51
|
Peng C, Huggins MA, Wanhainen KM, Knutson TP, Lu H, Georgiev H, Mittelsteadt KL, Jarjour NN, Wang H, Hogquist KA, Campbell DJ, Borges da Silva H, Jameson SC. Engagement of the costimulatory molecule ICOS in tissues promotes establishment of CD8 + tissue-resident memory T cells. Immunity 2022; 55:98-114.e5. [PMID: 34932944 PMCID: PMC8755622 DOI: 10.1016/j.immuni.2021.11.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 08/13/2021] [Accepted: 11/19/2021] [Indexed: 01/13/2023]
Abstract
Elevated gene expression of the costimulatory receptor Icos is a hallmark of CD8+ tissue-resident memory (Trm) T cells. Here, we examined the contribution of ICOS in Trm cell differentiation. Upon transfer into WT mice, Icos-/- CD8+ T cells exhibited defective Trm generation but produced recirculating memory populations normally. ICOS deficiency or ICOS-L blockade compromised establishment of CD8+ Trm cells but not their maintenance. ICOS ligation during CD8+ T cell priming did not determine Trm induction; rather, effector CD8+ T cells showed reduced Trm differentiation after seeding into Icosl-/- mice. IcosYF/YF CD8+ T cells were compromised in Trm generation, indicating a critical role for PI3K signaling. Modest transcriptional changes in the few Icos-/- Trm cells suggest that ICOS-PI3K signaling primarily enhances the efficiency of CD8+ T cell tissue residency. Thus, local ICOS signaling promotes production of Trm cells, providing insight into the contribution of costimulatory signals in the generation of tissue-resident populations.
Collapse
Affiliation(s)
- Changwei Peng
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew A. Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kelsey M. Wanhainen
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Todd P. Knutson
- Minnesota Supercomputing Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Hanbin Lu
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hristo Georgiev
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Current address: Institute of immunology, Hannover Medical School, Hannover D-30625, Germany
| | - Kristen L. Mittelsteadt
- Benaroya Research Institute and Department of Immunology University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Nicholas N. Jarjour
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Haiguang Wang
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristin A. Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel J. Campbell
- Benaroya Research Institute and Department of Immunology University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Henrique Borges da Silva
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Current address: Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Stephen C. Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA,Corresponding author and lead contact:
| |
Collapse
|
52
|
Hart AP, Laufer TM. A review of signaling and transcriptional control in T follicular helper cell differentiation. J Leukoc Biol 2022; 111:173-195. [PMID: 33866600 DOI: 10.1002/jlb.1ri0121-066r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
T follicular helper (Tfh) cells are a critical component of adaptive immunity and assist in optimal Ab-mediated defense. Multiple effector functions of Tfh support germinal center B cell survival, Ab class switching, and plasma cell maturation. In the past 2 decades, the phenotype and functional characteristics of GC Tfh have been clarified allowing for robust studies of the Th subset including activation signals and environmental cues controlling Tfh differentiation and migration during an immune response. A unique, 2-step differentiation process of Tfh has been proposed but the mechanisms underlying transition between unstable Tfh precursors and functional mature Tfh remain elusive. Likewise, newly identified transcriptional regulators of Tfh development have not yet been incorporated into our understanding of how these cells might function in disease. Here, we review the signals and downstream transcription factors that shape Tfh differentiation including what is known about the epigenetic processes that maintain Tfh identity. It is proposed that further evaluation of the stepwise differentiation pattern of Tfh will yield greater insights into how these cells become dysregulated in autoimmunity.
Collapse
Affiliation(s)
- Andrew P Hart
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Terri M Laufer
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Rheumatology, Department of Medicine, Corporal Michael C. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| |
Collapse
|
53
|
Wang HY, Ge W, Liu SQ, Long J, Jiang QQ, Zhou W, Zuo ZY, Liu DY, Zhao HM, Zhong YB. Curcumin Inhibits T Follicular Helper Cell Differentiation in Mice with Dextran Sulfate Sodium (DSS)-Induced Colitis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 50:275-293. [PMID: 34931590 DOI: 10.1142/s0192415x22500100] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Follicular helper T cells (Tfh) regulate the differentiation of germinal center B cells and maintain humoral immunity. Notably, imbalances in Tfh differentiation often lead to the development of autoimmune diseases, including inflammatory bowel disease (IBD). Curcumin, a natural product derived from Curcuma longa, is effective in relieving IBD in humans and animals, and its mechanisms of immune regulation need further elaboration. In this study, dextran sodium sulfate induced ulcerative colitis in BALB/c mice, and curcumin was administered simultaneously for 7 days. Curcumin effectively upregulated the change rate of mouse weight, colonic length, down-regulated colonic weight, index of colonic weight, colonic damage score and the levels of pro-inflammatory cytokines IL-6, IL-12, IL-23 and TGF-[Formula: see text]1 in colonic tissues of colitis mice. Importantly, curcumin regulated the differentiation balance of Tfh and their subpopulation in colitis mice; the percentages of Tfh (CD4[Formula: see text]CXCR5[Formula: see text]BCL-6[Formula: see text], CD4[Formula: see text]CXCR5[Formula: see text]PD-1[Formula: see text], CD4[Formula: see text]CXCR5[Formula: see text]PD-L1[Formula: see text], CD4[Formula: see text]CXCR5[Formula: see text]ICOS[Formula: see text], Tfh17 and Tem-Tfh were downregulated significantly, while CD4[Formula: see text]CXCR5[Formula: see text]Blimp-1[Formula: see text], Tfh1, Tfh10, Tfh21, Tfr, Tcm-Tfh and Tem-GC Tfh were upregulated. In addition, curcumin inhibited the expression of Tfh-related transcription factors BCL-6, p-STAT3, Foxp1, Roquin-1, Roquin-2 and SAP, and significantly upregulated the protein levels of Blimp-1 and STAT3 in colon tissue. In conclusion, curcumin may be effective in alleviating dextran sulfate sodium-induced colitis by regulating Tfh differentiation.
Collapse
Affiliation(s)
- Hai-Yan Wang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi, University of Chinese Medicine, 445 Bayi Avenue, Nanchang 330006, Jiangxi Province, P. R. China
| | - Su-Qing Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Jian Long
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Qing-Qing Jiang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Wen Zhou
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Zheng-Yun Zuo
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - Hai-Mei Zhao
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
| | - You-Bao Zhong
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang 330004, Jiangxi Province, P. R. China
- Department of Proctology, Affiliated Hospital of Jiangxi, University of Chinese Medicine, 445 Bayi Avenue, Nanchang 330006, Jiangxi Province, P. R. China
| |
Collapse
|
54
|
Rauschmeier R, Reinhardt A, Gustafsson C, Glaros V, Artemov AV, Dunst J, Taneja R, Adameyko I, Månsson R, Busslinger M, Kreslavsky T. Bhlhe40 function in activated B and TFH cells restrains the GC reaction and prevents lymphomagenesis. J Exp Med 2021; 219:212923. [PMID: 34919144 PMCID: PMC8689665 DOI: 10.1084/jem.20211406] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/01/2021] [Accepted: 11/23/2021] [Indexed: 12/22/2022] Open
Abstract
The generation of high-affinity antibodies against pathogens and vaccines requires the germinal center (GC) reaction, which relies on a complex interplay between specialized effector B and CD4 T lymphocytes, the GC B cells and T follicular helper (TFH) cells. Intriguingly, several positive key regulators of the GC reaction are common for both cell types. Here, we report that the transcription factor Bhlhe40 is a crucial cell-intrinsic negative regulator affecting both the B and T cell sides of the GC reaction. In activated CD4 T cells, Bhlhe40 was required to restrain proliferation, thus limiting the number of TFH cells. In B cells, Bhlhe40 executed its function in the first days after immunization by selectively restricting the generation of the earliest GC B cells but not of early memory B cells or plasmablasts. Bhlhe40-deficient mice with progressing age succumbed to a B cell lymphoma characterized by the accumulation of monoclonal GC B-like cells and polyclonal TFH cells in various tissues.
Collapse
Affiliation(s)
- René Rauschmeier
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Annika Reinhardt
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Gustafsson
- Center for Hematology and Regenerative Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vassilis Glaros
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Artem V. Artemov
- Department of Neuroimmunology, Medical University of Vienna, Vienna, Austria
- Endocrinology Research Centre, Moscow, Russian Federation
| | - Josefine Dunst
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Reshma Taneja
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Igor Adameyko
- Department of Neuroimmunology, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Månsson
- Center for Hematology and Regenerative Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Taras Kreslavsky
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
55
|
Zhong YB, Kang ZP, Wang MX, Long J, Wang HY, Huang JQ, Wei SY, Zhou W, Zhao HM, Liu DY. Curcumin ameliorated dextran sulfate sodium-induced colitis via regulating the homeostasis of DCs and Treg and improving the composition of the gut microbiota. J Funct Foods 2021; 86:104716. [DOI: 10.1016/j.jff.2021.104716] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
56
|
Wani SA, Sahu AR, Khan RIN, Praharaj MR, Saxena S, Rajak KK, Muthuchelvan D, Sahoo A, Mishra B, Singh RK, Mishra BP, Gandham RK. Proteome Modulation in Peripheral Blood Mononuclear Cells of Peste des Petits Ruminants Vaccinated Goats and Sheep. Front Vet Sci 2021; 8:670968. [PMID: 34631844 PMCID: PMC8493254 DOI: 10.3389/fvets.2021.670968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
In the present study, healthy goats and sheep (n = 5) that were confirmed negative for peste des petits ruminants virus (PPRV) antibodies by monoclonal antibody-based competitive ELISA and by serum neutralization test and for PPRV antigen by s-ELISA were vaccinated with Sungri/96. A quantitative study was carried out to compare the proteome of peripheral blood mononuclear cells (PBMCs) of vaccinated goat and sheep [5 days post-vaccination (dpv) and 14 dpv] vs. unvaccinated (0 day) to divulge the alteration in protein expression following vaccination. A total of 232 and 915 proteins were differentially expressed at 5 and 14 dpv, respectively, in goats. Similarly, 167 and 207 proteins were differentially expressed at 5 and 14 dpv, respectively, in sheep. Network generated by Ingenuity Pathway Analysis was “infectious diseases, antimicrobial response, and inflammatory response,” which includes the highest number of focus molecules. The bio functions, cell-mediated immune response, and humoral immune response were highly enriched in goats at 5 dpv and at 14 dpv. At the molecular level, the immune response produced by the PPRV vaccine virus in goats is effectively coordinated and stronger than that in sheep, though the vaccine provides protection from virulent virus challenge in both. The altered expression of certain PBMC proteins especially ISG15 and IRF7 induces marked changes in cellular signaling pathways to coordinate host immune responses.
Collapse
Affiliation(s)
- Sajad Ahmad Wani
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India.,College of Pharmacy, Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, United States
| | - Amit Ranjan Sahu
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Manas Ranjan Praharaj
- Systems Biology Lab, Department of Biotechnology -National Institute of Animal Biotechnology, Hyderabad, India
| | - Shikha Saxena
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Dhanavelu Muthuchelvan
- Division of Virology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Mukteswar, India
| | - Aditya Sahoo
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Bina Mishra
- Division of Biological Products, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - R K Singh
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Bishnu Prasad Mishra
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India
| | - Ravi Kumar Gandham
- Division of Veterinary Biotechnology, Indian Council of Agricultural Research - Indian Veterinary Research Institute, Bareilly, India.,Systems Biology Lab, Department of Biotechnology -National Institute of Animal Biotechnology, Hyderabad, India
| |
Collapse
|
57
|
Wan S, Ni L, Zhao X, Liu X, Xu W, Jin W, Wang X, Dong C. Costimulation molecules differentially regulate the ERK-Zfp831 axis to shape T follicular helper cell differentiation. Immunity 2021; 54:2740-2755.e6. [PMID: 34644536 DOI: 10.1016/j.immuni.2021.09.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/22/2021] [Accepted: 09/20/2021] [Indexed: 01/21/2023]
Abstract
T follicular helper (Tfh) cells play essential roles in regulating humoral immunity, especially germinal center reactions. However, how CD4+ T cells integrate the antigenic and costimulatory signals in Tfh cell development is still poorly understood. Here, we found that phorbol 12-myristate 13-acetate (PMA) + ionomycin (P+I) stimulation, together with interleukin-6 (IL-6), potently induce Tfh cell-like transcriptomic programs in vitro. The ERK kinase pathway was attenuated under P+I stimulation; ERK2 inhibition enhanced Tfh cell development in vitro and in vivo. We observed that inducible T cell costimulator (ICOS), but not CD28, lacked the ability to activate ERK, which was important in sustaining Tfh cell development. The transcription factor Zfp831, whose expression was repressed by ERK, promoted Tfh cell differentiation by directly upregulating the expression of the transcription factors Bcl6 and Tcf7. We have hence identified an ERK-Zfp831 axis, regulated by costimulation signaling, in critical regulation of Tfh cell development.
Collapse
Affiliation(s)
- Siyuan Wan
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Lu Ni
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohong Zhao
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei Xu
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Wei Jin
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohu Wang
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing, China; Shanghai Immune Therapy Institute, Shanghai Jiaotong University School of Medicine-affiliated Renji Hospital, Shanghai, China.
| |
Collapse
|
58
|
Cui D, Tang Y, Jiang Q, Jiang D, Zhang Y, Lv Y, Xu D, Wu J, Xie J, Wen C, Lu L. Follicular Helper T Cells in the Immunopathogenesis of SARS-CoV-2 Infection. Front Immunol 2021; 12:731100. [PMID: 34603308 PMCID: PMC8481693 DOI: 10.3389/fimmu.2021.731100] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a serious infectious disease that has led to a global pandemic with high morbidity and mortality. High-affinity neutralizing antibody is important for controlling infection, which is closely regulated by follicular helper T (Tfh) cells. Tfh cells play a central role in promoting germinal center reactions and driving cognate B cell differentiation for antibody secretion. Available studies indicate a close relationship between virus-specific Tfh cell-mediated immunity and SARS-CoV-2 infection progression. Although several lines of evidence have suggested that Tfh cells contribute to the control of SARS-CoV-2 infection by eliciting neutralizing antibody productions, further studies are needed to elucidate Tfh-mediated effector mechanisms in anti-SARS-CoV-2 immunity. Here, we summarize the functional features and roles of virus-specific Tfh cells in the immunopathogenesis of SARS-CoV-2 infection and in COVID-19 vaccines, and highlight the potential of targeting Tfh cells as therapeutic strategy against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Tang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Daixi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Zhang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Lv
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dandan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengping Wen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
59
|
Zhang X, Ge R, Chen H, Ahiafor M, Liu B, Chen J, Fan X. Follicular Helper CD4 + T Cells, Follicular Regulatory CD4 + T Cells, and Inducible Costimulator and Their Roles in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Mediators Inflamm 2021; 2021:2058964. [PMID: 34552387 PMCID: PMC8452443 DOI: 10.1155/2021/2058964] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Follicular helper CD4+ T (TFH) cells are a specialized subset of effector T cells that play a central role in orchestrating adaptive immunity. TFH cells mainly promote germinal center (GC) formation, provide help to B cells for immunoglobulin affinity maturation and class-switch recombination of B cells, and facilitate production of long-lived plasma cells and memory B cells. TFH cells express the nuclear transcriptional repressor B cell lymphoma 6 (Bcl-6), the chemokine (C-X-C motif) receptor 5 (CXCR5), the CD28 family members programmed cell death protein-1 (PD-1) and inducible costimulator (ICOS) and are also responsible for the secretion of interleukin-21 (IL-21) and IL-4. Follicular regulatory CD4+ T (TFR) cells, as a regulatory counterpart of TFH cells, participate in the regulation of GC reactions. TFR cells not only express markers of TFH cells but also express markers of regulatory T (Treg) cells containing FOXP3, glucocorticoid-induced tumor necrosis factor receptor (GITR), cytotoxic T lymphocyte antigen 4 (CTLA-4), and IL-10, hence owing to the dual characteristic of TFH cells and Treg cells. ICOS, expressed on activated CD4+ effector T cells, participates in T cell activation, differentiation, and effector process. The expression of ICOS is highest on TFH and TFR cells, indicating it as a key regulator of humoral immunity. Multiple sclerosis (MS) is a severe autoimmune disease that affects the central nervous system and results in disability, mediated by autoreactive T cells with evolving evidence of a remarkable contribution from humoral responses. This review summarizes recent advances regarding TFH cells, TFR cells, and ICOS, as well as their functional characteristics in relation to MS.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| | - Ruli Ge
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| | - Hongliang Chen
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| | - Maxwell Ahiafor
- School of International Studies, Binzhou Medical University, Yantai, 264003 Shandong, China
| | - Bin Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| | - Jinbo Chen
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| | - Xueli Fan
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong, China
| |
Collapse
|
60
|
Bundibugyo ebolavirus Survival Is Associated with Early Activation of Adaptive Immunity and Reduced Myeloid-Derived Suppressor Cell Signaling. mBio 2021; 12:e0151721. [PMID: 34372693 PMCID: PMC8406165 DOI: 10.1128/mbio.01517-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ebolaviruses Bundibugyo virus (BDBV) and Ebola virus (EBOV) cause fatal hemorrhagic disease in humans and nonhuman primates. While the host response to EBOV is well characterized, less is known about BDBV infection. Moreover, immune signatures that mediate natural protection against all ebolaviruses remain poorly defined. To explore these knowledge gaps, we transcriptionally profiled BDBV-infected rhesus macaques, a disease model that results in incomplete lethality. This approach enabled us to identify prognostic indicators. As expected, survival (∼60%) correlated with reduced clinical pathology and circulating infectious virus, although peak viral RNA loads were not significantly different between surviving and nonsurviving macaques. Survivors had higher anti-BDBV antibody titers and transcriptionally derived cytotoxic T cell-, memory B cell-, and plasma cell-type quantities, demonstrating activation of adaptive immunity. Conversely, a poor prognosis was associated with lack of an appropriate adaptive response, sustained innate immune signaling, and higher expression of myeloid-derived suppressor cell (MDSC)-related transcripts (S100A8, S100A9, CEBPB, PTGS2, CXCR1, and LILRA3). MDSCs are potent immunosuppressors of cellular and humoral immunity, and therefore, they represent a potential therapeutic target. Circulating plasminogen activator inhibitor 1 (PAI-1) and tissue plasminogen activator (tPA) levels were also elevated in nonsurvivors and in survivors exhibiting severe illness, emphasizing the importance of maintaining coagulation homeostasis to control disease progression.
Collapse
|
61
|
Jin J, Kim C, Xia Q, Gould TM, Cao W, Zhang H, Li X, Weiskopf D, Grifoni A, Sette A, Weyand CM, Goronzy JJ. Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Sci Immunol 2021; 6:eabg0791. [PMID: 34145066 PMCID: PMC8422387 DOI: 10.1126/sciimmunol.abg0791] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.
Collapse
Affiliation(s)
- Jun Jin
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Qiong Xia
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Timothy M Gould
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Huimin Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Xuanying Li
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Jorg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| |
Collapse
|
62
|
Spinelli L, Marchingo JM, Nomura A, Damasio MP, Cantrell DA. Phosphoinositide 3-Kinase p110 Delta Differentially Restrains and Directs Naïve Versus Effector CD8 + T Cell Transcriptional Programs. Front Immunol 2021; 12:691997. [PMID: 34220851 PMCID: PMC8250422 DOI: 10.3389/fimmu.2021.691997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Phosphoinositide 3-kinase p110 delta (PI3K p110δ) is pivotal for CD8+ T cell immune responses. The current study explores PI3K p110δ induction and repression of antigen receptor and cytokine regulated programs to inform how PI3K p110δ directs CD8+ T cell fate. The studies force a revision of the concept that PI3K p110δ controls metabolic pathways in T cells and reveal major differences in PI3K p110δ regulated transcriptional programs between naïve and effector cytotoxic T cells (CTL). These differences include differential control of the expression of cytolytic effector molecules and costimulatory receptors. Key insights from the work include that PI3K p110δ signalling pathways repress expression of the critical inhibitory receptors CTLA4 and SLAMF6 in CTL. Moreover, in both naïve and effector T cells the dominant role for PI3K p110δ is to restrain the production of the chemokines that orchestrate communication between adaptive and innate immune cells. The study provides a comprehensive resource for understanding how PI3K p110δ uses multiple processes mediated by Protein Kinase B/AKT, FOXO1 dependent and independent mechanisms and mitogen-activated protein kinases (MAPK) to direct CD8+ T cell fate.
Collapse
Affiliation(s)
| | | | | | | | - Doreen A. Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
63
|
Olatunde AC, Hale JS, Lamb TJ. Cytokine-skewed Tfh cells: functional consequences for B cell help. Trends Immunol 2021; 42:536-550. [PMID: 33972167 PMCID: PMC9107098 DOI: 10.1016/j.it.2021.04.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022]
Abstract
CD4+ follicular helper T (Tfh) cells play a vital role in providing help for B cells undergoing selection and differentiation into activated antibody-secreting cells in mammalian germinal centers (GCs). Increasing evidence suggests that Tfh cells are a heterogeneous population that generates cytokine-skewed immune responses - a reflection of the microenvironment during differentiation. This has important ramifications for Tfh-mediated B cell help. Because Tfh subsets can have opposing effects on GC B cell responses, we discuss current findings regarding the differentiation and functions of cytokine-skewed Tfh cells in modulating GC B cell differentiation. Antibodies are important weapons against infectious diseases but can also be pathogenic mediators in some autoimmune conditions. Since cytokine-skewed Tfh cells can influence the magnitude and quality of the humoral response, we address the roles of cytokine-skewed Tfh cells in disease.
Collapse
Affiliation(s)
- Adesola C Olatunde
- Department of Pathology, University of Utah, 15 North Medical Drive, Salt Lake City, UT 84112, USA
| | - J Scott Hale
- Department of Pathology, University of Utah, 15 North Medical Drive, Salt Lake City, UT 84112, USA
| | - Tracey J Lamb
- Department of Pathology, University of Utah, 15 North Medical Drive, Salt Lake City, UT 84112, USA.
| |
Collapse
|
64
|
Hao H, Nakayamada S, Tanaka Y. Differentiation, functions, and roles of T follicular regulatory cells in autoimmune diseases. Inflamm Regen 2021; 41:14. [PMID: 33934711 PMCID: PMC8088831 DOI: 10.1186/s41232-021-00164-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
T follicular helper cells participate in stimulating germinal center (GC) formation and supporting B cell differentiation and autoantibody production. However, T follicular regulatory (Tfr) cells suppress B cell activation. Since changes in the number and functions of Tfr cells lead to dysregulated GC reaction and autoantibody response, targeting Tfr cells may benefit the treatment of autoimmune diseases. Differentiation of Tfr cells is a multistage and multifactorial process with various positive and negative regulators. Therefore, understanding the signals regulating Tfr cell generation is crucial for the development of targeted therapies. In this review, we discuss recent studies that have elucidated the roles of Tfr cells in autoimmune diseases and investigated the modulators of Tfr cell differentiation. Additionally, potential immunotherapies targeting Tfr cells are highlighted.
Collapse
Affiliation(s)
- He Hao
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, Kitakyushu, 807-8555, Japan.,Department of Immuno-oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, Kitakyushu, 807-8555, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
65
|
Papillion A, Ballesteros-Tato A. The Potential of Harnessing IL-2-Mediated Immunosuppression to Prevent Pathogenic B Cell Responses. Front Immunol 2021; 12:667342. [PMID: 33986755 PMCID: PMC8112607 DOI: 10.3389/fimmu.2021.667342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 11/18/2022] Open
Abstract
Immunosuppressive drugs can partially control Antibody (Ab)-dependent pathology. However, these therapeutic regimens must be maintained for the patient's lifetime, which is often associated with severe side effects. As research advances, our understanding of the cellular and molecular mechanisms underlying the development and maintenance of auto-reactive B cell responses has significantly advanced. As a result, novel immunotherapies aimed to restore immune tolerance and prevent disease progression in autoimmune patients are underway. In this regard, encouraging results from clinical and preclinical studies demonstrate that subcutaneous administration of low-doses of recombinant Interleukin-2 (r-IL2) has potent immunosuppressive effects in patients with autoimmune pathologies. Although the exact mechanism by which IL-2 induces immunosuppression remains unclear, the clinical benefits of the current IL-2-based immunotherapies are attributed to its effect on bolstering T regulatory (Treg) cells, which are known to suppress overactive immune responses. In addition to Tregs, however, rIL-2 also directly prevent the T follicular helper cells (Tfh), T helper 17 cells (Th17), and Double Negative (DN) T cell responses, which play critical roles in the development of autoimmune disorders and have the ability to help pathogenic B cells. Here we discuss the broader effects of rIL-2 immunotherapy and the potential of combining rIL-2 with other cytokine-based therapies to more efficiently target Tfh cells, Th17, and DN T cells and subsequently inhibit auto-antibody (ab) production in autoimmune patients.
Collapse
Affiliation(s)
| | - André Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
66
|
Long Y, Feng J, Ma Y, Sun Y, Xu L, Song Y, Liu C. Altered follicular regulatory T (Tfr)- and helper T (Tfh)-cell subsets are associated with autoantibody levels in microscopic polyangiitis patients. Eur J Immunol 2021; 51:1809-1823. [PMID: 33764509 DOI: 10.1002/eji.202049093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/10/2021] [Accepted: 03/22/2021] [Indexed: 12/21/2022]
Abstract
Antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) is an autoimmune disease characterized by B cells-derived ANCAs, and ANCA was proved to be a key factor in its pathogenesis. Follicular regulatory T (Tfr) and follicular helper T (Tfh) cells were T-cell subsets that play important roles in B-cell maturation and antibody production. However, their significances in microscopic polyangiitis (MPA) patients, one type of AAV, has not been thoroughly studied. In this study, comprehensive pattern analyses of circulating Tfr and Tfh were performed in MPA patients and healthy controls (HCs), and we found Tfr levels and Tfr/Tfh ratios were significantly decreased in MPA patients. Compared with HCs, Helios+, CD45RA-FoxP3hi, and Ki-67+ Tfr were lower in MPA patients, while CD226+ Tfr cells were higher. These phenotypes suggest that function and proliferation ability of Tfr cells were relatively impaired. Tfh subsets, including ICOS+PD-1+ and Ki-67+ Tfh, were significantly increased, suggesting that the function of Tfh was enhanced in MPA although the total Tfh levels did not change significantly. Circulating memory B cells and plasmablasts were significantly elevated and negatively correlated with Tfr levels and Tfr/Tfh ratios in MPA patients. In addition, Tfr levels and Tfr/Tfh ratios were negatively while Tfh was positively correlated with serum myeloperoxidase (MPO)-ANCA levels. Furthermore, Tfr and Tfr/Tfh ratio were also reversely associated with SCr, BUN, IL-4, and IL-21 levels. Our results suggest that the imbalance of Tfr and Tfh functional subsets is related to increased level of autoantibodies in MPA patients, and we propose a new mechanism for the pathogenesis of MPA.
Collapse
Affiliation(s)
- Yan Long
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Jinghong Feng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yinting Ma
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuanyuan Sun
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Lijuan Xu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Ying Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
67
|
Zhang S, Li L, Xie D, Reddy S, Sleasman JW, Ma L, Zhong XP. Regulation of Intrinsic and Bystander T Follicular Helper Cell Differentiation and Autoimmunity by Tsc1. Front Immunol 2021; 12:620437. [PMID: 33936036 PMCID: PMC8079652 DOI: 10.3389/fimmu.2021.620437] [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: 10/22/2020] [Accepted: 03/24/2021] [Indexed: 11/13/2022] Open
Abstract
T Follicular helper (Tfh) cells promote germinal center (GC) B cell responses to develop effective humoral immunity against pathogens. However, dysregulated Tfh cells can also trigger autoantibody production and the development of autoimmune diseases. We report here that Tsc1, a regulator for mTOR signaling, plays differential roles in Tfh cell/GC B cell responses in the steady state and in immune responses to antigen immunization. In the steady state, Tsc1 in T cells intrinsically suppresses spontaneous GC-Tfh cell differentiation and subsequent GC-B cell formation and autoantibody production. In immune responses to antigen immunization, Tsc1 in T cells is required for efficient GC-Tfh cell expansion, GC-B cell induction, and antigen-specific antibody responses, at least in part via promoting GC-Tfh cell mitochondrial integrity and survival. Interestingly, in mixed bone marrow chimeric mice reconstituted with both wild-type and T cell-specific Tsc1-deficient bone marrow cells, Tsc1 deficiency leads to enhanced GC-Tfh cell differentiation of wild-type CD4 T cells and increased accumulation of wild-type T regulatory cells and T follicular regulatory cells. Such bystander GC-Tfh cell differentiation suggests a potential mechanism that could trigger self-reactive GC-Tfh cell/GC responses and autoimmunity via neighboring GC-Tfh cells.
Collapse
Affiliation(s)
- Shimeng Zhang
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lei Li
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danli Xie
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Srija Reddy
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States
| | - John W Sleasman
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiao-Ping Zhong
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Hematologic Malignancies and Cellular Therapies Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
68
|
Long Y, Xia C, Sun Y, Ma Y, Xu L, Song Y, Liu C. Increased circulating PD-1 hiCXCR5- peripheral helper T cells are associated with disease severity of active ulcerative colitis patients. Immunol Lett 2021; 233:2-10. [PMID: 33675861 DOI: 10.1016/j.imlet.2021.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/27/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE A recently identified population of T cells, phenotypically CD4+PD-1hiCXCR5-, has been firstly termed as peripheral helper T cells (Tph) and found to be pathogenic in autoimmune diseases. However, the potential role of Tph in ulcerative colitis (UC) remains unclear. We aim to investigate the changes of circulating Tph in UC patients and their potential significance in the pathogenesis of UC. METHODS Totally 68 UC patients and 34 age- and sex-matched healthy controls were enrolled. Circulating Tph and B cell subsets were analyzed by flow cytometry. Expressions of inducible T-cell co-stimulator (ICOS) on Tph cells were analyzed. Serum IL-4, IL-10, IL-12 and IL-21 were detected using ELISA. Correlation analyses were conducted between Tph cells and disease severity, functional B cell subsets and serum cytokines. RESULTS Both the frequency and absolute number of Tph were significantly increased in active UC patients and ICOS levels in Tph cells were also elevated, compared with remission UC patients and healthy controls. Tph and ICOS expression were significantly positively correlated with Mayo score and serum CRP in active UC patients, and were significantly decreased when achieving remission after treatment. Tph levels were correlated with new memory B cells, plasmablasts, serum IL-4 and IL-21. Meanwhile, serum IL-10 showed negative correlation while IL-12 exhibited positive correlation with circulating Tph cells in UC patients. CONCLUSIONS Circulating Tph cells are elevated in active UC patients and are associated with the disease activity, which may contribute to the pathogenesis of UC.
Collapse
Affiliation(s)
- Yan Long
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Changsheng Xia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuanyuan Sun
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yinting Ma
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Lijuan Xu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Ying Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
69
|
Schroeder AR, Zhu F, Hu H. Stepwise Tfh cell differentiation revisited: new advances and long-standing questions. Fac Rev 2021; 10. [PMID: 33644779 PMCID: PMC7894273 DOI: 10.12703/r/10-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
T follicular helper (Tfh) cells play an essential role in germinal center formation and the generation of high-affinity antibodies. Studies have proposed that Tfh cell differentiation is a multi-step process. However, it is still not fully understood how a subset of activated CD4+ T cells begin to express CXCR5 during the early stage of the response and, shortly after, how some CXCR5+ precursor Tfh (pre-Tfh) cells enter B cell follicles and differentiate further into germinal center Tfh (GC-Tfh) cells while others have a different fate. In this mini-review, we summarize the recent advances surrounding these two aspects of Tfh cell differentiation and discuss related long-standing questions, including Tfh memory.
Collapse
Affiliation(s)
- Andrew R Schroeder
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fangming Zhu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hui Hu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
70
|
Choi J, Crotty S. Bcl6-Mediated Transcriptional Regulation of Follicular Helper T cells (T FH). Trends Immunol 2021; 42:336-349. [PMID: 33663954 DOI: 10.1016/j.it.2021.02.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 02/01/2023]
Abstract
Follicular helper T cells (TFH) are essential B cell-help providers in the formation of germinal centers (GCs), affinity maturation of GC B cells, differentiation of high-affinity antibody-producing plasma cells, and production of memory B cells. The transcription factor (TF) B cell lymphoma 6 (Bcl6) is at the center of gene regulation in TFH biology, including differentiation and function, but how Bcl6 does this, and what additional TFs contribute, remain complex questions. This review focuses on advances in our understanding of Bcl6-mediated gene regulation of TFH functions, and the modulation of TFH by other TFs. These advances may have important implications in deciphering how repressor TFs can regulate many immunological cell types. An improved understanding of TFH biology will likely provide insights into biomedically relevant diseases.
Collapse
Affiliation(s)
- Jinyong Choi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA.
| |
Collapse
|
71
|
Sun Z, Yao Y, You M, Liu J, Guo W, Qi Z, Wang Z, Wang F, Yuan W, Yu S. The kinase PDK1 is critical for promoting T follicular helper cell differentiation. eLife 2021; 10:61406. [PMID: 33595435 PMCID: PMC7889074 DOI: 10.7554/elife.61406] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/08/2021] [Indexed: 01/03/2023] Open
Abstract
The kinase PDK1 is a crucial regulator for immune cell development by connecting PI3K to downstream AKT signaling. However, the roles of PDK1 in CD4+ T cell differentiation, especially in T follicular helper (Tfh) cell, remain obscure. Here we reported PDK1 intrinsically promotes the Tfh cell differentiation and germinal center responses upon acute infection by using conditional knockout mice. PDK1 deficiency in T cells caused severe defects in both early differentiation and late maintenance of Tfh cells. The expression of key Tfh regulators was remarkably downregulated in PDK1-deficient Tfh cells, including Tcf7, Bcl6, Icos, and Cxcr5. Mechanistically, ablation of PDK1 led to impaired phosphorylation of AKT and defective activation of mTORC1, resulting in substantially reduced expression of Hif1α and p-STAT3. Meanwhile, decreased p-AKT also suppresses mTORC2-associated GSK3β activity in PDK1-deficient Tfh cells. These integrated effects contributed to the dramatical reduced expression of TCF1 and ultimately impaired the Tfh cell differentiation.
Collapse
Affiliation(s)
- Zhen Sun
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingpeng Yao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Menghao You
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jingjing Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wenhui Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhihong Qi
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Fang Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, and Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shuyang Yu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
72
|
Abstract
Posttranscriptional control of mRNA regulates various biological processes, including inflammatory and immune responses. RNA-binding proteins (RBPs) bind cis-regulatory elements in the 3' untranslated regions (UTRs) of mRNA and regulate mRNA turnover and translation. In particular, eight RBPs (TTP, AUF1, KSRP, TIA-1/TIAR, Roquin, Regnase, HuR, and Arid5a) have been extensively studied and are key posttranscriptional regulators of inflammation and immune responses. These RBPs sometimes collaboratively or competitively bind the same target mRNA to enhance or dampen regulatory activities. These RBPs can also bind their own 3' UTRs to negatively or positively regulate their expression. Both upstream signaling pathways and microRNA regulation shape the interactions between RBPs and target RNA. Dysregulation of RBPs results in chronic inflammation and autoimmunity. Here, we summarize the functional roles of these eight RBPs in immunity and their associated diseases.
Collapse
Affiliation(s)
- Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0874, Japan.,Department of Host Defense, Division of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0874, Japan;
| | - Kazuhiko Maeda
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0874, Japan.,Department of Host Defense, Division of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0874, Japan;
| |
Collapse
|
73
|
Abstract
Akt kinases translate various external cues into intracellular signals that control cell survival, proliferation, metabolism and differentiation. This review discusses the requirement for Akt and its targets in determining the fate and function of T cells. We discuss the importance of Akt at various stages of T cell development including β-selection during which Akt fulfills the energy requirements of highly proliferative DN3 cells. Akt also plays an integral role in CD8 T cell biology where its regulation of Foxo transcription factors and mTORC1 metabolic activity controls effector versus memory CD8 T cell differentiation. Finally, Akt promotes the differentiation of naïve CD4 T cells into Th1, Th17 and Tfh cells but inhibits the development of Treg cells. We also highlight how modulating Akt in T cells is a promising avenue for enhancing cell-based cancer immunotherapy.
Collapse
|
74
|
Cao Y, Dong L, He Y, Hu X, Hou Y, Dong Y, Yang Q, Bi Y, Liu G. The direct and indirect regulation of follicular T helper cell differentiation in inflammation and cancer. J Cell Physiol 2021; 236:5466-5480. [PMID: 33421124 DOI: 10.1002/jcp.30263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/03/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Follicular T helper (Tfh) cells play important roles in facilitating B-cell differentiation and inducing the antibody response in humoral immunity and immune-associated inflammatory diseases, including infections, autoimmune diseases, and cancers. However, Tfh cell differentiation is mainly achieved through self-directed differentiation regulation and the indirect regulation mechanism of antigen-presenting cells (APCs). During the direct intrinsic differentiation of naïve CD4+ T cells into Tfh cells, Bcl-6, as the characteristic transcription factor, plays the core role of transcriptional regulation. APCs indirectly drive Tfh cell differentiation mainly by changing cytokine secretion mechanisms. Altered metabolic signaling is also critically involved in Tfh cell differentiation. This review summarizes the recent progress in understanding the direct and indirect regulatory signals and metabolic mechanisms of Tfh cell differentiation and function in immune-associated diseases.
Collapse
Affiliation(s)
- Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Ying He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Xuelian Hu
- Immunochina Pharmaceuticals Co., Ltd., No. 80, Xingshikou Road, Haidian District, Beijing, China
| | - Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, No. 20, East Street, Fengtai District, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing, China
| |
Collapse
|
75
|
Jin J, Zhang H, Weyand CM, Goronzy JJ. Lysosomes in T Cell Immunity and Aging. FRONTIERS IN AGING 2021; 2:809539. [PMID: 35822050 PMCID: PMC9261317 DOI: 10.3389/fragi.2021.809539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 05/01/2023]
Abstract
Lysosomes were initially recognized as degradation centers that regulate digestion and recycling of cellular waste. More recent studies document that the lysosome is an important signaling hub that regulates cell metabolism. Our knowledge of the role of lysosomes in immunity is mostly derived from innate immune cells, especially lysosomal degradation-specialized cells such as macrophages and dendritic cells. Their function in adaptive immunity is less understood. However, with the recent emphasis on metabolic regulation of T cell differentiation, lysosomes are entering center stage in T cell immunology. In this review, we will focus on the role of lysosomes in adaptive immunity and discuss recent findings on lysosomal regulation of T cell immune responses and lysosomal dysfunction in T cell aging.
Collapse
Affiliation(s)
- Jun Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Huimin Zhang
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Cornelia M. Weyand
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Jorg J. Goronzy
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Jorg J. Goronzy,
| |
Collapse
|
76
|
Delpoux A, Marcel N, Hess Michelini R, Katayama CD, Allison KA, Glass CK, Quiñones-Parra SM, Murre C, Loh L, Kedzierska K, Lappas M, Hedrick SM, Doedens AL. FOXO1 constrains activation and regulates senescence in CD8 T cells. Cell Rep 2021; 34:108674. [PMID: 33503413 DOI: 10.1016/j.celrep.2020.108674] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/25/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Naive and memory T cells are maintained in a quiescent state, yet capable of rapid response and differentiation to antigen challenge via molecular mechanisms that are not fully understood. In naive cells, the deletion of Foxo1 following thymic development results in the increased expression of multiple AP-1 family members, rendering T cells less able to respond to antigenic challenge. Similarly, in the absence of FOXO1, post-infection memory T cells exhibit the characteristics of extended activation and senescence. Age-based analysis of human peripheral T cells reveals that levels of FOXO1 and its downstream target, TCF7, are inversely related to host age, whereas the opposite is found for AP-1 factors. These characteristics of aging also correlate with the formation of T cells manifesting features of cellular senescence. Our work illustrates a role for FOXO1 in the active maintenance of stem-like properties in T cells at the timescales of acute infection and organismal life span.
Collapse
Affiliation(s)
- Arnaud Delpoux
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Nimi Marcel
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Rodrigo Hess Michelini
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Carol D Katayama
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Karmel A Allison
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Sergio M Quiñones-Parra
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Cornelis Murre
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Martha Lappas
- Obstetrics, Nutrition, and Endocrinology Group, Department of Obstetrics & Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Stephen M Hedrick
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA.
| | - Andrew L Doedens
- Division of Biological Sciences, Molecular Biology Section, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0377, USA.
| |
Collapse
|
77
|
Dong L, He Y, Cao Y, Wang Y, Jia A, Wang Y, Yang Q, Li W, Bi Y, Liu G. Functional differentiation and regulation of follicular T helper cells in inflammation and autoimmunity. Immunology 2020; 163:19-32. [PMID: 33128768 DOI: 10.1111/imm.13282] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Follicular T helper (TFH ) cells are specialized T cells that support B cells, which are essential for humoral immunity. TFH cells express the transcription factor B-cell lymphoma 6 (Bcl-6), chemokine (C-X-C motif) receptor (CXCR) 5, the surface receptors programmed cell death protein 1 (PD-1) and inducible T-cell costimulator (ICOS), the cytokine IL-21 and other molecules. The activation, proliferation and differentiation of TFH cells are closely related to dynamic changes in cellular metabolism. In this review, we summarize the progress made in understanding the development and functional differentiation of TFH cells. Specifically, we focus on the regulatory mechanisms of TFH cell functional differentiation, including regulatory signalling pathways and the metabolic regulatory mechanisms of TFH cells. In addition, TFH cells are closely related to immune-associated diseases, including infections, autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ying He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yuexin Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Wanjie Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
78
|
Xiao ZX, Hu X, Jarjour W, Zheng SG. The role of B7 family members in the generation of Immunoglobulin. J Leukoc Biol 2020; 109:377-382. [PMID: 33118237 DOI: 10.1002/jlb.1mr0420-003rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 10/23/2022] Open
Abstract
Ig is a Y-shaped protein produced by plasma cells and exerts multiple functions in humoral immunity. There are five groups of Igs including IgA, IgD, IgE, IgG, and IgM, which differ in their heavy chain class. The primary function of Igs includes the neutralization of extrinsic pathogens, agglutination of foreign cells for phagocytosis, precipitation of soluble antigens in serum, and complement fixation. The B cells activated by antigen(s) can differentiate into antibody-producing cells that are called plasma cells and usually matured in the germinal center (GC). Follicular T helper (Tfh) cells crosstalk with antigen-presenting cells and play a crucial role in the development of the GC. Moreover, Tfh cells regulate trafficking through the GC to allow formative interaction with GC B cells that ultimately results in affinity maturation, B-cell memory, and Ig class switching. The B7 family is a series of number of structurally related membrane proteins that bind with a specific receptor to deliver costimulatory or co-inhibitory signals that regulate the activation of T cells in GC. Here, we review and summarize the recent advance of the effects of B7 family members on Ig production and relative diseases.
Collapse
Affiliation(s)
- Ze Xiu Xiao
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaojiang Hu
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wael Jarjour
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
79
|
Amatore F, Ortonne N, Lopez M, Orlanducci F, Castellano R, Ingen-Housz-Oro S, De Croos A, Salvado C, Gorvel L, Goubard A, Collette Y, Bouabdallah R, Schiano JM, Bonnet N, Grob JJ, Gaulard P, Bagot M, Bensussan A, Berbis P, Olive D. ICOS is widely expressed in cutaneous T-cell lymphoma, and its targeting promotes potent killing of malignant cells. Blood Adv 2020; 4:5203-5214. [PMID: 33095875 PMCID: PMC7594390 DOI: 10.1182/bloodadvances.2020002395] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
The treatment of advanced-stage cutaneous T-cell lymphoma (CTCL) remains an unmet medical need. Mogamulizumab, anti-KIR3DL2, and brentuximab vedotin (BV), an anti-CD30 antibody-drug conjugate (ADC) coupled with monomethyl-auristatin-E (MMAE), provided encouraging results, but new targeted therapies are needed. Inducible T-cell costimulator (ICOS), a T-cell costimulatory receptor, is a promising therapeutic target, not only because it is expressed by malignant T cells in CTCL but also because of its connection with the suppressive activity of regulatory T (Treg) cells. Immunohistochemical analysis revealed that ICOS was widely expressed by malignant cells in skin biopsy specimens from 52 patients with mycosis fungoides and Sézary syndrome (SS), as well as in involved node biopsy specimens from patients with SS. Furthermore, flow cytometry demonstrated its strong expression by circulating tumor cells in all our patients with SS. Percentages of ICOS+ Treg cells were significantly higher in patients with SS than in healthy donors. We then investigated the preclinical efficacy of anti-ICOS ADCs generated by coupling murine anti-ICOS monoclonal antibodies with MMAE and pyrrolobenzodiazepine. In 3 CTCL cell lines (Myla, MJ, and HUT78), we observed a significant dose-dependent decrease in cell viability in the presence of anti-ICOS ADCs. In addition, anti-ICOS-MMAE ADCs had an in vitro and in vivo efficacy superior to BV in a mouse xenograft model (MyLa). Finally, we assessed the efficacy of anti-ICOS ADCs in ICOS+ patient-derived xenografts from patients with SS and angioimmunoblastic T-cell lymphoma. Collectively, our findings provide the preliminary basis for a therapeutic trial.
Collapse
Affiliation(s)
- Florent Amatore
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
- Department of Dermatology and Skin Cancers, Hôpital de la Timone, Aix Marseille Université, Marseille, France
- Department of Dermatology, Aix Marseille University, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | | | - Marc Lopez
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Florence Orlanducci
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Rémy Castellano
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Saskia Ingen-Housz-Oro
- Department of Dermatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Henri-Mondor, Créteil, France
| | | | | | - Laurent Gorvel
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Armelle Goubard
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Yves Collette
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Réda Bouabdallah
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Jean-Marc Schiano
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Nathalie Bonnet
- Department of Dermatology, Aix Marseille University, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | - Jean-Jacques Grob
- Department of Dermatology and Skin Cancers, Hôpital de la Timone, Aix Marseille Université, Marseille, France
| | | | - Martine Bagot
- Department of Dermatology, Saint-Louis Hospital, AP-HP, Paris University, INSERM U976, Paris, France; and
| | - Armand Bensussan
- Paris University, INSERM, UMR-976, Institut de Recherche Saint-Louis, Paris, France
| | - Philippe Berbis
- Department of Dermatology, Aix Marseille University, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | - Daniel Olive
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Centre National de la Recherche Scientifique U7258, Aix Marseille Université, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
80
|
Bouchard A, Witalis M, Chang J, Panneton V, Li J, Bouklouch Y, Suh WK. Hippo Signal Transduction Mechanisms in T Cell Immunity. Immune Netw 2020; 20:e36. [PMID: 33163244 PMCID: PMC7609160 DOI: 10.4110/in.2020.20.e36] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022] Open
Abstract
Hippo signaling pathways are evolutionarily conserved signal transduction mechanisms mainly involved in organ size control, tissue regeneration, and tumor suppression. However, in mammals, the primary role of Hippo signaling seems to be regulation of immunity. As such, humans with null mutations in STK4 (mammalian homologue of Drosophila Hippo; also known as MST1) suffer from recurrent infections and autoimmune symptoms. Although dysregulated T cell homeostasis and functions have been identified in MST1-deficient human patients and mouse models, detailed cellular and molecular bases of the immune dysfunction remain to be elucidated. Although the canonical Hippo signaling pathway involves transcriptional co-activator Yes-associated protein (YAP) or transcriptional coactivator with PDZ motif (TAZ), the major Hippo downstream signaling pathways in T cells are YAP/TAZ-independent and they widely differ between T cell subsets. Here we will review Hippo signaling mechanisms in T cell immunity and describe their implications for immune defects found in MST1-deficient patients and animals. Further, we propose that mutual inhibition of Mst and Akt kinases and their opposing roles on the stability and function of forkhead box O and β-catenin may explain various immune defects discovered in mutant mice lacking Hippo signaling components. Understanding these diverse Hippo signaling pathways and their interplay with other evolutionarily-conserved signaling components in T cells may uncover molecular targets relevant to vaccination, autoimmune diseases, and cancer immunotherapies.
Collapse
Affiliation(s)
- Antoine Bouchard
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Molecular Biology Program, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Mariko Witalis
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Molecular Biology Program, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Jinsam Chang
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Molecular Biology Program, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Vincent Panneton
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Department of Microbiology, Infectiology, and Immunology, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Joanna Li
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Yasser Bouklouch
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada.,Molecular Biology Program, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada.,Department of Microbiology, Infectiology, and Immunology, University of Montreal, Montreal, QC H3T 1J4, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
81
|
Abstract
Virtually all aspects of T and B lymphocyte development, homeostasis, activation, and effector function are impacted by the interaction of their clonally distributed antigen receptors with antigens encountered in their respective environments. Antigen receptors mediate their effects by modulating intracellular signaling pathways that ultimately impinge on the cytoskeleton, bioenergetic pathways, transcription, and translation. Although these signaling pathways are rather well described at this point, especially those steps that are most receptor-proximal, how such pathways contribute to more quantitative aspects of lymphocyte function is still being elucidated. One of the signaling pathways that appears to be involved in this “tuning” process is controlled by the lipid kinase PI3K. Here we review recent key findings regarding both the triggering/enhancement of PI3K signals (via BCAP and ICOS) as well as their regulation (via PIK3IP1 and PHLPP) and how these signals integrate and determine cellular processes. Lymphocytes display tremendous functional plasticity, adjusting their metabolism and gene expression programs to specific conditions depending on their tissue of residence and the nature of the infectious threat to which they are responding. We give an overview of recent findings that have contributed to this model, with a focus on T cells, including what has been learned from patients with gain-of-function mutations in PI3K as well as lessons from cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Benjamin Murter
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|
82
|
Abstract
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Collapse
|
83
|
Peng C, Jameson SC. The relationship between CD4+ follicular helper T cells and CD8+ resident memory T cells: sisters or distant cousins? Int Immunol 2020; 32:583-587. [PMID: 32620009 PMCID: PMC7478156 DOI: 10.1093/intimm/dxaa045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Independent studies over the last decade have characterized the properties of non-circulating CD8+ 'resident' memory T cells (TRM), which offer barrier protective immunity in non-lymphoid tissues and CD4+ follicular helper T cells (TFH), which mediate B-cell help in lymphoid sites. Despite their very different biological roles in the immune system, intriguing parallels have been noted between the trafficking properties and differentiation cues of these populations, parallels which have only sharpened with recent findings. In this review, we explore the features that underlie these similarities and discuss whether these indicate meaningful homologies in the development of CD8+ TRM and CD4+ TFH or reflect resemblances which are only 'skin-deep'.
Collapse
Affiliation(s)
- Changwei Peng
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Stephen C Jameson
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
84
|
Mhaidly R, Krug A, Gaulard P, Lemonnier F, Ricci JE, Verhoeyen E. New preclinical models for angioimmunoblastic T-cell lymphoma: filling the GAP. Oncogenesis 2020; 9:73. [PMID: 32796826 PMCID: PMC7427806 DOI: 10.1038/s41389-020-00259-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mouse models are essential to study and comprehend normal and malignant hematopoiesis. The ideal preclinical model should mimic closely the human malignancy. This means that these mice should recapitulate the clinical behavior of the human diseases such as cancer and therapeutic responses with high reproducibility. In addition, the genetic mutational status, the cell phenotype, the microenvironment of the tumor and the time until tumor development occurs, should be mimicked in a preclinical model. This has been particularly challenging for human angioimmunoblastic lymphoma (AITL), one of the most prominent forms of peripheral T-cell lymphomas. A complex network of interactions between AITL tumor cells and the various cells of the tumor microenvironment has impeded the study of AITL pathogenesis in vitro. Very recently, new mouse models that recapitulate faithfully the major features of human AITL disease have been developed. Here, we provide a summary of the pathology, the transcriptional profile and genetic and immune-phenotypic features of human AITL. In addition, we give an overview of preclinical models that recapitulate more or less faithfully human AITL characteristics and pathology. These recently engineered mouse models were essential in the evaluation of novel therapeutic agents for possible treatment of AITL, a malignancy in urgent need of new treatment options.
Collapse
Affiliation(s)
- Rana Mhaidly
- Université Côte d'Azur, INSERM, C3M, 06204, Nice, France
- Institut Curie, Stress and Cancer Laboratory, Equipe Labellisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'ULM, F-75248, Paris, France
- Inserm, U830, 26, rue d'ULM, Paris, F-75005, France
| | - Adrien Krug
- Université Côte d'Azur, INSERM, C3M, 06204, Nice, France
| | - Philippe Gaulard
- Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, INSERMU955; Institut Mondor de Recherche Biomédicale, INSERMU955, Université Paris Est Créteil, Créteil, France
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Créteil, France
| | - François Lemonnier
- Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, INSERMU955; Institut Mondor de Recherche Biomédicale, INSERMU955, Université Paris Est Créteil, Créteil, France
- Unité Hémopathies Lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance Publique des Hôpitaux de Paris, Créteil, France
| | | | - Els Verhoeyen
- Université Côte d'Azur, INSERM, C3M, 06204, Nice, France.
- CIRI, Université de Lyon, INSERM U1111, ENS de Lyon, Université Lyon1, CNRS, UMR 5308, 69007, Lyon, France.
| |
Collapse
|
85
|
Chiba S, Sakata-Yanagimoto M. Advances in understanding of angioimmunoblastic T-cell lymphoma. Leukemia 2020; 34:2592-2606. [PMID: 32704161 PMCID: PMC7376827 DOI: 10.1038/s41375-020-0990-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/27/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
It has been nearly half a century since angioimmunoblastic T-cell lymphoma (AITL) was characterized in the early 1970’s. Our understanding of the disease has dramatically changed due to multiple discoveries and insights. One of the key features of AITL is aberrant immune activity. Although AITL is now understood to be a neoplastic disease, pathologists appreciated that it was an inflammatory condition. The more we understand AITL at cellular and genetic levels, the more we view it as both a neoplastic and an inflammatory disease. Here, we review recent progress in our understanding of AITL, focusing on as yet unsolved questions.
Collapse
Affiliation(s)
- Shigeru Chiba
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Mamiko Sakata-Yanagimoto
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
86
|
Du T, Yang CL, Ge MR, Liu Y, Zhang P, Li H, Li XL, Li T, Liu YD, Dou YC, Yang B, Duan RS. M1 Macrophage Derived Exosomes Aggravate Experimental Autoimmune Neuritis via Modulating Th1 Response. Front Immunol 2020; 11:1603. [PMID: 32793234 PMCID: PMC7390899 DOI: 10.3389/fimmu.2020.01603] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 06/16/2020] [Indexed: 12/29/2022] Open
Abstract
Guillain–Barré syndrome (GBS), an immune-mediated disorder affecting the peripheral nervous system, is the most common and severe acute paralytic neuropathy. GBS remains to be potentially life-threatening and disabling despite the increasing availability of current standard therapeutic regimens. Therefore, more targeted therapeutics are in urgent need. Macrophages have been implicated in both initiation and resolution of experimental autoimmune neuritis (EAN), the animal model of GBS, but the exact mechanisms remain to be elucidated. It has been increasingly appreciated that exosomes, a type of extracellular vesicles (EVs), are of importance for functions of macrophages. Nevertheless, the roles of macrophage derived exosomes in EAN/GBS remain unclear. Here we determined the effects of macrophage derived exosomes on the development of EAN in Lewis rats. M1 macrophage derived exosomes (M1 exosomes) were found to aggravate EAN via boosting Th1 and Th17 response, while M2 macrophage derived exosomes (M2 exosomes) showed potentials to mitigate disease severity via a mechanism bypassing Th1 and Th17 response. Besides, both M1 and M2 exosomes increased germinal center reactions in EAN. Further in vitro studies confirmed that M1 exosomes could directly promote IFN-γ production in T cells and M2 exosomes were not capable of inhibiting IFN-γ expression. Thus, our data identify a previously undescribed means that M1 macrophages amplify Th1 response via exosomes and provide novel insights into the crosstalk between macrophages and T cells as well.
Collapse
Affiliation(s)
- Tong Du
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Chun-Lin Yang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Meng-Ru Ge
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ying Liu
- Department of Neuronal Electrophysiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tao Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yu-Dong Liu
- Department of Neuronal Electrophysiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Ying-Chun Dou
- College of Basic Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.,Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
87
|
Long X, Zhang L, Zhang Y, Min M, Lin B, Chen J, Ma X, Zhai S, Cai Z, Liu Y, Lu Y, Che N, Tan W, Qin J, Wang X. Histone methyltransferase Nsd2 is required for follicular helper T cell differentiation. J Exp Med 2020; 217:jem.20190832. [PMID: 31636135 PMCID: PMC7037247 DOI: 10.1084/jem.20190832] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/21/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
Regulation of Bcl6 expression during follicular helper T cell differentiation remains incompletely understood. Here, Long et al. show that T cell activation induces H3K36me2 methyltransferase Nsd2, in a CD28- and ICOS-dependent manner, to promote Bcl6 expression and Tfh differentiation. Follicular helper T (Tfh) cells provide essential help for humoral immune response. Transcriptional factor Bcl6 is the master regulator for Tfh generation and is induced very early after T cell activation in a CD28-dependent manner, but how CD28 signal promotes Bcl6 early expression remains unknown. Here we found that CD28 signal quickly induces expression of the H3K36me2 methytransferase Nsd2, which is required for Bcl6 expression as early as the first cell division after T cell activation. Nsd2 deficiency in T cells leads to decreased Bcl6 expression, impaired Tfh generation, compromised germinal center response, and delayed virus clearance. Ectopic Bcl6 expression rescues the Tfh defect of Nsd2 KO cells. ICOS signal is dispensable for early Nsd2 induction but required for sustained Nsd2 expression, which is critical for Tfh maintenance. Overexpression of Nsd2 increases Bcl6 expression and enhances Tfh generation; 4-mo-old mice even develop spontaneous Tfh. Overall, our study reveals Nsd2 as a critical epigenetic regulator for Tfh differentiation.
Collapse
Affiliation(s)
- Xuehui Long
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Le Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Min
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bichun Lin
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingjing Chen
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojie Ma
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sulan Zhai
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenming Cai
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yingxia Liu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanlai Lu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Che
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Qin
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Institute of Nutrition and Health Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
88
|
Olson WJ, Jakic B, Hermann‐Kleiter N. Regulation of the germinal center response by nuclear receptors and implications for autoimmune diseases. FEBS J 2020; 287:2866-2890. [PMID: 32246891 PMCID: PMC7497069 DOI: 10.1111/febs.15312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/15/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023]
Abstract
The immune system plays an essential role in protecting the host from infectious diseases and cancer. Notably, B and T lymphocytes from the adaptive arm of the immune system can co-operate to form long-lived antibody responses and are therefore the main target in vaccination approaches. Nevertheless, protective immune responses must be tightly regulated to avoid hyper-responsiveness and responses against self that can result in autoimmunity. Nuclear receptors (NRs) are perfectly adapted to rapidly alter transcriptional cellular responses to altered environmental settings. Their functional role is associated with both immune deficiencies and autoimmunity. Despite extensive linking of nuclear receptor function with specific CD4 T helper subsets, research on the functional roles and mechanisms of specific NRs in CD4 follicular T helper cells (Tfh) and germinal center (GC) B cells during the germinal center reaction is just emerging. We review recent advances in our understanding of NR regulation in specific cell types of the GC response and discuss their implications for autoimmune diseases such as systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- William J. Olson
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| | - Bojana Jakic
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
- Department of Immunology, Genetics and PathologyUppsala UniversitySweden
| | - Natascha Hermann‐Kleiter
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| |
Collapse
|
89
|
Choi J, Diao H, Faliti CE, Truong J, Rossi M, Bélanger S, Yu B, Goldrath AW, Pipkin ME, Crotty S. Bcl-6 is the nexus transcription factor of T follicular helper cells via repressor-of-repressor circuits. Nat Immunol 2020; 21:777-789. [PMID: 32572238 PMCID: PMC7449381 DOI: 10.1038/s41590-020-0706-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
T follicular helper (TFH) cells are a distinct type of CD4+ T cells that are essential for most antibody and B lymphocyte responses. TFH cell regulation and dysregulation is involved in a range of diseases. Bcl-6 is the lineage defining transcription factor of TFH cells and its activity is essential for TFH cell differentiation and function. However, how Bcl-6 controls TFH biology has largely remained unclear, at least in part due to intrinsic challenges of connecting repressors to gene upregulation in complex cell types with multiple possible differentiation fates. Multiple competing models were tested here by a series of experimental approaches to determine that Bcl-6 exhibited negative autoregulation and controlled pleiotropic attributes of TFH differentiation and function, including migration, costimulation, inhibitory receptors, and cytokines, via multiple repressor-of-repressor gene circuits.
Collapse
Affiliation(s)
- Jinyong Choi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Huitian Diao
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA
| | - Caterina E Faliti
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jacquelyn Truong
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Meghan Rossi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Simon Bélanger
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Bingfei Yu
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Ananda W Goldrath
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Matthew E Pipkin
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA. .,Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
90
|
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
|
91
|
Specific subfamilies of transposable elements contribute to different domains of T lymphocyte enhancers. Proc Natl Acad Sci U S A 2020; 117:7905-7916. [PMID: 32193341 PMCID: PMC7148579 DOI: 10.1073/pnas.1912008117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Transposable elements (TEs) compose nearly half of mammalian genomes and provide building blocks for cis-regulatory elements. Using high-throughput sequencing, we show that 84 TE subfamilies are overrepresented, and distributed in a lineage-specific fashion in core and boundary domains of CD8+ T cell enhancers. Endogenous retroviruses are most significantly enriched in core domains with accessible chromatin, and bear recognition motifs for immune-related transcription factors. In contrast, short interspersed elements (SINEs) are preferentially overrepresented in nucleosome-containing boundaries. A substantial proportion of these SINEs harbor a high density of the enhancer-specific histone mark H3K4me1 and carry sequences that match enhancer boundary nucleotide composition. Motifs with regulatory features are better preserved within enhancer-enriched TE copies compared to their subfamily equivalents located in gene deserts. TE-rich and TE-poor enhancers associate with both shared and unique gene groups and are enriched in overlapping functions related to lymphocyte and leukocyte biology. The majority of T cell enhancers are shared with other immune lineages and are accessible in common hematopoietic progenitors. A higher proportion of immune tissue-specific enhancers are TE-rich compared to enhancers specific to other tissues, correlating with higher TE occurrence in immune gene-associated genomic regions. Our results suggest that during evolution, TEs abundant in these regions and carrying motifs potentially beneficial for enhancer architecture and immune functions were particularly frequently incorporated by evolving enhancers. Their putative selection and regulatory cooption may have accelerated the evolution of immune regulatory networks.
Collapse
|
92
|
Chen X, Cao G, Wu J, Wang X, Pan Z, Gao J, Tian Q, Xu L, Li Z, Hao Y, Huang Q, Wang P, Xiao M, Xie L, Tang S, Liu Z, Hu L, Tang J, He R, Wang L, Zhou X, Wu Y, Chen M, Sun B, Zhu B, Huang J, Ye L. The histone methyltransferase EZH2 primes the early differentiation of follicular helper T cells during acute viral infection. Cell Mol Immunol 2020; 17:247-260. [PMID: 30842630 PMCID: PMC7052164 DOI: 10.1038/s41423-019-0219-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/17/2019] [Accepted: 02/17/2019] [Indexed: 12/12/2022] Open
Abstract
Epigenetic modifications to histones dictate the differentiation of naïve CD4+ T cells into different subsets of effector T helper (TH) cells. The histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been implicated in the mechanism regulating the differentiation of TH1, TH2 and regulatory T (Treg) cells. However, whether and how EZH2 regulates follicular helper T (TFH) cell differentiation remain unknown. Using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection, we observed abundant EZH2 expression and associated H3K27me3 modifications preferentially in the early committed virus-specific TFH cells compared to those in TH1 cells. Ablation of EZH2 in LCMV-specific CD4+ T cells leads to a selective impairment of early TFH cell fate commitment, but not late TFH differentiation or memory TFH maintenance. Mechanistically, EZH2 specifically stabilizes the chromatin accessibility of a cluster of genes that are important for TFH fate commitment, particularly B cell lymphoma 6 (Bcl6), and thus directs TFH cell commitment. Therefore, we identified the chromatin-modifying enzyme EZH2 as a novel regulator of early TFH differentiation during acute viral infection.
Collapse
Affiliation(s)
- Xiangyu Chen
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Guoshuai Cao
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Jialin Wu
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Xinxin Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, 400038, Chongqing, China
| | - Zhiwei Pan
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Jianbao Gao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, 400038, Chongqing, China
| | - Qin Tian
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Lifan Xu
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Zhirong Li
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Yaxing Hao
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Qizhao Huang
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, China
| | - Pengcheng Wang
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Minglu Xiao
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Luoyingzi Xie
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Shupei Tang
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Zhenyu Liu
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Li Hu
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Jianfang Tang
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Ran He
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Li Wang
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Xinyuan Zhou
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China
| | - Mengjie Chen
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210009, Jiangsu, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, 400038, Chongqing, China.
| | - Jun Huang
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA.
| | - Lilin Ye
- Institute of Immunology, PLA, Third Military Medical University, 400038, Chongqing, China.
| |
Collapse
|
93
|
Preite S, Gomez-Rodriguez J, Cannons JL, Schwartzberg PL. T and B-cell signaling in activated PI3K delta syndrome: From immunodeficiency to autoimmunity. Immunol Rev 2020; 291:154-173. [PMID: 31402502 DOI: 10.1111/imr.12790] [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: 05/20/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022]
Abstract
Phosphatidylinositol 3 kinases (PI3K) are a family of lipid kinases that are activated by a variety of cell-surface receptors, and regulate a wide range of downstream readouts affecting cellular metabolism, growth, survival, differentiation, adhesion, and migration. The importance of these lipid kinases in lymphocyte signaling has recently been highlighted by genetic analyses, including the recognition that both activating and inactivating mutations of the catalytic subunit of PI3Kδ, p110δ, lead to human primary immunodeficiencies. In this article, we discuss how studies on the human genetic disorder "Activated PI3K-delta syndrome" and mouse models of this disease (Pik3cdE1020K/+ mice) have provided fundamental insight into pathways regulated by PI3Kδ in T and B cells and their contribution to lymphocyte function and disease, including responses to commensal bacteria and the development of autoimmunity and tumors. We highlight critical roles of PI3Kδ in T follicular helper cells and the orchestration of the germinal center reaction, as well as in CD8+ T-cell function. We further present data demonstrating the ability of the AKT-resistant FOXO1AAA mutant to rescue IgG1 class switching defects in Pik3cdE1020K/+ B cells, as well as data supporting a role for PI3Kδ in promoting multiple T-helper effector cell lineages.
Collapse
Affiliation(s)
- Silvia Preite
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Julio Gomez-Rodriguez
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L Cannons
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Pamela L Schwartzberg
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
94
|
Mountz JD, Hsu HC, Ballesteros-Tato A. Dysregulation of T Follicular Helper Cells in Lupus. THE JOURNAL OF IMMUNOLOGY 2020; 202:1649-1658. [PMID: 30833421 DOI: 10.4049/jimmunol.1801150] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022]
Abstract
Although multiple and overlapping mechanisms are ultimately responsible for the immunopathology observed in patients with systemic lupus erythematosus, autoreactive Abs secreted by autoreactive plasma cells (PCs) are considered to play a critical role in disease progression and immunopathology. Given that PCs derive from the germinal centers (GC), long-term dysregulated GC reactions are often associated with the development of spontaneous autoantibody responses and immunopathology in systemic lupus erythematosus patients. In this review, we summarize the emerging evidence concerning the roles of T follicular helper cells in regulating pathogenic GC and autoreactive PC responses in lupus.
Collapse
Affiliation(s)
- John D Mountz
- Division of Clinical Immunology and Rheumatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and .,Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233
| | - Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Andre Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| |
Collapse
|
95
|
Gbedande K, Carpio VH, Stephens R. Using two phases of the CD4 T cell response to blood-stage murine malaria to understand regulation of systemic immunity and placental pathology in Plasmodium falciparum infection. Immunol Rev 2020; 293:88-114. [PMID: 31903675 PMCID: PMC7540220 DOI: 10.1111/imr.12835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Plasmodium falciparum infection and malaria remain a risk for millions of children and pregnant women. Here, we seek to integrate knowledge of mouse and human T helper cell (Th) responses to blood-stage Plasmodium infection to understand their contribution to protection and pathology. Although there is no complete Th subset differentiation, the adaptive response occurs in two phases in non-lethal rodent Plasmodium infection, coordinated by Th cells. In short, cellular immune responses limit the peak of parasitemia during the first phase; in the second phase, humoral immunity from T cell-dependent germinal centers is critical for complete clearance of rapidly changing parasite. A strong IFN-γ response kills parasite, but an excess of TNF compared with regulatory cytokines (IL-10, TGF-β) can cause immunopathology. This common pathway for pathology is associated with anemia, cerebral malaria, and placental malaria. These two phases can be used to both understand how the host responds to rapidly growing parasite and how it attempts to control immunopathology and variation. This dual nature of T cell immunity to Plasmodium is discussed, with particular reference to the protective nature of the continuous generation of effector T cells, and the unique contribution of effector memory T cells.
Collapse
Affiliation(s)
- Komi Gbedande
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Victor H Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Robin Stephens
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
96
|
Amatore F, Gorvel L, Olive D. Role of Inducible Co-Stimulator (ICOS) in cancer immunotherapy. Expert Opin Biol Ther 2019; 20:141-150. [PMID: 31738626 DOI: 10.1080/14712598.2020.1693540] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: The promotion of antitumor response by targeting co-stimulatory B7 superfamily members has become evident to create a new wave of cancer immunotherapy. Inducible Co-Stimulator (ICOS), which is expressed on activated T cells, gained interest in the translational medicine community.Areas covered: We performed an extensive literature review using the keywords 'ICOS' and 'cancer', and the Clinicaltrials.gov database for early phase clinical trials targeting ICOS. In this review, we highlight the dual role of ICOS in oncogenesis in different malignancies. We summarize the current state of knowledge about ICOS/ICOSL pathway targeting by immunotherapies.Expert opinion: Due to its multifaceted link with anti-tumor immunity, both antagonist and agonist antibodies might be of interest to target the ICOS/ICOSL pathway for tumor treatment. Indeed, ICOS activation might potentiate the effect of an inhibitory checkpoint blockade, while its neutralization could decrease the function of immunosuppressive Tregs and inhibit lymphoid tumor cells expressing Tfh markers.
Collapse
Affiliation(s)
- Florent Amatore
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| | - Laurent Gorvel
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| | - Daniel Olive
- Centre de recherche en Cancérologie de Marseille, INSERM U1068, CNRS U7258, Aix Marseille Université, Institut Paoli - Calmettes, Marseille, France
| |
Collapse
|
97
|
Ding T, Niu H, Zhao X, Gao C, Li X, Wang C. T-Follicular Regulatory Cells: Potential Therapeutic Targets in Rheumatoid Arthritis. Front Immunol 2019; 10:2709. [PMID: 31849938 PMCID: PMC6901970 DOI: 10.3389/fimmu.2019.02709] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is an incurable aggressive chronic inflammatory joint disease with a worldwide prevalence. High levels of autoantibodies and chronic inflammation may be involved in the pathology. Notably, T follicular regulatory (Tfr) cells are critical mediators of T follicular helper (Tfh) cell generation and antibody production in the germinal center (GC) reaction. Changes in the number and function of Tfr cells may lead to dysregulation of the GC reaction and the production of aberrant autoantibodies. Regulation of the function and number of Tfr cells could be an effective strategy for precisely controlling antibody production, reestablishing immune homeostasis, and thereby improving the outcome of RA. This review summarizes advances in our understanding of the biology and functions of Tfr cells. The involvement of Tfr cells and other immune cell subsets in RA is also discussed. Furthermore, we highlight the potential therapeutic targets related to Tfr cells and restoring the Tfr/Tfh balance via cytokines, microRNAs, the mammalian target of rapamycin (mTOR) signaling pathway, and the gut microbiota, which will facilitate further research on RA and other immune-mediated diseases.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongqing Niu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiangcong Zhao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital and Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
98
|
Tao H, Li L, Gao Y, Wang Z, Zhong XP. Differential Control of iNKT Cell Effector Lineage Differentiation by the Forkhead Box Protein O1 (Foxo1) Transcription Factor. Front Immunol 2019; 10:2710. [PMID: 31824499 PMCID: PMC6881238 DOI: 10.3389/fimmu.2019.02710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
The invariant NKT (iNKT) cells recognize glycolipid antigens presented by the non-classical MHC like molecule CD1d. They represent an innate T-cell lineage with the ability to rapidly produce a variety of cytokines in response to agonist stimulation to bridge innate and adaptive immunity. In thymus, most iNKT cells complete their maturation and differentiate to multiple effector lineages such as iNKT-1, iNKT-2, and iNKT-17 cells that possess the capability to produce IFNγ, IL-4, and IL-17A, respectively, and play distinct roles in immune responses and diseases. Mechanisms that control iNKT lineage fate decisions are still not well understood. Evidence has revealed critical roles of Foxo1 of the forkhead box O1 subfamily of transcription factors in the immune system. However, its role in iNKT cells has been unknown. In this report, we demonstrate that deletion of Foxo1 causes severe decreases of iNKT cell total numbers due to impairment of late but not early iNKT cell development. Deficiency of Foxo1 results in decreases of iNKT-1 but increases of iNKT-17 cells. Our data reveal that Foxo1 controls iNKT effector lineage fate decision by promoting iNKT-1 but suppressing iNKT-17 lineages.
Collapse
Affiliation(s)
- Huishan Tao
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Gao
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Ping Zhong
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,The Hematologic Malignancies and Cellular Therapy Research Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
99
|
Kikuchi T, Nakae J, Kawano Y, Watanabe N, Onodera M, Itoh H. Foxo in T Cells Regulates Thermogenic Program through Ccr4/Ccl22 Axis. iScience 2019; 22:81-96. [PMID: 31756626 PMCID: PMC6880116 DOI: 10.1016/j.isci.2019.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 09/21/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
Crosstalk between immunity and the thermogenic program has provided insight into metabolic energy regulation. Here, we generated thermogenic program-accelerating mice (T-QKO), in which Foxo1 is knockout and Foxo3 is hetero-knockout in CD4+ T cells. T-QKO exhibit lean phenotype under HFD due to increased energy expenditure. Cold exposure significantly increased expression of the thermogenic genes (Ppargc1a and Ucp1), Th2 cytokines (Il4 and Il13), and Th2 marker gene (Gata3) in subcutaneous adipose tissue (SC) of T-QKO. Furthermore, Ccr4 expression was significantly increased in Th2 cells of T-QKO, and cold exposure induced Ccl22 expression in SC, leading to increased accumulation of Th2 cell population in SC of T-QKO. These data reveal a mechanism by which cold exposure induces selective recruitment of Th2 cells into SC, leading to regulation of energy expenditure by generating beige adipocyte and suggest that inhibition of Foxo in T cells may support a strategy to prevent and treat obesity. T-QKO increases Gata3 and Ccr4 expression in CD4+ T cells Cold exposure increases Ccl22 expression in subcutaneous adipose tissue Cold exposure increases SC-specific recruitment of Th2 cells in T-QKO Recruited Th2 cells secrete IL-4 and IL-13 and increase beiging of adipocytes
Collapse
Affiliation(s)
- Tetsuhiro Kikuchi
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jun Nakae
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Physiology, International University of Health and Welfare School of Medicine, Narita 286-8686, Japan.
| | - Yoshinaga Kawano
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Hiroshi Itoh
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
100
|
Mondragón L, Mhaidly R, De Donatis GM, Tosolini M, Dao P, Martin AR, Pons C, Chiche J, Jacquin M, Imbert V, Proïcs E, Boyer L, Doye A, Luciano F, Neels JG, Coutant F, Fabien N, Sormani L, Rubio-Patiño C, Bossowski JP, Muller F, Marchetti S, Villa E, Peyron JF, Gaulard P, Lemonnier F, Asnafi V, Genestier L, Benhida R, Fournié JJ, Passeron T, Ricci JE, Verhoeyen E. GAPDH Overexpression in the T Cell Lineage Promotes Angioimmunoblastic T Cell Lymphoma through an NF-κB-Dependent Mechanism. Cancer Cell 2019; 36:268-287.e10. [PMID: 31447347 DOI: 10.1016/j.ccell.2019.07.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 04/17/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
GAPDH is emerging as a key player in T cell development and function. To investigate the role of GAPDH in T cells, we generated a transgenic mouse model overexpressing GAPDH in the T cell lineage. Aged mice developed a peripheral Tfh-like lymphoma that recapitulated key molecular, pathological, and immunophenotypic features of human angioimmunoblastic T cell lymphoma (AITL). GAPDH induced non-canonical NF-κB pathway activation in mouse T cells, which was strongly activated in human AITL. We developed a NIK inhibitor to reveal that targeting the NF-κB pathway prolonged AITL-bearing mouse survival alone and in combination with anti-PD-1. These findings suggest the therapeutic potential of targeting NF-κB signaling in AITL and provide a model for future AITL therapeutic investigations.
Collapse
Affiliation(s)
| | - Rana Mhaidly
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Marie Tosolini
- Pôle Technologique du CRCT - Plateau Bioinformatique INSERM-UMR 1037, Toulouse, France
| | - Pascal Dao
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Anthony R Martin
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Caroline Pons
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Marie Jacquin
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Emma Proïcs
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Laurent Boyer
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Anne Doye
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Jaap G Neels
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Frédéric Coutant
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon, Edouard Herriot Hospital, Lyon, France
| | - Nicole Fabien
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Laura Sormani
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | | | | | | | - Elodie Villa
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Philippe Gaulard
- Université Paris-Est Créteil, Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France; Département de Pathologie, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Créteil, France
| | - François Lemonnier
- Université Paris-Est Créteil, Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Créteil, France
| | - Vahid Asnafi
- Université Paris 5, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker-Enfants Malades, Paris, France
| | - Laurent Genestier
- CRCL, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon I, 69921 Oullins Cedex, France
| | - Rachid Benhida
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Jean-Jacques Fournié
- CRCT, INSERM U1037 - Université Paul Sabatier - CNRS ERL5294, Université de Toulouse, Laboratoire d'Excellence TOUCAN, Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France; IUCT, 31037 Toulouse, France
| | - Thierry Passeron
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France; Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Dermatology, 06204 Nice, France
| | | | - Els Verhoeyen
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France; CIRI, Université de Lyon, INSERM U1111, ENS de Lyon, Université Lyon 1, CNRS, UMR 5308, 69007 Lyon, France.
| |
Collapse
|