DOI: 10.1016/j.jaci.2020.04.059] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS Different adjuvants induce distinct IgG+ GC B-cell responses with specific transcriptomes and expression levels of the α2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3- follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-γ+ TFH1 cells, IL-6/IL-23-dependent IL-17A+ TFH17 cells, and high ratios of TFH cells to Foxp3+ follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.
Collapse
Affiliation(s)
- Yannic C Bartsch
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Simon Eschweiler
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Alexei Leliavski
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hanna B Lunding
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Sander Wagt
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany; Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Janina Petry
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Gina-Maria Lilienthal
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany; Department of Anesthesiology and Intensive Care, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Noortje de Haan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Raghu Erapaneedi
- Institute for Pathology, University of Würzburg, Würzburg, Germany
| | - Anastasios D Giannou
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lilian Aly
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Germany
| | - Ryota Sato
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Louise A de Neef
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - André Winkler
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany; Laboratory of Tolerance and Autoimmunity at the German Rheumatism Research Center, a Leibniz Institute, Berlin, Germany
| | - Dominique Braumann
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany; Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Juliane Hobusch
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Kyra Kuhnigk
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Vanessa Krémer
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Moritz Steinhaus
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Véronique Blanchard
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Timo Gemoll
- Section for Translational Surgical Oncology & Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Jens K Habermann
- Section for Translational Surgical Oncology & Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Mattias Collin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Gabriela Salinas
- NGS-Integrative Genomics, Institute Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Thomas Korn
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Germany; Munich Cluster for Systems Neurology, SyNergy, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nir Yogev
- Clinic and Polyclinic for Dermatology and Venerology, University Hospital Cologne, Cologne, Germany
| | - Samuel Huber
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rabe
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | | | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Friederike Berberich-Siebelt
- Institute for Pathology, University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, Borstel, Germany; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany; Laboratory of Tolerance and Autoimmunity at the German Rheumatism Research Center, a Leibniz Institute, Berlin, Germany; Airway Research Center North, University of Lübeck, German Center for Lung Research, Lübeck, Germany.
| |
Collapse
|
132
|
Kelly HG, Tan HX, Juno JA, Esterbauer R, Ju Y, Jiang W, Wimmer VC, Duckworth BC, Groom JR, Caruso F, Kanekiyo M, Kent SJ, Wheatley AK. Self-assembling influenza nanoparticle vaccines drive extended germinal center activity and memory B cell maturation. JCI Insight 2020; 5:136653. [PMID: 32434990 DOI: 10.1172/jci.insight.136653] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/15/2020] [Indexed: 01/10/2023] Open
Abstract
Protein-based, self-assembling nanoparticles elicit superior immunity compared with soluble protein vaccines, but the immune mechanisms underpinning this effect remain poorly defined. Here, we investigated the immunogenicity of a prototypic ferritin-based nanoparticle displaying influenza hemagglutinin (HA) in mice and macaques. Vaccination of mice with HA-ferritin nanoparticles elicited higher serum antibody titers and greater protection against experimental influenza challenge compared with soluble HA protein. Germinal centers in the draining lymph nodes were expanded and persistent following HA-ferritin vaccination, with greater deposition of antigen that colocalized with follicular dendritic cells. Our findings suggest that a highly ordered and repetitive antigen array may directly drive germinal centers through a B cell-intrinsic mechanism that does not rely on ferritin-specific T follicular helper cells. In contrast to mice, enhanced immunogenicity of HA-ferritin was not observed in pigtail macaques, where antibody titers and lymph node immunity were comparable to soluble vaccination. An improved understanding of factors that drive nanoparticle vaccine immunogenicity in small and large animal models will facilitate the clinical development of nanoparticle vaccines for broad and durable protection against diverse pathogens.
Collapse
Affiliation(s)
- Hannah G Kelly
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| | - Yi Ju
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Department of Chemical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Wenbo Jiang
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | - Brigette C Duckworth
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Joanna R Groom
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Department of Chemical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and
| |
Collapse
|
133
|
Qi H, Tian D, Li M, Zhang C, Jin H, Liu L, Zhao X, Ma L, Zhao W, Ge Q, Duan T, Zhang D. Foxo3 Promotes the Differentiation and Function of Follicular Helper T Cells. Cell Rep 2020; 31:107621. [DOI: 10.1016/j.celrep.2020.107621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 01/27/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022] Open
|
134
|
Liu Z, Wang Y, Li F, Xie F, Liu M, Shi J, Dong N. Circulating follicular T helper cells and humoral reactivity in rheumatic heart disease. Life Sci 2020; 245:117390. [PMID: 32007574 DOI: 10.1016/j.lfs.2020.117390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/16/2020] [Accepted: 01/29/2020] [Indexed: 11/18/2022]
Abstract
AIMS This study aimed to profile circulating T follicular helper cells (cTfh) and their effect on B cells in rheumatic heart disease (RHD). MATERIALS AND METHODS Participants were divided into healthy control (HC, n = 30) and RHD (n = 30) groups. Percentages of cTfh subpopulations, based on CD4, CXCR5, CXCR3, CCR6, Foxp3, Ki67, and PD-1 cell markers, and of CD19+ B cell subgroups were measured by flow cytometry and compared between the groups. Also, IL-21 concentration in plasma and mitral valve were quantitated by cytometric bead array, immunofluorescence, and western blotting. KEY FINDINGS The PD-1+ cTfh, B cells (naive B cells, plasmablasts, and plasma B cells) proportion and cTfh17/cTfh ratios in RHD group were significantly increased, compared to HC (p < 0.01 in all cases), while different types of memory B cells were diminished (p < 0.001). In RHD patients, percentages of PD-1+ cTfh and switched memory B cells were negatively correlated (r = -0.565, p = 0.009); meanwhile, percentages of plasmablasts and PD-1+ cTfh cells were positively correlated (r = 0.594, p = 0.005). Additionally, IL-21 levels in plasma and mitral valve of RHD group were higher than those in HC. Also, IL-21 levels correlated with PD-1+ cTfh(r = 0.557, p = 0.010), cTfh17 (r = 0.567, p = 0.009), and plasmablast (r = -0.5957, p = 0.005) cell proportions, and (cTh2 + cTh17)/cTfh1 ratio (r = -0.547, p = 0.013). SIGNIFICANCE The activation of PD-1+ cTfh and cTfh17 subtype was highly correlated with plasmablast maturation and IL-21 production in rheumatic heart disease. Thus indicating the prominent role of cTfh and humoral reactivity in the immune pathogenesis of RHD.
Collapse
Affiliation(s)
- Zongtao Liu
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yixuan Wang
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Li
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xie
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Liu
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Nianguo Dong
- From the Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
135
|
Choi SC, Morel L. Immune metabolism regulation of the germinal center response. Exp Mol Med 2020; 52:348-355. [PMID: 32132626 PMCID: PMC7156389 DOI: 10.1038/s12276-020-0392-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/16/2019] [Accepted: 01/14/2020] [Indexed: 01/16/2023] Open
Abstract
The humoral immune response requires germinal centers to produce high-affinity antigen-specific antibodies that counter pathogens. Numerous studies have provided a better understanding of how metabolic pathways regulate the development, activation and functions of immune cells. Germinal centers are transient, highly dynamic microanatomic structures that develop in lymphoid organs during a T-cell-dependent humoral immune response. Analysis of germinal centers provides an opportunity to understand how metabolic programs control the differentiation and function of highly specialized germinal center B cells and follicular helper CD4+ T cells. Targeting immunometabolism during the germinal center response may afford the possibility to improve vaccine design and to develop new therapies to alleviate autoimmunity. In this review, we discuss the major metabolic pathways that are used by germinal center B and T cells, as well as the plasma cells that they produce, all of which are influenced by the microenvironment of this unique structure of the adaptive immune system. Studies of the metabolic mechanisms involved in antibody production will inform vaccine design and autoimmune disease treatments. Germinal centers (GCs) are transient sites in lymph nodes and the spleen, formed when white blood cells called T-cell lymphocytes respond to infection. GCs act as factories where another lymphocyte group, B cells, proliferates and mutates before producing infection-appropriate antibodies. GCs therefore play a critical role in adaptive immunity, but the metabolic pathways involved are unclear. Laurence Morel and Seung-Chui Choi at the University of Florida, Gainesville, USA, reviewed understanding of the metabolic pathways used by T cells, B cells and the antibodies they produce. The cells within GCs require different energy sources and metabolic pathways according to their developmental stage, to ensure optimal immune responses. The researchers call for extensive profiling of this complex metabolic system.
Collapse
Affiliation(s)
- Seung-Chul Choi
- 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, USA.
| |
Collapse
|
136
|
Verma A, Schmidt BA, Elizaldi SR, Nguyen NK, Walter KA, Beck Z, Trinh HV, Dinasarapu AR, Lakshmanappa YS, Rane NN, Matyas GR, Rao M, Shen X, Tomaras GD, LaBranche CC, Reimann KA, Foehl DH, Gach JS, Forthal DN, Kozlowski PA, Amara RR, Iyer SS. Impact of T h1 CD4 Follicular Helper T Cell Skewing on Antibody Responses to an HIV-1 Vaccine in Rhesus Macaques. J Virol 2020; 94:e01737-19. [PMID: 31827000 PMCID: PMC7158739 DOI: 10.1128/jvi.01737-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Generating durable humoral immunity through vaccination depends upon effective interactions of follicular helper T (Tfh) cells with germinal center (GC) B cells. Th1 polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing costimulatory and cytokine-dependent Tfh help to B cells. However, the question remains as to whether adjuvant-dependent modulation of Tfh cells enhances HIV-1 vaccine-induced antienvelope (anti-Env) antibody responses. We investigated whether an HIV-1 vaccine platform designed to increase the number of Th1-polarized Tfh cells enhances the magnitude and quality of anti-Env antibodies. Utilizing a novel interferon-induced protein 10 (IP-10)-adjuvanted HIV-1 DNA prime followed by a monophosphoryl lipid A and QS-21 (MPLA+QS-21)-adjuvanted Env protein boost (DIP-10 PALFQ) in macaques, we observed higher anti-Env serum IgG titers with greater cross-clade reactivity, specificity for V1V2, and effector functions than in macaques primed with DNA lacking IP-10 and boosted with MPLA-plus-alum-adjuvanted Env protein (DPALFA) The DIP-10 PALFQ vaccine regimen elicited higher anti-Env IgG1 and lower IgG4 antibody levels in serum, showing for the first time that adjuvants can dramatically impact the IgG subclass profile in macaques. The DIP-10 PALFQ regimen also increased vaginal and rectal IgA antibodies to a greater extent. Within lymph nodes, we observed augmented GC B cell responses and the promotion of Th1 gene expression profiles in GC Tfh cells. The frequency of GC Tfh cells correlated with both the magnitude and avidity of anti-Env serum IgG. Together, these data suggest that adjuvant-induced stimulation of Th1-Tfh cells is an effective strategy for enhancing the magnitude and quality of anti-Env antibody responses.IMPORTANCE The results of the RV144 trial demonstrated that vaccination could prevent HIV transmission in humans and that longevity of anti-Env antibodies may be key to this protection. Efforts to improve upon the prime-boost vaccine regimen used in RV144 have indicated that booster immunizations can increase serum anti-Env antibody titers but only transiently. Poor antibody durability hampers efforts to develop an effective HIV-1 vaccine. This study was designed to identify the specific elements involved in the immunological mechanism necessary to produce robust HIV-1-specific antibodies in rhesus macaques. By clearly defining immune-mediated pathways that improve the magnitude and functionality of the anti-HIV-1 antibody response, we will have the foundation necessary for the rational development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Anil Verma
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Brian A Schmidt
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Sonny R Elizaldi
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- Graduate Group in Immunology, UC Davis, Davis, California, USA
| | - Nancy K Nguyen
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Korey A Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zoltan Beck
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ashok R Dinasarapu
- Emory Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | | | - Niharika N Rane
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Keith A Reimann
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - David H Foehl
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Johannes S Gach
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Smita S Iyer
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- California National Primate Research Center, School of Veterinary Medicine, UC Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
137
|
Duan X, Shen C, Zhang X, Wu L, Chen J, Ma B, Wang Q, Sun P, Lan Y, Su C. Toll-like receptor 7 agonist imiquimod prevents the progression of SLE in MRL/lpr mice via inhibiting the differentiation of T follicular helper cells. Int Immunopharmacol 2020; 80:106239. [PMID: 32007709 DOI: 10.1016/j.intimp.2020.106239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/04/2020] [Accepted: 01/20/2020] [Indexed: 11/29/2022]
Abstract
Previous research has recently indicated that TLR7 is able to induce CD4+T cell anergy, which is the opposite of the role it plays in innate immune cells. Therefore, TLR7 ligands may be used as a manner in which to induce CD4+T cells "tolerance" in autoimmune diseases. T follicular helper (Tfh) cells were demonstrated to be a subset of CD4+T cells that help B cells produce antibodies. The abnormal activity of Tfh cells, though, is their function as a primary pathogenic factor in systemic lupus erythematosus (SLE). However, the role of TLR7 in Tfh cells is not clear. Our study was aimed at determining the influence of TLR7 on Tfh cells in a murine model of SLE (MRL/lpr mice). We were surprised to find that the frequency of Tfh cells and germinal center (GC) B cells was significantly reduced after treatment with the TLR7 agonist imiquimod. Imiquimod also significantly reduced the expression of inducible costimulatory molecule (ICOS) and programmed death 1(PD-1) in Tfh cells and decreased IL-21 secretion. Moreover, imiquimod significantly reduced the mRNA expression of several transcription factors, including Bcl-6, c-Maf, Batf3, Nfatc2 and Stat3, and enhanced the expression of Prdm1 and Stat5b in CD4+T cells. Imiquimod also ameliorated the progression of SLE in MRL/lpr mice by inhibiting anti-dsDNA antibodies and antinuclear antibody (ANA) secretion in the serum. Our findings indicated that TLR7 inhibited the development of Tfh cells both in vivo and ex vivo, which depended on many transcription factors aside from Bcl-6. Our results demonstrated that a TLR7 agonist has the potential to be used to inhibit Tfh cell responses during SLE.
Collapse
Affiliation(s)
- Xiangguo Duan
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, 750004 Yinchuan, PR China; Department of Laboratory Surgery, General Hospital of Ningxia Medical University, 750004 Yinchuan, PR China
| | - Chunxiu Shen
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China
| | - Xiaoyu Zhang
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China
| | - Lihua Wu
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, 750004 Yinchuan, PR China
| | - Jian Chen
- Guolong Hospital, 750004 Yinchuan, PR China
| | - Bin Ma
- Department of Oncology Surgery, The First People's Hospital of Yinchuan, 750001 Yinchuan, PR China
| | - Qi Wang
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China
| | - Peng Sun
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China
| | - Yaru Lan
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China
| | - Chunxia Su
- School of Basic Medical Sciences , Ningxia Medical University, 750004 Yinchuan, PR China.
| |
Collapse
|
138
|
|
139
|
Yi JS, Rosa-Bray M, Staats J, Zakroysky P, Chan C, Russo MA, Dumbauld C, White S, Gierman T, Weinhold KJ, Guptill JT. Establishment of normative ranges of the healthy human immune system with comprehensive polychromatic flow cytometry profiling. PLoS One 2019; 14:e0225512. [PMID: 31825961 PMCID: PMC6905525 DOI: 10.1371/journal.pone.0225512] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Existing normative flow cytometry data have several limitations including small sample sizes, incompletely described study populations, variable flow cytometry methodology, and limited depth for defining lymphocyte subpopulations. To overcome these issues, we defined high-dimensional flow cytometry reference ranges for the healthy human immune system using Human Immunology Project Consortium methodologies after carefully screening 127 subjects deemed healthy through clinical and laboratory testing. We enrolled subjects in the following age cohorts: 18–29 years, 30–39, 40–49, and 50–66 and enrolled cohorts to ensure an even gender distribution and at least 30% non-Caucasians. From peripheral blood mononuclear cells, flow cytometry reference ranges were defined for >50 immune subsets including T-cell (activation, maturation, T follicular helper and regulatory T cell), B-cell, and innate cells. We also developed a web tool for visualization of the dataset and download of raw data. This dataset provides the immunology community with a resource to compare and extract data from rigorously characterized healthy subjects across age groups, gender and race.
Collapse
Affiliation(s)
- John S Yi
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
| | | | - Janet Staats
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
| | - Pearl Zakroysky
- Duke Clinical Research Institute, Durham, NC, United States of America
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States of America
| | - Melissa A Russo
- Department of Neurology, Duke University School of Medicine, Durham, NC, United States of America
| | - Chelsae Dumbauld
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
| | - Scott White
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
| | - Todd Gierman
- Biomat USA-Grifols Plasma Operations, United States of America
| | - Kent J Weinhold
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
| | - Jeffrey T Guptill
- Duke Clinical Research Institute, Durham, NC, United States of America.,Department of Neurology, Duke University School of Medicine, Durham, NC, United States of America
| |
Collapse
|
140
|
Sanz I, Wei C, Jenks SA, Cashman KS, Tipton C, Woodruff MC, Hom J, Lee FEH. Challenges and Opportunities for Consistent Classification of Human B Cell and Plasma Cell Populations. Front Immunol 2019; 10:2458. [PMID: 31681331 PMCID: PMC6813733 DOI: 10.3389/fimmu.2019.02458] [Citation(s) in RCA: 366] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
The increasingly recognized role of different types of B cells and plasma cells in protective and pathogenic immune responses combined with technological advances have generated a plethora of information regarding the heterogeneity of this human immune compartment. Unfortunately, the lack of a consistent classification of human B cells also creates significant imprecision on the adjudication of different phenotypes to well-defined populations. Additional confusion in the field stems from: the use of non-discriminatory, overlapping markers to define some populations, the extrapolation of mouse concepts to humans, and the assignation of functional significance to populations often defined by insufficient surface markers. In this review, we shall discuss the current understanding of human B cell heterogeneity and define major parental populations and associated subsets while discussing their functional significance. We shall also identify current challenges and opportunities. It stands to reason that a unified approach will not only permit comparison of separate studies but also improve our ability to define deviations from normative values and to create a clean framework for the identification, functional significance, and disease association with new populations.
Collapse
Affiliation(s)
- Ignacio Sanz
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Chungwen Wei
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Scott A Jenks
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Kevin S Cashman
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Christopher Tipton
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Matthew C Woodruff
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Jennifer Hom
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - F Eun-Hyung Lee
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States.,Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
141
|
Zhao J, Shi J, Qu M, Zhao X, Wang H, Huang M, Liu Z, Li Z, He Q, Zhang S, Zhang Z. Hyperactive Follicular Helper T Cells Contribute to Dysregulated Humoral Immunity in Patients With Liver Cirrhosis. Front Immunol 2019; 10:1915. [PMID: 31456809 PMCID: PMC6700335 DOI: 10.3389/fimmu.2019.01915] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022] Open
Abstract
Objectives: Liver cirrhosis (LC) is usually accompanied by cirrhosis associated immune dysfunction (CAID), including reduced naïve T cells and memory B cells. However, little is known regarding on follicular helper T (Tfh) cell compartments in cirrhotic patients, especially in the secondary lymphoid organs such as spleen. This study characterizes splenic Tfh cells and explores its association with humoral immunity and disease progression in cirrhotic patients. Methods: Using flow cytometry and histological staining, we analyzed the frequency and cytokine production of splenic Tfh cells from LC patients and healthy controls (HCs). Co-culture experiments of sorted Tfh and B cells were performed for functional analysis in vitro. The correlations between Tfh cells and disease progression markers as well as B cell subset perturbations were also examined. Results: PD-1highICOS+CXCR5+ Tfh cells were preferentially enriched in the spleen of cirrhotic patients, where they expressed higher levels of CXCR3 and produced more interleukin (IL)-21. Histologically, more splenic Tfh cells occupied the B cell follicular structure in LC patients where they shaped more active germinal centers (GCs) than those in HC spleens. In vitro, splenic Tfh cells in cirrhotic patients robustly induce plasma cell differentiation through IL-21 dependent manner. Finally, increased Tfh cell frequency is positively correlated with the plasma cells and disease severity in LC patients. Conclusions: We conclude that hyperactive Tfh cells contribute to dysregulated humoral immunity in patients with liver cirrhosis.
Collapse
Affiliation(s)
- Juanjuan Zhao
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Institute for Hepatology, Shenzhen Third People's Hospital, Shenzhen, China.,Research Center for Clinical & Translational Medicine, Fifth Medical Center for General Hospital of PLA, Beijing, China
| | - Jijing Shi
- The Central Laboratory, The First People's Hospital of Zhengzhou, Zhengzhou, China
| | - Mengmeng Qu
- Research Center for Clinical & Translational Medicine, Fifth Medical Center for General Hospital of PLA, Beijing, China
| | - Xin Zhao
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Department of Surgery, Fifth Medical Center for General Hospital of PLA, Beijing, China
| | - Hongbo Wang
- Department for Liver Transplantation, Fifth Medical Center for General Hospital of PLA, Beijing, China
| | - Man Huang
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Institute for Hepatology, Shenzhen Third People's Hospital, Shenzhen, China
| | - Zhenwen Liu
- Department for Liver Transplantation, Fifth Medical Center for General Hospital of PLA, Beijing, China
| | - Zhiwei Li
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Department for Liver Transplantation, Shenzhen Third People's Hospital, Shenzhen, China
| | - Qing He
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Institute for Hepatology, Shenzhen Third People's Hospital, Shenzhen, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zheng Zhang
- The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.,Institute for Hepatology, Shenzhen Third People's Hospital, Shenzhen, China.,Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.,Key Laboratory of Immunology, School of Basic Medical Sciences, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
142
|
Seth A, Craft J. Spatial and functional heterogeneity of follicular helper T cells in autoimmunity. Curr Opin Immunol 2019; 61:1-9. [PMID: 31374450 DOI: 10.1016/j.coi.2019.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022]
Abstract
Follicular helper T cells provide signals that promote B cell development, proliferation, and production of affinity matured and appropriately isotype switched antibodies. In addition to their classical locations within B cell follicles and germinal centers therein, B cell helper T cells are also found in extrafollicular spaces - either in secondary lymphoid or non-lymphoid tissues. Both follicular and extrafollicular T helper cells drive autoantibody-mediated autoimmunity. Interfering with B cell help provided by T cells can ameliorate autoimmune disease in animal models and human patients. The next frontier in Tfh cell biology will be identification of Tfh cell-specific pathogenic changes in autoimmunity and exploiting them for therapeutic purposes.
Collapse
Affiliation(s)
- Abhinav Seth
- Department of Internal Medicine, Section of Rheumatology, New Haven, CT, United States
| | - Joe Craft
- Department of Internal Medicine, Section of Rheumatology, New Haven, CT, United States; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
143
|
Shao P, Li F, Wang J, Chen X, Liu C, Xue HH. Cutting Edge: Tcf1 Instructs T Follicular Helper Cell Differentiation by Repressing Blimp1 in Response to Acute Viral Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:801-806. [PMID: 31300510 DOI: 10.4049/jimmunol.1900581] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/24/2019] [Indexed: 01/07/2023]
Abstract
Differentiation of T follicular helper (TFH) cells is regulated by a complex transcriptional network, with mutually antagonistic Bcl6-Blimp1 as a core regulatory axis. It is well established that Tcf1 acts upstream of Bcl6 for its optimal induction to program TFH cell differentiation. In this study, we show that whereas genetic ablation of Tcf1 in mice greatly diminished TFH cells in response to viral infection, compound deletion of Blimp1 with Tcf1 restored TFH cell frequency, numbers, and generation of germinal center B cells. Aberrant upregulation of T-bet and Id2 in Tcf1-deficient TFH cells was also largely rectified by ablating Blimp1. Tcf1 chromatin immunoprecipitation sequencing in TFH cells identified two strong Tcf1 binding sites in the Blimp1 gene at a 24-kb upstream and an intron-3 element. Deletion of the intron-3 element, but not the 24-kb upstream element, compromised production of TFH cells. Our data demonstrate that Tcf1-mediated Blimp1 repression is functionally critical for safeguarding TFH cell differentiation.
Collapse
Affiliation(s)
- Peng Shao
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Fengyin Li
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Jinyong Wang
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Xia Chen
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Chengyu Liu
- Transgenic Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Hai-Hui Xue
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; .,Iowa City Veterans Affairs Health Care System, Iowa City, IA 52246
| |
Collapse
|
144
|
Pahwa S. Searching for the origin of the enigmatic circulating T follicular helper cells. J Clin Invest 2019; 129:3048-3051. [PMID: 31264972 DOI: 10.1172/jci130311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
T follicular helper (Tfh) cells in germinal centers of secondary lymphoid organs are pivotal for B and T cell interactions required for induction of humoral immunity. It has long been debated whether Tfh cells exit from lymph nodes into the blood as circulating Tfh cells. In this issue of the JCI, Vella et al. have taken the bull by the horns and applied considerable technical muscle to answer this question. By analyzing phenotype, transcriptome, epigenetic profile, and T cell receptor clonotype, the authors provide evidence that a subset of cTfh cells do indeed originate in lymph nodes and traffic into the blood via the thoracic duct.
Collapse
|