1
|
Zhang Z, Zhang L, Wang K, Xie T, Zhang X, Yu W, Li Y, Shen L, Li R, Peng Z. Single-cell landscape of bronchoalveolar immune cells in patients with immune checkpoint inhibitor-related pneumonitis. NPJ Precis Oncol 2024; 8:226. [PMID: 39369126 PMCID: PMC11455925 DOI: 10.1038/s41698-024-00715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024] Open
Abstract
The pathophysiology of immune checkpoint inhibitor-related pneumonitis remains incompletely understood. We conducted single-cell and T-cell receptor transcriptomic sequencing on the bronchoalveolar lavage fluid from five patients with grade ≥2 immune checkpoint inhibitor-related pneumonitis. Our analyses revealed a prominent enrichment of T cells in the bronchoalveolar lavage fluid of patients with immune checkpoint inhibitor-related pneumonitis. Within the CD4 + T cell subset, Tfh-like T cells were highly enriched and exhibited signatures associated with inflammation and clonal expansion. Regulatory T cells were also enriched and displayed enhanced inhibitory functions. Within the CD8 + T-cell subset, effector memory/tissue-resident memory T cells with an elevated cytotoxic phenotype were highly infiltrated. Among myeloid cells, alveolar macrophages were depleted, while pro-inflammatory intermediate monocytes were elevated. Dendritic cells demonstrated enhanced antigen presentation capabilities. Cytokines CXCR4, CXCL13, TNF-α, IFN-α, IFN-γ, and TWEAK were elevated. Through a comprehensive single-cell analysis, we depicted the landscape of immune checkpoint inhibitor-related pneumonitis.
Collapse
Affiliation(s)
- Zhening Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Immunobiology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Lei Zhang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Keqiang Wang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Tong Xie
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yanyan Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ran Li
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China.
| | - Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
2
|
Evans ET, Page EF, Choi AS, Shonibare Z, Kahn AG, Arend RC, Mythreye K. Activin levels correlate with lymphocytic infiltration in epithelial ovarian cancer. Cancer Med 2024; 13:e7368. [PMID: 39248018 PMCID: PMC11381957 DOI: 10.1002/cam4.7368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 09/10/2024] Open
Abstract
OBJECTIVE The TGF-β superfamily member activin, a dimer of the gene products of INHBA and/or INHBB, has been implicated in immune cell maturation and recruitment, but its immune impact within epithelial ovarian cancer (EOC) is not well characterized. We sought to explore differences in activin (INHBA/ Inhibin-βA and INHBB/ Inhibin-βB) between malignant and ovarian tissues at the RNA and protein level and assess the relationship between activin and immune cells in EOC. METHODS Publicly available RNA sequencing data were accessed from GEO (#GSE143897) with normalization and quantification performed via DESeq2. Immune gene expression profile was further explored within the TCGA-OV cohort derived from The Cancer Genome Atlas (TCGA). Immunohistochemical analysis was performed to evaluate activin A and T-cell markers CD8 and FoxP3 at the protein level. ELISA to activin-A was used to assess levels in the ascites of advanced EOC patients. Kaplan-Meier curves were generated to visualize survival outcomes. RESULTS Gene expression levels of components of the activin signaling pathway were elevated within EOC when compared to a benign cohort, with differences in activin type I/II receptor gene profiles identified. Additionally, INHBA gene expression was linked to lymphocytic immune markers in EOC samples. Immunohistochemistry analysis revealed a positive correlation of CD8 and FOXP3 staining with activin A at the protein level in both primary and metastatic epithelial ovarian cancer samples. Furthermore, Activin-A (inhibin-βA) is significantly elevated in EOC patient ascites. CONCLUSION INHBA expression is elevated within EOC, correlating with worse survival, with activin protein levels correlating with specific immune infiltration. Our findings suggest that activin-A may play a role in suppressing anti-tumor immunity in EOC, highlighting its potential as a therapeutic target.
Collapse
MESH Headings
- Humans
- Female
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Ovarian Epithelial/immunology
- Carcinoma, Ovarian Epithelial/metabolism
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/mortality
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/genetics
- Inhibin-beta Subunits/genetics
- Inhibin-beta Subunits/metabolism
- Activins/metabolism
- Activins/genetics
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Middle Aged
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
Collapse
Affiliation(s)
- Elizabeth T Evans
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Emily F Page
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Alex Seok Choi
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Zainab Shonibare
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Andrea G Kahn
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Rebecca C Arend
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Karthikeyan Mythreye
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
3
|
Nieuwenhuizen NE, Nouailles G, Sutherland JS, Zyla J, Pasternack AH, Heyckendorf J, Frye BC, Höhne K, Zedler U, Bandermann S, Abu Abed U, Brinkmann V, Gutbier B, Witzenrath M, Suttorp N, Zissel G, Lange C, Ritvos O, Kaufmann SHE. Activin A levels are raised during human tuberculosis and blockade of the activin signaling axis influences murine responses to M. tuberculosis infection. mBio 2024; 15:e0340823. [PMID: 38376260 PMCID: PMC10936190 DOI: 10.1128/mbio.03408-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Activin A strongly influences immune responses; yet, few studies have examined its role in infectious diseases. We measured serum activin A levels in two independent tuberculosis (TB) patient cohorts and in patients with pneumonia and sarcoidosis. Serum activin A levels were increased in TB patients compared to healthy controls, including those with positive tuberculin skin tests, and paralleled severity of disease, assessed by X-ray scores. In pneumonia patients, serum activin A levels were also raised, but in sarcoidosis patients, levels were lower. To determine whether blockade of the activin A signaling axis could play a functional role in TB, we harnessed a soluble activin type IIB receptor fused to human IgG1 Fc, ActRIIB-Fc, as a ligand trap in a murine TB model. The administration of ActRIIB-Fc to Mycobacterium tuberculosis-infected mice resulted in decreased bacterial loads and increased numbers of CD4 effector T cells and tissue-resident memory T cells in the lung. Increased frequencies of tissue-resident memory T cells corresponded with downregulated T-bet expression in lung CD4 and CD8 T cells. Altogether, the results suggest a disease-exacerbating role of ActRIIB signaling pathways. Serum activin A may be useful as a biomarker for diagnostic triage of active TB or monitoring of anti-tuberculosis therapy. IMPORTANCE Tuberculosis remains the leading cause of death by a bacterial pathogen. The etiologic agent of tuberculosis, Mycobacterium tuberculosis, can remain dormant in the infected host for years before causing disease. Significant effort has been made to identify biomarkers that can discriminate between latently infected and actively diseased individuals. We found that serum levels of the cytokine activin A were associated with increased lung pathology and could discriminate between active tuberculosis and tuberculin skin-test-positive healthy controls. Activin A signals through the ActRIIB receptor, which can be blocked by administration of the ligand trap ActRIIB-Fc, a soluble activin type IIB receptor fused to human IgG1 Fc. In a murine model of tuberculosis, we found that ActRIIB-Fc treatment reduced mycobacterial loads. Strikingly, ActRIIB-Fc treatment significantly increased the number of tissue-resident memory T cells. These results suggest a role for ActRIIB signaling pathways in host responses to Mycobacterium tuberculosis and activin A as a biomarker of ongoing disease.
Collapse
Affiliation(s)
- Natalie E. Nieuwenhuizen
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Geraldine Nouailles
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jayne S. Sutherland
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Joanna Zyla
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Arja H. Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jan Heyckendorf
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Björn C. Frye
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kerstin Höhne
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrike Zedler
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Silke Bandermann
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Ulrike Abu Abed
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Volker Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
| | - Birgitt Gutbier
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Gernot Zissel
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Lange
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - the CAPNETZ Study group
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - the DZIF TB study group
- Department of Immunology, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Institute for Hygiene and Microbiology, Julius Maximilian University of Würzburg, Würzburg, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Pneumology, Clinic, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- CAPNETZ STIFTUNG, Hannover, Germany
- German Center for Lung Research (DZL), Berlin, Germany
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
- Baylor College of Medicine and Texas Children´s Hospital, Global TB Program, Houston, Texas, USA
- Max Planck Institute for Multidisciplinary Sciences, Emeritus Group Systems Immunology, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
4
|
Zheng Y, Lebid A, Chung L, Fu J, Wang X, Otrocol A, Zarif JC, Yu H, Llosa NJ, Pardoll DM. Targeting the activin receptor 1C on CD4+ T cells for cancer immunotherapy. Oncoimmunology 2024; 13:2297503. [PMID: 38235319 PMCID: PMC10793694 DOI: 10.1080/2162402x.2023.2297503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
Activins, members of the TGF-beta superfamily, have been isolated and identified in the endocrine system, but have not been substantially investigated in the context of the immune system and endocrine-unrelated cancers. Here, we demonstrated that tumor-bearing mice had elevated systemic activin levels, which correlated directly with tumor burden. Likewise, cancer patients have elevated plasma activin levels compared to healthy controls. We observed that both tumor and immune cells could be sources of activins. Importantly, our in vitro studies suggest that activins promote differentiation of naïve CD4+ cells into Foxp3-expressing induced regulatory T cells (Tregs), particularly when TGF-beta was limited in the culture medium. Database and qRT-PCR analysis of sorted major immune cell subsets in mice revealed that activin receptor 1c (ActRIC) was uniquely expressed on Tregs and that both ActRIC and ActRIIB (activin receptor 2b) were highly upregulated during iTreg differentiation. ActRIC-deficient naïve CD4+ cells were found to be defective in iTreg generation both in vitro and in vivo. Treg suppression assays were also performed, and ActRIC deficiency did not change the function or stability of iTregs. Mice lacking ActRIC or mice treated with monoclonal anti-ActRIC antibody were more resistant to tumor progression than wild-type controls. This phenotype was correlated with reduced expression of Foxp3 in CD4+ cells in the tumor microenvironment. In light of the information presented above, blocking activin-ActRIC signaling is a promising and disease-specific strategy to impede the accumulation of immunosuppressive iTregs in cancer. Therefore, it is a potential candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Zheng
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andriana Lebid
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liam Chung
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juan Fu
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoxu Wang
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrea Otrocol
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jelani C. Zarif
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hong Yu
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicolas J. Llosa
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M. Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Yull S, Shafiei S, Park CB, Kazemi P, Tiemann EB, Pagé MHG, Dufort D. Uterine Nodal expression supports maternal immunotolerance and establishment of the FOXP3 + regulatory T cell population during the preimplantation period. Front Immunol 2023; 14:1276979. [PMID: 38022561 PMCID: PMC10646213 DOI: 10.3389/fimmu.2023.1276979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Pregnancy success is dependent on the establishment of maternal tolerance during the preimplantation period. The immunosuppressive function of regulatory T cells is critical to limit inflammation arising from implantation of the semi-allogeneic blastocyst. Insufficient maternal immune adaptations to pregnancy have been frequently associated with cases of female infertility and recurrent implantation failure. The role of Nodal, a secreted morphogen of the TGFβ superfamily, was recently implicated during murine pregnancy as its conditional deletion (NodalΔ/Δ) in the female reproductive tract resulted in severe subfertility. Here, it was determined that despite normal preimplantation processes and healthy, viable embryos, NodalΔ/Δ females had a 50% implantation failure rate compared to NodalloxP/loxP controls. Prior to implantation, the expression of inflammatory cytokines MCP-1, G-CSF, IFN-γ and IL-10 was dysregulated in the NodalΔ/Δ uterus. Further analysis of the preimplantation leukocyte populations in NodalΔ/Δ uteri showed an overabundance of infiltrating, pro-inflammatory CD11bhigh Ly6C+ macrophages coupled with the absence of CD4+ FOXP3+ regulatory T cells. Therefore, it is proposed that uterine Nodal expression during the preimplantation period has a novel role in the establishment of maternal immunotolerance, and its dysregulation should be considered as a potential contributor to cases of female infertility and recurrent implantation failure.
Collapse
Affiliation(s)
- Sarah Yull
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Shiva Shafiei
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Craig B. Park
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Parinaz Kazemi
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | | | - Marie-Hélène Godin Pagé
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Daniel Dufort
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
7
|
Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023; 20:1002-1022. [PMID: 37217798 PMCID: PMC10468540 DOI: 10.1038/s41423-023-01036-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Balanced immunity is pivotal for health and homeostasis. CD4+ helper T (Th) cells are central to the balance between immune tolerance and immune rejection. Th cells adopt distinct functions to maintain tolerance and clear pathogens. Dysregulation of Th cell function often leads to maladies, including autoimmunity, inflammatory disease, cancer, and infection. Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance. It is therefore critical to understand how Treg and Th17 cells are regulated in health and disease. Cytokines are instrumental in directing Treg and Th17 cell function. The evolutionarily conserved TGF-β (transforming growth factor-β) cytokine superfamily is of particular interest because it is central to the biology of both Treg cells that are predominantly immunosuppressive and Th17 cells that can be proinflammatory, pathogenic, and immune regulatory. How TGF-β superfamily members and their intricate signaling pathways regulate Treg and Th17 cell function is a question that has been intensely investigated for two decades. Here, we introduce the fundamental biology of TGF-β superfamily signaling, Treg cells, and Th17 cells and discuss in detail how the TGF-β superfamily contributes to Treg and Th17 cell biology through complex yet ordered and cooperative signaling networks.
Collapse
Affiliation(s)
- Junying Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xingqi Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
8
|
Wiley MB, Bauer J, Mehrotra K, Zessner-Spitzenberg J, Kolics Z, Cheng W, Castellanos K, Nash MG, Gui X, Kone L, Maker AV, Qiao G, Reddi D, Church DN, Kerr RS, Kerr DJ, Grippo PJ, Jung B. Non-Canonical Activin A Signaling Stimulates Context-Dependent and Cellular-Specific Outcomes in CRC to Promote Tumor Cell Migration and Immune Tolerance. Cancers (Basel) 2023; 15:3003. [PMID: 37296966 PMCID: PMC10252122 DOI: 10.3390/cancers15113003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
We have shown that activin A (activin), a TGF-β superfamily member, has pro-metastatic effects in colorectal cancer (CRC). In lung cancer, activin activates pro-metastatic pathways to enhance tumor cell survival and migration while augmenting CD4+ to CD8+ communications to promote cytotoxicity. Here, we hypothesized that activin exerts cell-specific effects in the tumor microenvironment (TME) of CRC to promote anti-tumoral activity of immune cells and the pro-metastatic behavior of tumor cells in a cell-specific and context-dependent manner. We generated an Smad4 epithelial cell specific knockout (Smad4-/-) which was crossed with TS4-Cre mice to identify SMAD-specific changes in CRC. We also performed IHC and digital spatial profiling (DSP) of tissue microarrays (TMAs) obtained from 1055 stage II and III CRC patients in the QUASAR 2 clinical trial. We transfected the CRC cells to reduce their activin production and injected them into mice with intermittent tumor measurements to determine how cancer-derived activin alters tumor growth in vivo. In vivo, Smad4-/- mice displayed elevated colonic activin and pAKT expression and increased mortality. IHC analysis of the TMA samples revealed increased activin was required for TGF-β-associated improved outcomes in CRC. DSP analysis identified that activin co-localization in the stroma was coupled with increases in T-cell exhaustion markers, activation markers of antigen presenting cells (APCs), and effectors of the PI3K/AKT pathway. Activin-stimulated PI3K-dependent CRC transwell migration, and the in vivo loss of activin lead to smaller CRC tumors. Taken together, activin is a targetable, highly context-dependent molecule with effects on CRC growth, migration, and TME immune plasticity.
Collapse
Affiliation(s)
- Mark B. Wiley
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Jessica Bauer
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Kunaal Mehrotra
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Jasmin Zessner-Spitzenberg
- Clinical Department for Gastroenterology and Hepatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Zoe Kolics
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Wenxuan Cheng
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| | - Karla Castellanos
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Michael G. Nash
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Xianyong Gui
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Lyonell Kone
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ajay V. Maker
- Department of Surgery, University of California-San Francisco, San Francisco, CA 94115, USA
| | - Guilin Qiao
- Department of Surgery, University of California-San Francisco, San Francisco, CA 94115, USA
| | - Deepti Reddi
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - David N. Church
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 4BH, UK
- NIHR Oxford Comprehensive Biomedical Research Center, Oxford University Hospitals NHS Foundation Trust, University of Oxford, Oxford OX1 4BH, UK
| | - Rachel S. Kerr
- Department of Oncology, University of Oxford, Oxford OX1 4BH, UK
| | - David J. Kerr
- Radcliffe Department of Medicine, University of Oxford, Oxford OX1 4BH, UK
| | - Paul J. Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Barbara Jung
- Department of Medicine, University of Washington, Seattle, WA 98195, USA; (M.B.W.); (K.M.)
| |
Collapse
|
9
|
Evans ET, Horst B, Arend RC, Mythreye K. Evolving roles of activins and inhibins in ovarian cancer pathophysiology. Am J Physiol Cell Physiol 2023; 324:C428-C437. [PMID: 36622068 PMCID: PMC9902228 DOI: 10.1152/ajpcell.00178.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023]
Abstract
Activins and inhibins are unique members of the transforming growth factor-β (TGFβ) family of growth factors, with the ability to exert autocrine, endocrine, and paracrine effects in a wide range of complex physiologic and pathologic processes. Although first isolated within the pituitary, emerging evidence suggests broader influence beyond reproductive development and function. Known roles of activin and inhibin in angiogenesis and immunity along with correlations between gene expression and cancer prognosis suggest potential roles in tumorigenesis. Here, we present a review of the current understanding of the biological role of activins and inhibins as it relates to ovarian cancers, summarizing the underlying signaling mechanisms and physiologic influence, followed by detailing their roles in cancer progression, diagnosis, and treatment.
Collapse
Affiliation(s)
- Elizabeth T Evans
- Department of Gynecologic Oncology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama
| | - Ben Horst
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rebecca C Arend
- Department of Gynecologic Oncology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama
| | - Karthikeyan Mythreye
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Cheng Z, Xiong X, Wu F, Zhao Y, Dong R, Jiang S, Fang K, Huang P, Chen G. Network Pharmacology Research Indicates that Wu-Mei-Wan Treats Obesity by Inhibiting Th17 Cell Differentiation and Alleviating Metabolic Inflammation. Comb Chem High Throughput Screen 2023; 26:30-48. [PMID: 35189797 DOI: 10.2174/1386207325666220221121919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Wu-Mei-Wan (WMW), a traditional Chinese medicine (TCM) formula, has a good effect on the treatment of obesity and has been proven helpful to promote the metabolism of adipose tissue. However, its underlying mechanism remains to be studied. This study aims to explore the potential pharmacological mechanism of WMW in the treatment of obesity. METHODS Network pharmacology was used to sort out the relationship between WMW putative targets and obesity-related drug targets or disease targets, which indicated the mechanism of WMW in treating obesity from two aspects of clinical drugs approved by the Food and Drug Administration (FDA) and obesity-related diseases. Databases such as Traditional Chinese Medicine Systems Pharmacology (TCMSP), PubChem, DrugBank, DisGeNET, and Genecards were used to collect information about targets. String platform was used to convert the data into gene symbol of "homo sapiens", and perform gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. With the Human Protein Reference Database (HPRD) as background data, Cytoscape 3.6.0 software was used to construct a new protein-protein interaction (PPI) network. Mechanism diagrams of key pathways were obtained from the KEGG database. AutoDock Vina software was used to conduct molecular docking verification. RESULTS The number of targets in the overlap between WMW putative targets and obesity-related drug targets accounted for more than 50% of the latter, and HTR3A, SLC6A4, and CYP3A4 were core targets. In obesity-related disease targets-WMW putative targets PPI network, the Th17 cell differentiation pathway, and the IL-17 signaling pathway were key pathways, and the 1st module and the 7th module were central function modules that were highly associated with immunity and inflammation. Molecular docking verified that STAT3, TGFB1, MMP9, AHR, IL1B, and CCL2 were core targets in the treatment of WMW on obesity. CONCLUSION WMW has similar effects on lipid and drug metabolism as the current obesity-related drugs, and is likely to treat obesity by inhibiting Th17 cell differentiation and alleviating metabolic inflammation.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinyu Xiong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Wu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Zhao
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruolan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shujun Jiang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Panpan Huang
- Key Laboratory of Chinese Medicine Compound and Chinese Medicine Resources Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, China
- Basic Medical Sciences College, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
11
|
Ganjoo S, Puebla-Osorio N, Nanez S, Hsu E, Voss T, Barsoumian H, Duong LK, Welsh JW, Cortez MA. Bone morphogenetic proteins, activins, and growth and differentiation factors in tumor immunology and immunotherapy resistance. Front Immunol 2022; 13:1033642. [PMID: 36353620 PMCID: PMC9638036 DOI: 10.3389/fimmu.2022.1033642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2024] Open
Abstract
The TGF-β superfamily is a group of secreted polypeptides with key roles in exerting and regulating a variety of physiologic effects, especially those related to cell signaling, growth, development, and differentiation. Although its central member, TGF-β, has been extensively reviewed, other members of the family-namely bone morphogenetic proteins (BMPs), activins, and growth and differentiation factors (GDFs)-have not been as thoroughly investigated. Moreover, although the specific roles of TGF-β signaling in cancer immunology and immunotherapy resistance have been extensively reported, little is known of the roles of BMPs, activins, and GDFs in these domains. This review focuses on how these superfamily members influence key immune cells in cancer progression and resistance to treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Pinjusic K, Dubey OA, Egorova O, Nassiri S, Meylan E, Faget J, Constam DB. Activin-A impairs CD8 T cell-mediated immunity and immune checkpoint therapy response in melanoma. J Immunother Cancer 2022; 10:jitc-2022-004533. [PMID: 35580932 PMCID: PMC9125758 DOI: 10.1136/jitc-2022-004533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 12/16/2022] Open
Abstract
Background Activin-A, a transforming growth factor β family member, is secreted by many cancer types and is often associated with poor disease prognosis. Previous studies have shown that Activin-A expression can promote cancer progression and reduce the intratumoral frequency of cytotoxic T cells. However, the underlying mechanisms and the significance of Activin-A expression for cancer therapies are unclear. Methods We analyzed the expression of the Activin-A encoding gene INHBA in melanoma patients and the influence of its gain- or loss-of-function on the immune infiltration and growth of BRAF-driven YUMM3.3 and iBIP2 mouse melanoma grafts and in B16 models. Using antibody depletion strategies, we investigated the dependence of Activin-A tumor-promoting effect on different immune cells. Immune-regulatory effects of Activin-A were further characterized in vitro and by an adoptive transfer of T cells. Finally, we assessed INHBA expression in melanoma patients who received immune checkpoint therapy and tested whether it impairs the response in preclinical models. Results We show that Activin-A secretion by melanoma cells inhibits adaptive antitumor immunity irrespective of BRAF status by inhibiting CD8+ T cell infiltration indirectly and even independently of CD4 T cells, at least in part by attenuating the production of CXCL9/10 by myeloid cells. In addition, we show that Activin-A/INHBA expression correlates with anti-PD1 therapy resistance in melanoma patients and impairs the response to dual anti-cytotoxic T-Lymphocyte associated protein 4/anti-PD1 treatment in preclinical models. Conclusions Our findings suggest that strategies interfering with Activin-A induced immune-regulation offer new therapeutic opportunities to overcome CD8 T cell exclusion and immunotherapy resistance.
Collapse
Affiliation(s)
- Katarina Pinjusic
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Olivier Andreas Dubey
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Olga Egorova
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Sina Nassiri
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Etienne Meylan
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.,Laboratory of Immuno-Oncology, Bordet Cancer Research Laboratories, Institut Jules Bordet, Faculty of Medicine, and Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Julien Faget
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.,Equipe Immunity and Cancer IRCM, INSERM U1194, Montpellier, France
| | - Daniel Beat Constam
- School of Life Sciences (SV), ISREC, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Abstract
The transforming growth factor-β (TGF-β) family includes cytokines controlling cell behavior, differentiation and homeostasis of various tissues including components of the immune system. Despite well recognized importance of TGF-β in controlling T cell functions, the immunomodulatory roles of many other members of the TGF-β cytokine family, especially bone morphogenetic proteins (BMPs), start to emerge. Bone Morphogenic Protein Receptor 1α (BMPR1α) is upregulated by activated effector and Foxp3+ regulatory CD4+ T cells (Treg cells) and modulates functions of both of these cell types. BMPR1α inhibits generation of proinflammatory Th17 cells and sustains peripheral Treg cells. This finding underscores the importance of the BMPs in controlling Treg cell plasticity and transition between Treg and Th cells. BMPR1α deficiency in in vitro induced and peripheral Treg cells led to upregulation of Kdm6b (Jmjd3) demethylase, an antagonist of polycomb repressive complex 2 (PRC2), and cell cycle inhibitor Cdkn1a (p21Cip1) promoting cell senescence. This indicates that BMPs and BMPR1α may represent regulatory modules shaping epigenetic landscape and controlling proinflammatory reprogramming of Th and Treg cells. Revealing functions of other BMP receptors and their crosstalk with receptors for TGF-β will contribute to our understanding of peripheral immunoregulation.
Collapse
Affiliation(s)
- Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
14
|
Browning LM, Miller C, Kuczma M, Pietrzak M, Jing Y, Rempala G, Muranski P, Ignatowicz L, Kraj P. Bone Morphogenic Proteins Are Immunoregulatory Cytokines Controlling FOXP3 + T reg Cells. Cell Rep 2021; 33:108219. [PMID: 33027660 DOI: 10.1016/j.celrep.2020.108219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 07/28/2020] [Accepted: 09/10/2020] [Indexed: 12/23/2022] Open
Abstract
Bone morphogenic proteins (BMPs) are members of the transforming growth factor β (TGF-β) cytokine family promoting differentiation, homeostasis, and self-renewal of multiple tissues. We show that signaling through the bone morphogenic protein receptor 1α (BMPR1α) sustains expression of FOXP3 in Treg cells in peripheral lymphoid tissues. BMPR1α signaling promotes molecular circuits supporting acquisition and preservation of Treg cell phenotype and inhibiting differentiation of pro-inflammatory effector Th1/Th17 CD4+ T cell. Mechanistically, increased expression of KDM6B (JMJD3) histone demethylase, an antagonist of the polycomb repressive complex 2, underlies lineage-specific changes of T cell phenotypes associated with abrogation of BMPR1α signaling. These results reveal that BMPs are immunoregulatory cytokines mediating maturation and stability of peripheral FOXP3+ regulatory T cells (Treg cells) and controlling generation of iTreg cells. Thus, we establish that BMPs, a large cytokine family, are an essential link between stromal tissues and the adaptive immune system involved in sustaining tissue homeostasis by promoting immunological tolerance.
Collapse
Affiliation(s)
- Lauren M Browning
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Caroline Miller
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Michal Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Yu Jing
- Center for Bioelectrics, Old Dominion University, Norfolk, VA 23529, USA
| | - Grzegorz Rempala
- College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Pawel Muranski
- Columbia University Medical Center, New York, NY 10032, USA
| | - Leszek Ignatowicz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
15
|
Roy U, de Oliveira RS, Galvez EJC, Gronow A, Basic M, Perez LG, Gagliani N, Bleich A, Huber S, Strowig T. Induction of IL-22-Producing CD4+ T Cells by Segmented Filamentous Bacteria Independent of Classical Th17 Cells. Front Immunol 2021; 12:671331. [PMID: 34566952 PMCID: PMC8456099 DOI: 10.3389/fimmu.2021.671331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/11/2021] [Indexed: 01/13/2023] Open
Abstract
The intestinal microbiota modulates IL-22 production in the intestine, including the induction of IL-22-producing CD4+ T helper cells. Which specific bacteria are responsible for the induction of these cells is less well understood. Here, we demonstrate through the use of novel gnotobiotic knock-in reporter mice that segmented filamentous bacteria (SFB), which are known for their ability to induce Th17 cells, also induce distinct IL-17A negative CD4+ T cell populations in the intestine. A subset of these cells instead produces IL-22 upon restimulation ex vivo and also during enteric infections. Furthermore, they produce a distinct set of cytokines compared to Th17 cells including the differential expression of IL-17F and IFN-γ. Importantly, genetic models demonstrate that these cells, presumably Th22 cells, develop independently of intestinal Th17 cells. Together, our data identifies that besides Th17, SFB also induces CD4+ T cell populations, which serve as immediate source of IL-22 during intestinal inflammation.
Collapse
Affiliation(s)
- Urmi Roy
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Rômulo S. de Oliveira
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Eric J. C. Galvez
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Achim Gronow
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Laura Garcia Perez
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
16
|
Ruffner MA, Zhang Z, Maurer K, Muir AB, Cianferoni A, Sullivan KE, Spergel JM. RNA sequencing identifies global transcriptional changes in peripheral CD4 + cells during active oesophagitis and following epicutaneous immunotherapy in eosinophilic oesophagitis. Clin Transl Immunology 2021; 10:e1314. [PMID: 34322233 PMCID: PMC8296633 DOI: 10.1002/cti2.1314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 04/28/2021] [Accepted: 06/25/2021] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE There are no disease-modifying therapies for the treatment of eosinophilic oesophagitis (EoE), which is driven by non-IgE-mediated allergic inflammation. A recent clinical trial of milk epicutaneous immunotherapy (EPIT) has shown initial promise, with 47% of treated EoE patients tolerating milk without recurrence of disease. Mechanisms of EPIT in EoE have not been studied in humans. Here, we identify transcriptional changes in the peripheral CD4+ T-cell compartment during active EoE and following EPIT. METHODS RNA isolation, sequencing and integrative data analysis were performed on peripheral CD4+ T cells isolated from 15 of 20 patients enrolled in a clinical trial of EPIT for EoE. Gene expression changes in peripheral CD4+ T cells were examined during diet therapy and following trial of milk antigen EPIT. RESULTS We identify 244 differentially expressed genes in peripheral blood CD4+ cells of EoE patients consuming versus those eliminating milk, and 129 DEGs in CD4+ cells were isolated after EPIT versus after placebo (FDR ≤ 0.05). Gene set enrichment analysis identifies enrichment of hallmark interferon-α and interferon-γ response pathways in peripheral CD4+ T cells from EoE patients during active disease on a milk-containing diet. We demonstrate overlap of this gene signature with the altered gene expression signature seen in EoE patient biopsy tissue. EPIT therapy response is associated with significant enrichment in pathways related to T-cell receptor signalling (P = 1.16 × 10-14), antigen presentation and costimulation, and cytokine signalling (P = 1.11 × 10-16), as well as upregulation of genes associated with regulatory T-cell function. CONCLUSIONS EoE is associated with distinct global transcriptional changes in CD4+ T cells, one feature of which is an IFN response signature. Clinically favorable response to EPIT is likely multifactorial but is associated with a distinct transcriptional profile in peripheral CD4+ cells supporting the hypothesis that EPIT alters peripheral CD4+ responses in EoE patients.
Collapse
Affiliation(s)
- Melanie A Ruffner
- Division of Allergy and ImmunologyThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsThe Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPAUSA
| | - Zhe Zhang
- Department of Biomedical and Health InformaticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Kelly Maurer
- Division of Allergy and ImmunologyThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Amanda B Muir
- Department of PediatricsThe Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPAUSA
- Division of Gastroenterology, Hepatology, and NutritionThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Antonella Cianferoni
- Division of Allergy and ImmunologyThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsThe Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPAUSA
| | - Kathleen E Sullivan
- Division of Allergy and ImmunologyThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsThe Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPAUSA
| | - Jonathan M Spergel
- Division of Allergy and ImmunologyThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsThe Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPAUSA
| |
Collapse
|
17
|
Critical Roles of Balanced T Helper 9 Cells and Regulatory T Cells in Allergic Airway Inflammation and Tumor Immunity. J Immunol Res 2021; 2021:8816055. [PMID: 33748292 PMCID: PMC7943311 DOI: 10.1155/2021/8816055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 01/02/2023] Open
Abstract
CD4+T helper (Th) cells are important mediators of immune responses in asthma and cancer. When counteracted by different classes of pathogens, naïve CD4+T cells undergo programmed differentiation into distinct types of Th cells. Th cells orchestrate antigen-specific immune responses upon their clonal T-cell receptor (TCR) interaction with the appropriate peptide antigen presented on MHC class II molecules expressed by antigen-presenting cells (APCs). T helper 9 (Th9) cells and regulatory T (Treg) cells and their corresponding cytokines have critical roles in tumor and allergic immunity. In the context of asthma and cancer, the dynamic internal microenvironment, along with chronic inflammatory stimuli, influences development, differentiation, and function of Th9 cells and Treg cells. Furthermore, the dysregulation of the balance between Th9 cells and Treg cells might trigger aberrant immune responses, resulting in development and exacerbation of asthma and cancer. In this review, the development, differentiation, and function of Th9 cells and Treg cells, which are synergistically regulated by various factors including cytokine signals, transcriptional factors (TFs), costimulatory signals, microenvironment cues, metabolic pathways, and different signal pathways, will be discussed. In addition, we focus on the recent progress that has helped to achieve a better understanding of the roles of Th9 cells and Treg cells in allergic airway inflammation and tumor immunity. We also discuss how various factors moderate their responses in asthma and cancer. Finally, we summarize the recent findings regarding potential mechanisms for regulating the balance between Th9 and Treg cells in asthma and cancer. These advances provide opportunities for novel therapeutic strategies that are aimed at reestablishing the balance of these cells in the diseases.
Collapse
|
18
|
Guignabert C, Humbert M. Targeting transforming growth factor-β receptors in pulmonary hypertension. Eur Respir J 2021; 57:13993003.02341-2020. [PMID: 32817256 DOI: 10.1183/13993003.02341-2020] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
Abstract
The transforming growth factor-β (TGF-β) superfamily includes several groups of multifunctional proteins that form two major branches, namely the TGF-β-activin-nodal branch and the bone morphogenetic protein (BMP)-growth differentiation factor (GDF) branch. The response to the activation of these two branches, acting through canonical (small mothers against decapentaplegic (Smad) 2/3 and Smad 1/5/8, respectively) and noncanonical signalling pathways, are diverse and vary for different environmental conditions and cell types. An extensive body of data gathered in recent years has demonstrated a central role for the cross-talk between these two branches in a number of cellular processes, which include the regulation of cell proliferation and differentiation, as well as the transduction of signalling cascades for the development and maintenance of different tissues and organs. Importantly, alterations in these pathways, which include heterozygous germline mutations and/or alterations in the expression of several constitutive members, have been identified in patients with familial/heritable pulmonary arterial hypertension (PAH) or idiopathic PAH (IPAH). Consequently, loss or dysfunction in the delicate, finely-tuned balance between the TGF-β-activin-nodal branch and the BMP-GDF branch are currently viewed as the major molecular defect playing a critical role in PAH predisposition and disease progression. Here we review the role of the TGF-β-activin-nodal branch in PAH and illustrate how this knowledge has not only provided insight into understanding its pathogenesis, but has also paved the way for possible novel therapeutic approaches.
Collapse
Affiliation(s)
- Christophe Guignabert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Marc Humbert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Dept of Respiratory and Intensive Care Medicine, French Pulmonary Hypertension Reference Center, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| |
Collapse
|
19
|
Wu B, Zhang S, Guo Z, Bi Y, Zhou M, Li P, Seyedsadr M, Xu X, Li JL, Markovic-Plese S, Wan YY. The TGF-β superfamily cytokine Activin-A is induced during autoimmune neuroinflammation and drives pathogenic Th17 cell differentiation. Immunity 2021; 54:308-323.e6. [PMID: 33421362 DOI: 10.1016/j.immuni.2020.12.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/12/2020] [Accepted: 12/16/2020] [Indexed: 01/02/2023]
Abstract
Th17 cells are known to exert pathogenic and non-pathogenic functions. Although the cytokine transforming growth factor β1 (TGF-β1) is instrumental for Th17 cell differentiation, it is dispensable for generation of pathogenic Th17 cells. Here, we examined the T cell-intrinsic role of Activin-A, a TGF-β superfamily member closely related to TGF-β1, in pathogenic Th17 cell differentiation. Activin-A expression was increased in individuals with relapsing-remitting multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. Stimulation with interleukin-6 and Activin-A induced a molecular program that mirrored that of pathogenic Th17 cells and was inhibited by blocking Activin-A signaling. Genetic disruption of Activin-A and its receptor ALK4 in T cells impaired pathogenic Th17 cell differentiation in vitro and in vivo. Mechanistically, extracellular-signal-regulated kinase (ERK) phosphorylation, which was essential for pathogenic Th17 cell differentiation, was suppressed by TGF-β1-ALK5 but not Activin-A-ALK4 signaling. Thus, Activin-A drives pathogenic Th17 cell differentiation, implicating the Activin-A-ALK4-ERK axis as a therapeutic target for Th17 cell-related diseases.
Collapse
Affiliation(s)
- Bing Wu
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Song Zhang
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zengli Guo
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanmin Bi
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mingxia Zhou
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ping Li
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Xiaojiang Xu
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Silva Markovic-Plese
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
De Martino M, Daviaud C, Diamond JM, Kraynak J, Alard A, Formenti SC, Miller LD, Demaria S, Vanpouille-Box C. Activin A Promotes Regulatory T-cell-Mediated Immunosuppression in Irradiated Breast Cancer. Cancer Immunol Res 2020; 9:89-102. [PMID: 33093219 DOI: 10.1158/2326-6066.cir-19-0305] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/01/2020] [Accepted: 10/14/2020] [Indexed: 11/16/2022]
Abstract
Increased regulatory T cells (Treg) after radiotherapy have been reported, but the mechanisms of their induction remain incompletely understood. TGFβ is known to foster Treg differentiation within tumors and is activated following radiotherapy. Thus, we hypothesized that TGFβ blockade would result in decreased Tregs within the irradiated tumor microenvironment. We found increased Tregs in the tumors of mice treated with focal radiotherapy and TGFβ blockade. This increase was mediated by upregulation of another TGFβ family member, activin A. In vitro, activin A secretion was increased following irradiation of mouse and human breast cancer cells, and its expression was further enhanced upon TGFβ blockade. In vivo, dual blockade of activin A and TGFβ was required to decrease intratumoral Tregs in the context of radiotherapy. This resulted in an increase in CD8+ T-cell priming and was associated with a reduced tumor recurrence rate. Combination of immune checkpoint inhibitors with the dual blockade of activin A and TGFβ led to the development of tumor-specific memory responses in irradiated breast cancer. Supporting the translational value of activin A targeting to reduce Treg-mediated immunosuppression, retrospective analysis of a public dataset of patients with breast cancer revealed a positive correlation between activin A gene expression and Treg abundance. Overall, these results shed light on an immune escape mechanism driven by activin A and suggest that dual targeting of activin A and TGFβ may be required to optimally unleash radiation-induced antitumor immunity against breast cancer.
Collapse
Affiliation(s)
- Mara De Martino
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Camille Daviaud
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Julie M Diamond
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York.,Department of Pathology, New York University School of Medicine, New York, New York.,Endless Frontier Labs, New York, New York
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Amandine Alard
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, New York, New York
| | - Lance D Miller
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, New York, New York.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York. .,Sandra and Edward Meyer Cancer Center, New York, New York
| |
Collapse
|
21
|
Wound Repair, Scar Formation, and Cancer: Converging on Activin. Trends Mol Med 2020; 26:1107-1117. [PMID: 32878730 DOI: 10.1016/j.molmed.2020.07.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Wound repair is a highly regulated process that requires the interaction of various cell types. It has been shown that cancers use the mechanisms of wound healing to promote their own growth. Therefore, it is of importance to identify common regulators of wound repair and tumor formation and to unravel their functions and mechanisms of action. An exciting example is activin, which acts on multiple cell types in wounds and tumors, thereby promoting healing, but also scar formation and tumorigenesis. Here, we summarize current knowledge on the role of activin in these processes and highlight the therapeutic potential of activin or activin antagonists for the treatment of impaired healing or excessive scarring and cancer, respectively.
Collapse
|
22
|
Activin-A limits Th17 pathogenicity and autoimmune neuroinflammation via CD39 and CD73 ectonucleotidases and Hif1-α-dependent pathways. Proc Natl Acad Sci U S A 2020; 117:12269-12280. [PMID: 32409602 DOI: 10.1073/pnas.1918196117] [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] [Indexed: 11/18/2022] Open
Abstract
In multiple sclerosis (MS), Th17 cells are critical drivers of autoimmune central nervous system (CNS) inflammation and demyelination. Th17 cells exhibit functional heterogeneity fostering both pathogenic and nonpathogenic, tissue-protective functions. Still, the factors that control Th17 pathogenicity remain incompletely defined. Here, using experimental autoimmune encephalomyelitis, an established mouse MS model, we report that therapeutic administration of activin-A ameliorates disease severity and alleviates CNS immunopathology and demyelination, associated with decreased activation of Th17 cells. In fact, activin-A signaling through activin-like kinase-4 receptor represses pathogenic transcriptional programs in Th17-polarized cells, while it enhances antiinflammatory gene modules. Whole-genome profiling and in vivo functional studies revealed that activation of the ATP-depleting CD39 and CD73 ectonucleotidases is essential for activin-A-induced suppression of the pathogenic signature and the encephalitogenic functions of Th17 cells. Mechanistically, the aryl hydrocarbon receptor, along with STAT3 and c-Maf, are recruited to promoter elements on Entpd1 and Nt5e (encoding CD39 and CD73, respectively) and other antiinflammatory genes, and control their expression in Th17 cells in response to activin-A. Notably, we show that activin-A negatively regulates the metabolic sensor, hypoxia-inducible factor-1α, and key inflammatory proteins linked to pathogenic Th17 cell states. Of translational relevance, we demonstrate that activin-A is induced in the CNS of individuals with MS and restrains human Th17 cell responses. These findings uncover activin-A as a critical controller of Th17 cell pathogenicity that can be targeted for the suppression of autoimmune CNS inflammation.
Collapse
|
23
|
Nono JK, Lutz MB, Brehm K. Expansion of Host Regulatory T Cells by Secreted Products of the Tapeworm Echinococcus multilocularis. Front Immunol 2020; 11:798. [PMID: 32457746 PMCID: PMC7225322 DOI: 10.3389/fimmu.2020.00798] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/07/2020] [Indexed: 01/15/2023] Open
Abstract
Background Alveolar echinococcosis (AE), caused by the metacestode larval stage of the fox-tapeworm Echinococcus multilocularis, is a chronic zoonosis associated with significant modulation of the host immune response. A role of regulatory T-cells (Treg) in generating an immunosuppressive environment around the metacestode during chronic disease has been reported, but the molecular mechanisms of Treg induction by E. multilocularis, particularly parasite immunoregulatory factors involved, remain elusive so far. Methodology/Principal Findings We herein demonstrate that excretory/secretory (E/S) products of the E. multilocularis metacestode promote the formation of Foxp3+ Treg from CD4+ T-cells in vitro in a TGF-β-dependent manner, given that this effect was abrogated by treatment with antibody to mammalian TGF-β. We also show that host T-cells secrete elevated levels of the immunosuppressive cytokine IL-10 in response to metacestode E/S products. Within the E/S fraction of the metacestode we identified an E. multilocularis activin A homolog (EmACT) that displays significant similarities to mammalian Transforming Growth Factor-β (TGF-β/activin subfamily members. EmACT obtained from heterologous expression failed to directly induce Treg expansion from naïve T cells but required addition of recombinant host TGF-β to promote CD4+ Foxp3+ Treg conversion in vitro. Furthermore, like in the case of metacestode E/S products, EmACT-treated CD4+ T-cells secreted higher levels of IL-10. These observations suggest a contribution of EmACT to in vitro expansion of Foxp3+ Treg by the E. multilocularis metacestode. Using infection experiments we show that intraperitoneally injected metacestode tissue expands host Foxp3+ Treg, confirming the expansion of this cell type in vivo during parasite establishment. Conclusion/Significance In conclusion, we herein demonstrate that E. multilocularis larvae secrete factors that induce the secretion of IL-10 by T-cells and contribute to the expansion of TGF-b-driven Foxp3+ Treg, a cell type that has been reported crucial for generating a tolerogenic environment to support parasite establishment and proliferation. Among the E/S factors of the parasite we identified a factor with structural and functional homologies to mammalian activin A which might play an important role in these activities.
Collapse
Affiliation(s)
- Justin Komguep Nono
- Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
- Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
- The Medical Research Centre, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaounde, Cameroon
| | - Manfred B. Lutz
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Klaus Brehm
- Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Kariyawasam HH, Gane SB. Allergen-induced asthma, chronic rhinosinusitis and transforming growth factor-β superfamily signaling: mechanisms and functional consequences. Expert Rev Clin Immunol 2019; 15:1155-1170. [PMID: 31549888 DOI: 10.1080/1744666x.2020.1672538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Often co-associated, asthma and chronic rhinosinusitis (CRS) are complex heterogeneous disease syndromes. Severity in both is related to tissue inflammation and abnormal repair (termed remodeling). Understanding signaling factors that can modulate, integrate the activation, and regulation of such key processes together is increasingly important. The transforming growth factor (TGF)-β superfamily of ligands comprise a versatile system of immunomodulatory molecules that are gaining recognition as having an essential function in the immunopathogenesis of asthma. Early data suggest an important role in CRS as well. Abnormal or dysregulated signaling may contribute to disease pathogenesis and severity.Areas covered: The essential biology of this complex family of growth factors in relation to the excess inflammation and remodeling that occurs in allergic asthma and CRS is reviewed. The need to understand the integration of signaling pathways together is highlighted. Studies in human airway tissue are evaluated and only selected key animal models relevant to human disease discussed given the highly context-dependent signaling and function of these ligands.Expert opinion: Abnormal or dysregulated TGF-β superfamily signaling may be central to the excess inflammation and tissue remodeling in asthma, and possibly CRS. Therefore, the TGF-β superfamily signaling pathways represent an emerging and attractive therapeutic target.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Department of Adult Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| | - Simon B Gane
- Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| |
Collapse
|
25
|
Zessner-Spitzenberg J, Thomas AL, Krett NL, Jung B. TGFβ and activin A in the tumor microenvironment in colorectal cancer. GENE REPORTS 2019; 17. [PMID: 32154442 DOI: 10.1016/j.genrep.2019.100501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although overall survival in colorectal cancer (CRC) is increasing steadily due to progress in screening, therapeutic options and precise diagnostic tools remain scarce. As the understanding of CRC as a complex and multifactorial condition moves forward, the tumor microenvironment has come into focus as a source of diagnostic markers and potential therapeutic targets. The role of TGFβ in shifting the epithelial cancer compartment towards invasiveness and a pro-migratory phenotype via stromal signaling has been widely investigated. Accordingly, recent studies have proposed that CRC patients could be stratified into distinct subtypes and have identified one poor prognosis subset of CRC that is characterized by high stromal activity and elevated levels of TGFβ. The TGFβ superfamily member activin A is crucial for the pro-metastatic properties of the TGFβ pathway, yet it has been under-researched in CRC carcinogenesis. In this review, we will elucidate the signaling network and interdependency of both ligands in the context of the tumor microenvironment in CRC.
Collapse
Affiliation(s)
- Jasmin Zessner-Spitzenberg
- Division of Gastroenterology and Hepatology, University of Illinois Medical College, Chicago, IL 60612, USA.,Medical University of Vienna, Spitalgasse 23, 1090 Wien, Austria
| | - Alexandra L Thomas
- Division of Gastroenterology and Hepatology, University of Illinois Medical College, Chicago, IL 60612, USA
| | - Nancy L Krett
- Division of Gastroenterology and Hepatology, University of Illinois Medical College, Chicago, IL 60612, USA
| | - Barbara Jung
- Division of Gastroenterology and Hepatology, University of Illinois Medical College, Chicago, IL 60612, USA
| |
Collapse
|
26
|
Rautela J, Dagley LF, de Oliveira CC, Schuster IS, Hediyeh-Zadeh S, Delconte RB, Cursons J, Hennessy R, Hutchinson DS, Harrison C, Kita B, Vivier E, Webb AI, Degli-Esposti MA, Davis MJ, Huntington ND, Souza-Fonseca-Guimaraes F. Therapeutic blockade of activin-A improves NK cell function and antitumor immunity. Sci Signal 2019; 12:12/596/eaat7527. [PMID: 31455725 DOI: 10.1126/scisignal.aat7527] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Natural killer (NK) cells are innate lymphocytes that play a major role in immunosurveillance against tumor initiation and metastatic spread. The signals and checkpoints that regulate NK cell fitness and function in the tumor microenvironment are not well defined. Transforming growth factor-β (TGF-β) is a suppressor of NK cells that inhibits interleukin-15 (IL-15)-dependent signaling events and increases the abundance of receptors that promote tissue residency. Here, we showed that NK cells express the type I activin receptor ALK4, which, upon binding to its ligand activin-A, phosphorylated SMAD2/3 to suppress IL-15-mediated NK cell metabolism. Activin-A impaired human and mouse NK cell proliferation and reduced the production of granzyme B to impair tumor killing. Similar to TGF-β, activin-A also induced SMAD2/3 phosphorylation and stimulated NK cells to increase their cell surface expression of several markers of ILC1 cells. Activin-A also induced these changes in TGF-β receptor-deficient NK cells, suggesting that activin-A and TGF-β stimulate independent pathways that drive SMAD2/3-mediated NK cell suppression. Last, inhibition of activin-A by follistatin substantially slowed orthotopic melanoma growth in mice. These data highlight the relevance of examining TGF-β-independent SMAD2/3 signaling mechanisms as a therapeutic axis to relieve NK cell suppression and promote antitumor immunity.
Collapse
Affiliation(s)
- Jai Rautela
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Laura F Dagley
- Systems Biology and Personalized Medicine Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Carolina C de Oliveira
- Laboratório de Células Inflamatórias e Neoplásicas, Departamento de Biologia Celular, SCB, Centro Politecnico, Universidade Federal do Paraná, Curitiba, CEP 81531-980, PR, Brazil
| | - Iona S Schuster
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Crawley, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia.,Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Soroor Hediyeh-Zadeh
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology and Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Rebecca B Delconte
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joseph Cursons
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology and Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Robert Hennessy
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Dana S Hutchinson
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Craig Harrison
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Badia Kita
- Paranta Biosciences Limited, Melbourne, Victoria 3004, Australia
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, 13288 Marseille, France
| | - Andrew I Webb
- Systems Biology and Personalized Medicine Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Crawley, Western Australia, Australia.,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia.,Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Melissa J Davis
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology and Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Nicholas D Huntington
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia. .,University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Activin-A in the regulation of immunity in health and disease. J Autoimmun 2019; 104:102314. [PMID: 31416681 DOI: 10.1016/j.jaut.2019.102314] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 02/08/2023]
Abstract
The TGF-β superfamily of cytokines plays pivotal roles in the regulation of immune responses protecting against or contributing to diseases, such as, allergy, autoimmunity and cancer. Activin-A, a member of the TGF-β superfamily, was initially identified as an inducer of follicle-stimulating hormone secretion. Extensive research over the past decades illuminated fundamental roles for activin-A in essential biologic processes, including embryonic development, stem cell maintenance and differentiation, haematopoiesis, cell proliferation and tissue fibrosis. Activin-A signals through two type I and two type II receptors which, upon ligand binding, activate their kinase activity, phosphorylate the SMAD2 and 3 intracellular signaling mediators that form a complex with SMAD4, translocate to the nucleus and activate or silence gene expression. Most immune cell types, including macrophages, dendritic cells (DCs), T and B lymphocytes and natural killer cells have the capacity to produce and respond to activin-A, although not in a similar manner. In innate immune cells, including macrophages, DCs and neutrophils, activin-A exerts a broad range of pro- or anti-inflammatory functions depending on the cell maturation and activation status and the spatiotemporal context. Activin-A also controls the differentiation and effector functions of Th cell subsets, including Th9 cells, TFH cells, Tr1 Treg cells and Foxp3+ Treg cells. Moreover, activin-A affects B cell responses, enhancing mucosal IgA secretion and inhibiting pathogenic autoantibody production. Interestingly, an array of preclinical and clinical studies has highlighted crucial functions of activin-A in the initiation, propagation and resolution of human diseases, including autoimmune diseases, such as, systemic lupus erythematosus, rheumatoid arthritis and pulmonary alveolar proteinosis, in allergic disorders, including allergic asthma and atopic dermatitis, in cancer and in microbial infections. Here, we provide an overview of the biology of activin-A and its signaling pathways, summarize recent studies pertinent to the role of activin-A in the modulation of inflammation and immunity, and discuss the potential of targeting activin-A as a novel therapeutic approach for the control of inflammatory diseases.
Collapse
|
28
|
Lis A, Wiley M, Vaughan J, Gray PC, Blader IJ. The Activin Receptor, Activin-Like Kinase 4, Mediates Toxoplasma Gondii Activation of Hypoxia Inducible Factor-1. Front Cell Infect Microbiol 2019; 9:36. [PMID: 30891432 PMCID: PMC6411701 DOI: 10.3389/fcimb.2019.00036] [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: 09/14/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
To grow and cause disease, intracellular pathogens modulate host cell processes. Identifying these processes as well as the mechanisms used by the pathogens to manipulate them is important for the development of more effective therapeutics. As an example, the intracellular parasite Toxoplasma gondii induces a wide variety of changes to its host cell, including altered membrane trafficking, cytoskeletal reorganization, and differential gene expression. Although several parasite molecules and their host targets have been identified that mediate- these changes, few are known to be required for parasite replication. One exception is the host cell transcription factor, hypoxia-inducible factor-1 (HIF-1), which is required for parasite replication in an oxygen-dependent manner. Toxoplasma activates HIF-1 by stabilizing the HIF-1α subunit, and this is dependent on the signaling from the Activin-Like Kinase (ALK) receptor superfamily. Here, we demonstrate that specific overexpression of the ALK family member, ALK4, increased HIF-1 activity in Toxoplasma-infected cells, and this increase required ALK4 kinase activity. Moreover, Toxoplasma stimulated ALK4 to dimerize with its co-receptor, ActRII, and also increased ALK4 kinase activity, thereby demonstrating that Toxoplasma activates the ALK4 receptor. ALK4 activation of HIF-1 was independent of canonical SMAD signaling but rather was dependent on the non-canonical Rho GTPase and JNK MAP kinase signaling pathways. Finally, Toxoplasma increased rates of ALK4 ubiquitination and turnover. These data provide the first evidence indicating that ALK4 signaling is a target for a microbial pathogen to manipulate its host cell.
Collapse
Affiliation(s)
- Agnieszka Lis
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Mandi Wiley
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | | | | - Ira J Blader
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
29
|
Niu J, Wang Y, Liu B, Yao Y. Mesenchymal stem cells prolong the survival of orthotopic liver transplants by regulating the expression of TGF-β1. TURKISH JOURNAL OF GASTROENTEROLOGY 2019; 29:601-609. [PMID: 30260784 DOI: 10.5152/tjg.2018.17395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND/AIMS Recent studies have shown that transforming growth factor-β1 (TGF-β1) is prominently associated with acute rejection. This study aimed to explore the role of mesenchymal stem cells (MSCs) in the maintenance of the long-term survival of orthotopic liver transplants (OLTs) via the regulation of TGF-β1 in an experimental rat model. MATERIALS AND METHODS We used Lewis rats as donors and ACI rats as recipients. Hematoxylin and eosin staining was performed to evaluate histomorphological changes, and Western blot was performed to measure protein expression. RESULTS The expression of TGF-β1 in the liver allografts and spleen and protein levels of forkhead box P3 (FoxP3), interleukin-10 (IL-10), and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) were measured using Western blot. The suppressive capacity of CD4+CD25+ regulatory T cells was evaluated using the MTT assay. Cell-mediated immunotoxicity was evaluated using the mixed lymphocyte reaction of CD4+ T cells and cytotoxic T lymphocyte (CTL) assay of CD8+ T cells. The results showed that MSCs prolonged the survival of the OLT mice by regulating the expression of TGF-β1 at different time points. The administration of MSCs promoted a prolonged survival in the ACI recipients (105±6.6 d) compared with the MSC-untreated recipients (16.2±4.0 d). On the postoperative day (POD) 7, the MSC-treated recipients showed a significantly higher expression of TGF-β1, FoxP3, IL-10, and CTLA-4 than the MSC-untreated recipients. However, on POD 100, the MSC-treated recipients showed a lower expression of TGF-β1 and FOxP3 than that on POD 7. Moreover, on POD 7, CD4+CD25+ regulatory T cells extracted from the MSC-treated recipients showed a higher expression of FoxP3, IL-10, CTLA-4, and suppressive capacity. On POD 7, CD4+ T cells from the MSC-treated recipients showed more significantly diminished proliferative functions than the MSC-untreated recipients; further, a reduced allospecific CTL activity of CD8+ T cells was observed in the MSC-treated recipients. CONCLUSION MSCs may represent a promising cell therapeutic approach for inducing immunosuppression or transplant tolerance.
Collapse
Affiliation(s)
- Jian Niu
- Department of General Surgery, Xuzhou Medical College Hospital, Jiangsu, China
| | - Yue Wang
- Department of General Surgery, Xuzhou Medical College Hospital, Jiangsu, China
| | - Bin Liu
- Department of General Surgery, Xuzhou Medical College Hospital, Jiangsu, China
| | - Yuanhu Yao
- Department of General Surgery, Xuzhou Medical College Hospital, Jiangsu, China
| |
Collapse
|
30
|
TGFβ Superfamily Members as Regulators of B Cell Development and Function-Implications for Autoimmunity. Int J Mol Sci 2018; 19:ijms19123928. [PMID: 30544541 PMCID: PMC6321615 DOI: 10.3390/ijms19123928] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022] Open
Abstract
The TGFβ superfamily is composed of more than 33 growth and differentiation factors, including TGFβ1, β2, β3, BMPs, GDFs, nodal-related proteins, and activins. These members usually exert pleiotropic actions on several tissues and control multiple cellular processes, such as cell growth, cell survival, cell migration, cell fate specification, and differentiation, both during embryonic development and postnatal life. Although the effects of these factors on immune responses were elucidated long ago, most studies have been focused on the actions of TGFβs on T cells, as major regulators of adaptive immunity. In this review, we discuss new findings about the involvement of TGFβ superfamily members in the control of B cell development and function. Moreover, the potential contribution of TGFβ signaling to control B cell-mediated autoimmune diseases and its utility in the design of new therapies are also discussed.
Collapse
|
31
|
Lyons DO, Plewes MR, Pullen NA. Soluble transforming growth factor beta-1 enhances murine mast cell release of Interleukin 6 in IgE-independent and Interleukin 13 in IgE-dependent settings in vitro. PLoS One 2018; 13:e0207704. [PMID: 30444930 PMCID: PMC6239331 DOI: 10.1371/journal.pone.0207704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/03/2018] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION For immune cells transforming growth factor beta-1 (TGF-β1) can enhance or repress effector functions. Here, we characterize the effects of TGF-β1 on IgE-mediated and IL-33-mediated activation of primary murine mast cells derived from hematopoietic stem cells (bone marrow derived mast cells; BMMC). We also investigated potential interactions between TGF-β1 and stem cell factor (SCF). We conclude TGF-β1 plays a selectively stimulatory role for mast cell cultures in vitro. METHODS BMMCs from C57BL/6 mice were differentiated with IL-3 and then treated with TGF-β1. BMMCs were exposed to TGF-β1, primed with IgE, activated with antigen, and then IL-6 and IL-13 cytokine release was quantified using ELISA. Additionally, the effects of TGF-β1 on both IgE and IL-33-mediated short term activation were observed via flow cytometric analysis of both surface LAMP-1 expression and intracellular IL-6. Receptor colocalization was visualized using fluorescence confocal microscopy and individual receptor expression levels were also quantified. RESULTS Resting IL-6 production increased with TGF-β1 but significance was lost following BMMC activation via IgE receptor (FcεRI) crosslinking. This was similar to a comparison effect due to SCF treatment alone, which also enhanced resting levels of IL-6. TGF-β1 treatment enhanced release of IL-13 only with FcεRI-IgE-mediated activation. TGF-β1 suppressed mobilization of IL-6 with short-term BMMC activation when stimulated with IL-33. Lastly, colocalization patterns of the SCF receptor (CD117) and FcεRI with IgE crosslinking were unaffected by TGF-β1 treatment, but individual expression levels for FcεRI, CD117, and TGFβRII were all reduced following either IgE activation or TGF-β1 treatment; this reduction was partially recovered in BMMCs that were both activated by IgE and treated with TGF-β1. DISCUSSION These data reveal a novel positive effect of soluble TGF-β1 on mast cell activation in vitro, suggesting mast cells may be activated through a non-canonical pathway by TGF-β1. Understanding this interaction will provide insight into the potential role of mast cells in settings where TGF-β1 is produced in an aberrant manner, such as in and around high grade tumors.
Collapse
Affiliation(s)
- David O. Lyons
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| | - Michele R. Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Nicholas A. Pullen
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| |
Collapse
|
32
|
Liu HJ, Chen G, Chen L, Zhou M, Xiong XZ, Meng ZJ, Sun SW, Tao XN. Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract. Int J Mol Med 2018; 42:3404-3414. [PMID: 30320351 PMCID: PMC6202106 DOI: 10.3892/ijmm.2018.3919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/01/2018] [Indexed: 11/06/2022] Open
Abstract
In CD4+ T helper (Th) cells, transforming growth factor β (TGF‑β) is indispensable for the induction of both regulatory T (Treg) and interleukin‑17‑producing effector T helper (Th17) cells. Although BMP and activin membrane‑bound inhibitor (BAMBI) is part of a rheostat‑like mechanism for the regulation of TGF‑β signalling and autoimmune arthritis in mouse models, the underlying activity of BAMBI on the human Th17/Treg cell axis, particularly during exposure to cigarette smoke, remains to be elucidated. The present study aimed to further characterize BAMBI expression in human CD4+ cells, as well as immune imbalance during activation and cigarette smoke exposure. Results from the present study indicated that exposure to cigarette smoke extract partially suppressed Treg differentiation and promoted Th17 cell generation under stimulation by anti‑CD3/28 antibodies and TGF‑β1. Additionally, exposure to cigarette smoke induced an inhibition of phosphorylated‑Smad2/Smad3, which may have arisen from a concomitant enhancement of BAMBI expression. In conclusion, human BAMBI may function as a molecular switch to control TGF‑β signalling strength and the Th17/Treg cell balance, which may be used not only as a biomarker but also as a target of new treatment strategies for maintaining immune tolerance and for the treatment of smoking‑induced immune disorders.
Collapse
Affiliation(s)
- Hong-Ju Liu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Chen
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Long Chen
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Mei Zhou
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xian-Zhi Xiong
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhao-Ji Meng
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Sheng-Wen Sun
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiao-Nan Tao
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
33
|
Zhang S. The role of transforming growth factor β in T helper 17 differentiation. Immunology 2018; 155:24-35. [PMID: 29682722 PMCID: PMC6099164 DOI: 10.1111/imm.12938] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/11/2018] [Accepted: 04/11/2018] [Indexed: 12/12/2022] Open
Abstract
T helper 17 (Th17) cells play critical roles in inflammatory and autoimmune diseases. The lineage-specific transcription factor RORγt is the key regulator for Th17 cell fate commitment. A substantial number of studies have established the importance of transforming growth factor β (TGF-β) -dependent pathways in inducing RORγt expression and Th17 differentiation. TGF-β superfamily members TGF-β1 , TGF-β3 or activin A, in concert with interleukin-6 or interleukin-21, differentiate naive T cells into Th17 cells. Alternatively, Th17 differentiation can occur through TGF-β-independent pathways. However, the mechanism of how TGF-β-dependent and TGF-β-independent pathways control Th17 differentiation remains controversial. This review focuses on the perplexing role of TGF-β in Th17 differentiation, depicts the requirement of TGF-β for Th17 development, and underscores the multiple mechanisms underlying TGF-β-promoted Th17 generation, pathogenicity and plasticity. With new insights and comprehension from recent findings, this review specifically tackles the involvement of the canonical TGF-β signalling components, SMAD2, SMAD3 and SMAD4, summarizes diverse SMAD-independent mechanisms, and highlights the importance of TGF-β signalling in balancing the reciprocal conversion of Th17 and regulatory T cells. Finally, this review includes discussions and perspectives and raises important mechanistic questions about the role of TGF-β in Th17 generation and function.
Collapse
Affiliation(s)
- Song Zhang
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
34
|
Browning LM, Pietrzak M, Kuczma M, Simms CP, Kurczewska A, Refugia JM, Lowery DJ, Rempala G, Gutkin D, Ignatowicz L, Muranski P, Kraj P. TGF-β-mediated enhancement of T H17 cell generation is inhibited by bone morphogenetic protein receptor 1α signaling. Sci Signal 2018; 11:eaar2125. [PMID: 30154100 PMCID: PMC8713300 DOI: 10.1126/scisignal.aar2125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The cytokines of the transforming growth factor-β (TGF-β) family promote the growth and differentiation of multiple tissues, but the role of only the founding member, TGF-β, in regulating the immune responses has been extensively studied. TGF-β is critical to prevent the spontaneous activation of self-reactive T cells and sustain immune homeostasis. In contrast, in the presence of proinflammatory cytokines, TGF-β promotes the differentiation of effector T helper 17 (TH17) cells. Abrogating TGF-β receptor signaling prevents the development of interleukin-17 (IL-17)-secreting cells and protects mice from TH17 cell-mediated autoimmunity. We found that the receptor of another member of TGF-β family, bone morphogenetic protein receptor 1α (BMPR1α), regulates T helper cell activation. We found that the differentiation of TH17 cells from naive CD4+ T cells was inhibited in the presence of BMPs. Abrogation of BMPR1α signaling during CD4+ T cell activation induced a developmental program that led to the generation of inflammatory effector cells expressing large amounts of IL-17, IFN-γ, and TNF family cytokines and transcription factors defining the TH17 cell lineage. We found that TGF-β and BMPs cooperated to establish effector cell functions and the cytokine profile of activated CD4+ T cells. Together, our data provide insight into the immunoregulatory function of BMPs.
Collapse
Affiliation(s)
- Lauren M Browning
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Michal Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Colin P Simms
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Agnieszka Kurczewska
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Justin M Refugia
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Dustin J Lowery
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Grzegorz Rempala
- College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Dmitriy Gutkin
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15240, USA
| | - Leszek Ignatowicz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Pawel Muranski
- Columbia University Medical Center, New York, NY 10032, USA
| | - Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
35
|
Komai T, Okamura T, Inoue M, Yamamoto K, Fujio K. Reevaluation of Pluripotent Cytokine TGF-β3 in Immunity. Int J Mol Sci 2018; 19:ijms19082261. [PMID: 30071700 PMCID: PMC6121403 DOI: 10.3390/ijms19082261] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/28/2018] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor (TGF)-βs are pluripotent cytokines with stimulatory and inhibitory properties for multiple types of immune cells. Analyses of genetic knockouts of each isoform of TGF-β have revealed differing expression patterns and distinct roles for the three mammalian isoforms of TGF-β. Considerable effort has been focused on understanding the molecular mechanisms of TGF-β1-mediated immune regulation, given its pivotal role in prohibiting systemic autoimmune disease. In recent years, functional similarities and differences between the TGF-β isoforms have delineated their distinct roles in the development of immunopathology and immune tolerance, with increased recent attention being focused on TGF-β3. In addition to the characteristic properties of each TGF-β isoform, recent progress has identified determinants of context-dependent functionality, including various cellular targets, cytokine concentrations, tissue microenvironments, and cytokine synergy, which combine to shape the physiological and pathophysiological roles of the TGF-βs in immunity. Controlling TGF-β production and signaling is being tested as a novel therapeutic strategy in multiple clinical trials for several human diseases. This review highlights advances in the understanding of the cellular sources, activation processes, contextual determinants, and immunological roles of TGF-β3 with comparisons to other TGF-β isoforms.
Collapse
Affiliation(s)
- Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
- Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo 153-8505, Japan.
| | - Mariko Inoue
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
- Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo 153-8505, Japan.
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Kanagawa 230-0045, Japan.
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| |
Collapse
|
36
|
Ni X, Tao J, Barbi J, Chen Q, Park BV, Li Z, Zhang N, Lebid A, Ramaswamy A, Wei P, Zheng Y, Zhang X, Wu X, Vignali P, Yang CP, Li H, Pardoll D, Lu L, Pan D, Pan F. YAP Is Essential for Treg-Mediated Suppression of Antitumor Immunity. Cancer Discov 2018; 8:1026-1043. [PMID: 29907586 DOI: 10.1158/2159-8290.cd-17-1124] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/05/2018] [Accepted: 06/06/2018] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) are critical for maintaining self-tolerance and immune homeostasis, but their suppressive function can impede effective antitumor immune responses. FOXP3 is a transcription factor expressed in Tregs that is required for their function. However, the pathways and microenvironmental cues governing FOXP3 expression and Treg function are not completely understood. Herein, we report that YAP, a coactivator of the Hippo pathway, is highly expressed in Tregs and bolsters FOXP3 expression and Treg function in vitro and in vivo. This potentiation stemmed from YAP-dependent upregulation of activin signaling, which amplifies TGFβ/SMAD activation in Tregs. YAP deficiency resulted in dysfunctional Tregs unable to suppress antitumor immunity or promote tumor growth in mice. Chemical YAP antagonism and knockout or blockade of the YAP-regulated activin receptor similarly improved antitumor immunity. Thus, we identify YAP as an unexpected amplifier of a Treg-reinforcing pathway with significant potential as an anticancer immunotherapeutic target.Significance: Tregs suppress antitumor immunity, and pathways supporting their function can be novel immunotherapy targets. Here, the selective expression of YAP by Tregs, its importance for their function, and its unexpected enhancement of pro-Treg Activin/SMAD signaling are reported, as are validations of potential cancer-fighting antagonists of YAP and its regulatory targets. Cancer Discov; 8(8); 1026-43. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.
Collapse
Affiliation(s)
- Xuhao Ni
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Translational Medicine Research Center, Affiliated Jiangning Hospital, and Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jinhui Tao
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Rheumatology & Immunology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Joseph Barbi
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Qian Chen
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Thorgene Co., Ltd., Beijing, China
| | - Benjamin V Park
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhiguang Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Nailing Zhang
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andriana Lebid
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anjali Ramaswamy
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ping Wei
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ying Zheng
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xuehong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xingmei Wu
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Paolo Vignali
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cui-Ping Yang
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Gastroenterology, Rujin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huabin Li
- Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Drew Pardoll
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ling Lu
- Translational Medicine Research Center, Affiliated Jiangning Hospital, and Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Duojia Pan
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Fan Pan
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
37
|
Decorin-Modified Umbilical Cord Mesenchymal Stem Cells (MSCs) Attenuate Radiation-Induced Lung Injuries via Regulating Inflammation, Fibrotic Factors, and Immune Responses. Int J Radiat Oncol Biol Phys 2018; 101:945-956. [PMID: 29976507 DOI: 10.1016/j.ijrobp.2018.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 03/01/2018] [Accepted: 04/03/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the therapeutic effects of decorin (DCN)-modified mesenchymal stem cells (MSCs) on radiation-induced lung injuries (RILIs) and to clarify the underlying mechanisms. METHODS AND MATERIALS Umbilical cord-derived mesenchymal stem cells (MSCs) were modified with Ad(E1-).DCN to generate DCN-expressing MSCs (DCN-modified MSCs [MSCs.DCN]). In an experimental mouse model of RILI, MSCs.DCN and MSCs.Null [MSCs modified with Ad(E1-).Null] were intravenously engrafted at 6 hours or 28 days after irradiation. The therapeutic effects on lung inflammation and fibrosis were evaluated by histopathologic analysis at 28 days and 3 months after irradiation. Inflammatory cytokines and chemokines were analyzed in both sera and lung tissues, and subtypes of T lymphocytes including regulatory T cells (Tregs) were analyzed in the peripheral blood and spleen. RESULTS Both MSC treatments could alleviate histopathologic injuries by reducing lymphocyte infiltration, decreasing apoptosis, increasing proliferation of epithelial cells, and inhibiting fibrosis in the later phase. However, treatment with MSCs.DCN resulted in much more impressive therapeutic effects. Moreover, we discovered that MSC treatment reduced the expression of chemokines and inflammatory cytokines and increased the expression of anti-inflammatory cytokines in both the peripheral blood and local pulmonary tissues. An important finding was that MSCs.DCN were much more effective in inducing interferon-γ expression, inhibiting collagen type III α1 expression in pulmonary tissues, and decreasing the proportion of Tregs. Furthermore, our data suggested that treatment during the acute phase (6 hours) after irradiation evoked much stronger responses both in attenuating inflammation and in inhibiting fibrosis than in the later phase (28 days). CONCLUSIONS MSCs.DCN could attenuate acute inflammation after irradiation and significantly inhibit later fibrosis. Likewise, DCN enhanced the functions of MSCs by targeting profibrotic factors and Tregs.
Collapse
|
38
|
Yin C, Cai XB, Wang HJ, Gu BJ, Yang XF, Zhang R, Ji XH. Pathological significance and regulatory mechanism of lymphotoxin β receptor overexpression in T cells of patients with systemic lupus erythematosus. J Biomed Res 2018; 32:113-122. [PMID: 28963441 PMCID: PMC5895565 DOI: 10.7555/jbr.27.20130046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease. Lymphotoxin β receptor (LTβR) signaling plays an important role in autoimmune inflammations. LTβR-Ig fusion protein, LTβR blocking agent, has been used to treat SLE, while its mechanism remains to be fully elucidated. In this study, to investigate the expression of LTβR in the T cells of SLE patients and its roles in the pathogenesis of SLE, we isolated the peripheral blood T cells of SLE patients and normal controls to detect expression of LTβR by flow cytometry and RNA assay. T cells were also stimulated with LIGHT, a ligand of LTβR, and then detected for their LTβR expressions and apoptosis by flow cytometry. Also, their expressions of inflammatory factors and receptors were determined by RNA assay. The results showed that LTβR positive cells were 22.75%±6.98% in CD3+ cells of SLE patients, while there were almost no LTβR positive cells in CD3+ cells of normal persons. Moreover, LTβR expression was remarkably higher in CD3, CD4 and CD8 positive T cells of active SLE patients than non/low active patients (all P<0.05), and positively correlated with increased Ig level, decreased complement level and renal damage. Moreover, the stimulation of SLE T cells with LIGHT promoted higher expression of LTβR, IL-23R and IL-17A, and apoptosis of T cells. In conclusion, we demonstrated a high expression of LTβR in the T cells of SLE patients which may be associated with pathogenesis of SLE.
Collapse
Affiliation(s)
- Cheng Yin
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Red Cross Blood Center, Nanjing, Jiangsu 210003, China
| | - Xu-Bing Cai
- Red Cross Blood Center, Nanjing, Jiangsu 210003, China
| | - Hui-Juan Wang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bing-Jie Gu
- Rheumatology Department of Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Xiao-Fan Yang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Rong Zhang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Hui Ji
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
39
|
Haertel E, Joshi N, Hiebert P, Kopf M, Werner S. Regulatory T cells are required for normal and activin‐promoted wound repair in mice. Eur J Immunol 2018; 48:1001-1013. [DOI: 10.1002/eji.201747395] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/23/2018] [Accepted: 02/12/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Eric Haertel
- Department of BiologyInstitute of Molecular Health Sciences ETH Zurich Switzerland
| | - Natasha Joshi
- Department of BiologyInstitute of Molecular Health Sciences ETH Zurich Switzerland
| | - Paul Hiebert
- Department of BiologyInstitute of Molecular Health Sciences ETH Zurich Switzerland
| | - Manfred Kopf
- Department of BiologyInstitute of Molecular Health Sciences ETH Zurich Switzerland
| | - Sabine Werner
- Department of BiologyInstitute of Molecular Health Sciences ETH Zurich Switzerland
| |
Collapse
|
40
|
Li X, Li D, Huang X, Zhou P, Shi Q, Zhang B, Ju X. Helios expression in regulatory T cells promotes immunosuppression, angiogenesis and the growth of leukemia cells in pediatric acute lymphoblastic leukemia. Leuk Res 2018; 67:60-66. [PMID: 29455107 DOI: 10.1016/j.leukres.2018.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/23/2018] [Accepted: 02/12/2018] [Indexed: 01/09/2023]
Abstract
Regulatory T cells (Tregs) characterized by the transcription factor forkhead box P3 (FoxP3) are crucial for maintaining immune tolerance and preventing autoimmunity. However, FoxP3 does not function alone and Helios is considered a potential candidate for defining Treg subsets. In this study, we investigated the expression and function of Helios for identifying Tregs in childhood precursor B-cell acute lymphoblastic leukemia (pre-B ALL). Our results demonstrated that patients with pre-B ALL had a higher percentage of Helios+ FoxP3+ CD4+ Tregs. And there was a positive correlation between the expression of Helios and the suppressive function of Tregs, the risk gradation of ALL. Helios in combination with CD4 and FoxP3 may be an effective way to detect functional Tregs in pre-B ALL by promoting the secretion of transforming growth factor (TGF)-β1. Furthermore, Helios+ Tregs could regulate angiogenesis in the BM niche of pre-B ALL via the VEGFA/VEGFR2 pathway. We also found Helios+ Tregs decreased apoptosis rate of nalm-6 cells by up-regulating the expression of anti-apoptosis protein Bcl-2. In summary, these data strongly imply the physiological importance of Helios expression in Tregs, and suggest that the manipulation of Helios may serve as a novel strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Xue Li
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Dong Li
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Xiaoyang Huang
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Panpan Zhou
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Qing Shi
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Bing Zhang
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital, Shandong University, Ji'nan, Shandong 250012, China.
| |
Collapse
|
41
|
Luz-Crawford P, Espinosa-Carrasco G, Ipseiz N, Contreras R, Tejedor G, Medina DA, Vega-Letter AM, Ngo D, Morand EF, Pène J, Hernandez J, Jorgensen C, Djouad F. Gilz-Activin A as a Novel Signaling Axis Orchestrating Mesenchymal Stem Cell and Th17 Cell Interplay. Am J Cancer Res 2018; 8:846-859. [PMID: 29344311 PMCID: PMC5771098 DOI: 10.7150/thno.21793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/09/2017] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSC) are highly immunosuppressive cells able to reduce chronic inflammation through the active release of mediators. Recently, we showed that glucocorticoid-induced leucine zipper (Gilz) expression by MSC is involved in their therapeutic effect by promoting the generation of regulatory T cells. However, the mechanisms underlying this pivotal role of Gilz remain elusive. Methods and Results In this study, we have uncovered evidence that Gilz modulates the phenotype and function of Th1 and Th17 cells likely by upregulating the level of Activin A and NO2 secreted by MSC. Adoptive transfer experiments sustained this Gilz-dependent suppressive effect of MSC on Th1 and Th17 cell functions. In immunoregulatory MSC, obtained by priming with IFN-γ and TNF-α, Gilz was translocated to the nucleus and bound to the promoters of inos and Activin βA to induce their expression. The increased expression of Activin A directly impacted on Th17 cells fate by repressing their differentiation program through the activation of Smad3/2 and enhancing IL-10 production. Conclusion Our results reveal how Gilz controls inos and Activin βA gene expression to ultimately assign immunoregulatory status to MSC able to repress the pathogenic Th17 cell differentiation program and uncover Activin A as a novel mediator of MSC in this process.
Collapse
|
42
|
Ortega-Francisco S, de la Fuente-Granada M, Alvarez Salazar EK, Bolaños-Castro LA, Fonseca-Camarillo G, Olguin-Alor R, Alemán-Muench GR, López-Casillas F, Raman C, García-Zepeda EA, Soldevila G. TβRIII is induced by TCR signaling and downregulated in FoxP3+ regulatory T cells. Biochem Biophys Res Commun 2017; 494:82-87. [DOI: 10.1016/j.bbrc.2017.10.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/16/2017] [Indexed: 12/12/2022]
|
43
|
Arora S, Dev K, Agarwal B, Das P, Syed MA. Macrophages: Their role, activation and polarization in pulmonary diseases. Immunobiology 2017; 223:383-396. [PMID: 29146235 PMCID: PMC7114886 DOI: 10.1016/j.imbio.2017.11.001] [Citation(s) in RCA: 381] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 02/08/2023]
Abstract
Macrophages, circulating in the blood or concatenated into different organs and tissues constitute the first barrier against any disease. They are foremost controllers of both innate and acquired immunity, healthy tissue homeostasis, vasculogenesis and congenital metabolism. Two hallmarks of macrophages are diversity and plasticity due to which they acquire a wobbling array of phenotypes. These phenotypes are appropriately synchronized responses to a variety of different stimuli from either the tissue microenvironment or - microbes or their products. Based on the phenotype, macrophages are classified into classically activated/(M1) and alternatively activated/(M2) which are further sub-categorized into M2a, M2b, M2c and M2d based upon gene expression profiles. Macrophage phenotype metamorphosis is the regulating factor in initiation, progression, and termination of numerous inflammatory diseases. Several transcriptional factors and other factors controlling gene expression such as miRNAs contribute to the transformation of macrophages at different points in different diseases. Understanding the mechanisms of macrophage polarization and modulation of their phenotypes to adjust to the micro environmental conditions might provide us a great prospective for designing novel therapeutic strategy. In view of the above, this review summarises the activation of macrophages, the factors intricated in activation along with benefaction of macrophage polarization in response to microbial infections, pulmonary toxicity, lung injury and other inflammatory diseases such as chronic obstructive pulmonary dysplasia (COPD), bronchopulmonary dysplasia (BPD), asthma and sepsis, along with the existing efforts to develop therapies targeting this facet of macrophage biology.
Collapse
Affiliation(s)
- Shweta Arora
- Translational Research Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Kapil Dev
- Translational Research Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Beamon Agarwal
- Department of Hematopathology, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467-2401, United States.
| | - Pragnya Das
- Drexel University College of Medicine, Philadelphia, PA 19134, United States.
| | - Mansoor Ali Syed
- Translational Research Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
44
|
Pickup MW, Owens P, Moses HL. TGF-β, Bone Morphogenetic Protein, and Activin Signaling and the Tumor Microenvironment. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022285. [PMID: 28062564 DOI: 10.1101/cshperspect.a022285] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cellular and noncellular components surrounding the tumor cells influence many aspects of tumor progression. Transforming growth factor β (TGF-β), bone morphogenetic proteins (BMPs), and activins have been shown to regulate the phenotype and functions of the microenvironment and are attractive targets to attenuate protumorigenic microenvironmental changes. Given the pleiotropic nature of the cytokines involved, a full understanding of their effects on numerous cell types in many contexts is necessary for proper clinical intervention. In this review, we will explore the various effects of TGF-β, BMP, and activin signaling on stromal phenotypes known to associate with cancer progression. We will summarize these findings in the context of their tumor suppressive or promoting effects, as well as the molecular changes that these cytokines induce to influence stromal phenotypes.
Collapse
Affiliation(s)
- Michael W Pickup
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Philip Owens
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Harold L Moses
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| |
Collapse
|
45
|
Activin-A co-opts IRF4 and AhR signaling to induce human regulatory T cells that restrain asthmatic responses. Proc Natl Acad Sci U S A 2017; 114:E2891-E2900. [PMID: 28320933 DOI: 10.1073/pnas.1616942114] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Type 1 regulatory T (Tr1) cells play a pivotal role in restraining human T-cell responses toward environmental allergens and protecting against allergic diseases. Still, the precise molecular cues that underlie their transcriptional and functional specification remain elusive. Here, we show that the cytokine activin-A instructs the generation of CD4+ T cells that express the Tr1-cell-associated molecules IL-10, inducible T-Cell costimulator (ICOS), lymphocyte activation gene 3 protein (LAG-3), and CD49b, and exert strongly suppressive functions toward allergic responses induced by naive and in vivo-primed human T helper 2 cells. Moreover, mechanistic studies reveal that activin-A signaling induces the activation of the transcription factor interferon regulatory factor (IRF4), which, along with the environmental sensor aryl hydrocarbon receptor, forms a multipartite transcriptional complex that binds in IL-10 and ICOS promoter elements and controls gene expression in human CD4+ T cells. In fact, IRF4 silencing abrogates activin-A-driven IL10 and ICOS up-regulation and impairs the suppressive functions of human activin-A-induced Tr1-like (act-A-iTr1) cells. Importantly, using a humanized mouse model of allergic asthma, we demonstrate that adoptive transfer of human act-A-iTr1 cells, both in preventive and therapeutic protocols, confers significant protection against cardinal asthma manifestations, including pulmonary inflammation. Overall, our findings uncover an activin-A-induced IRF4-aryl hydrocarbon receptor (AhR)-dependent transcriptional network, which generates suppressive human Tr1 cells that may be harnessed for the control of allergic diseases.
Collapse
|
46
|
Valencia J, Jiménez E, Martínez VG, Del Amo BG, Hidalgo L, Entrena A, Fernández-Sevilla LM, Del Río F, Varas A, Vicente Á, Sacedón R. Characterization of human fibroblastic reticular cells as potential immunotherapeutic tools. Cytotherapy 2017; 19:640-653. [PMID: 28262465 DOI: 10.1016/j.jcyt.2017.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 01/09/2023]
Abstract
Fibroblastic reticular cells (FRCs) are essential players during adaptive immune responses not only as a structural support for the encounter of antigen-presenting cells and naive T lymphocytes but also as a source of modulatory signals. However, little is known about this cell population in humans. To address the phenotypical and functional analysis of human FRCs here we established splenic (SP) and mesenteric lymph node (LN) CD45-CD31-CD90+podoplanin+ myofibroblastic cell cultures. They shared the phenotypical characteristics distinctive of FRCs, including the expression of immunomodulatory factors and peripheral tissue antigens. Nevertheless, human FRCs also showed particular features, some differing from mouse FRCs, like the lack of nitric oxide synthase (NOS2) expression after interferon (IFN)γstimulation. Interestingly, SP-FRCs expressed higher levels of interleukin (IL)-6, BMP4, CCL2, CXCL12 and Notch molecules, and strongly adapted their functional profile to lipopolysaccharide (LPS), polyinosinic:polycytidylic acid (Poly I:C) and IFNγ stimulation. In contrast, we found higher expression of transforming growth factor (TGF)β and Activin A in LN-FRCs that barely responded via Toll-Like Receptor (TLR)3 and constitutively expressed retinaldehyde dehydrogenase 1 enzyme, absent in SP-FRCs. This study reveals human FRCs can be valuable models to increase our knowledge about the physiology of human secondary lymphoid organs in health and disease and to explore the therapeutic options of FRCs.
Collapse
Affiliation(s)
- Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Eva Jiménez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Víctor G Martínez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Beatriz G Del Amo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Laura Hidalgo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Ana Entrena
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | - Francisco Del Río
- Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Alberto Varas
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Ángeles Vicente
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Rosa Sacedón
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
47
|
Brehm K, Koziol U. Echinococcus-Host Interactions at Cellular and Molecular Levels. ADVANCES IN PARASITOLOGY 2017; 95:147-212. [PMID: 28131363 DOI: 10.1016/bs.apar.2016.09.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The potentially lethal zoonotic diseases alveolar and cystic echinococcosis are caused by the metacestode larval stages of the tapeworms Echinococcus multilocularis and Echinococcus granulosus, respectively. In both cases, metacestode growth and proliferation occurs within the inner organs of mammalian hosts, which is associated with complex molecular host-parasite interactions that regulate nutrient uptake by the parasite as well as metacestode persistence and development. Using in vitro cultivation systems for parasite larvae, and informed by recently released, comprehensive genome and transcriptome data for both parasites, these molecular host-parasite interactions have been subject to significant research during recent years. In this review, we discuss progress in this field, with emphasis on parasite development and proliferation. We review host-parasite interaction mechanisms that occur early during an infection, when the invading oncosphere stage undergoes a metamorphosis towards the metacestode, and outline the decisive role of parasite stem cells during this process. We also discuss special features of metacestode morphology, and how this parasite stage takes up nutrients from the host, utilizing newly evolved or expanded gene families. We comprehensively review mechanisms of host-parasite cross-communication via evolutionarily conserved signalling systems and how the parasite signalling systems might be exploited for the development of novel chemotherapeutics. Finally, we point to an urgent need for the development of functional genomic techniques in this parasite, which will be imperative for hypothesis-driven analyses into Echinococcus stem cell biology, developmental mechanisms and immunomodulatory activities, which are all highly relevant for the development of anti-infective measures.
Collapse
Affiliation(s)
- K Brehm
- University of Würzburg, Würzburg, Germany
| | - U Koziol
- University of Würzburg, Würzburg, Germany; Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
48
|
|
49
|
Lee N, Shin JU, Jin S, Yun KN, Kim JY, Park CO, Kim SH, Noh JY, Lee KH. Upregulation of CD47 in Regulatory T Cells in Atopic Dermatitis. Yonsei Med J 2016; 57:1435-45. [PMID: 27593872 PMCID: PMC5011276 DOI: 10.3349/ymj.2016.57.6.1435] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Regulatory T (Treg) cells are key modulators in the immune system. Recent studies have shown that atopic dermatitis (AD) patients have higher numbers of Treg cells; however, little is known about the specific phenotype and function of Treg cells in AD. MATERIALS AND METHODS To identify differentially expressed proteins in peripheral induced Treg cells in AD and naturally derived Treg cells in normal controls, CD4⁺CD25⁺ Treg cells were isolated from thymus tissue of normal mice and the spleens of AD mice. Membrane proteins were extracted, and quantitative proteomics labeling with Tandem Mass Tags (TMT) was performed, followed by one-dimensional liquid chromatography/tandem mass spectrometry analysis. RESULTS Using TMT labeling, we identified 510 proteins, including 63 membrane proteins and 16 plasma membrane proteins. CD47 was one of the upregulated proteins in Treg cells in AD spleens. Although CD47 was expressed in all CD4⁺ and CD8⁺ T cells, a significantly higher expression of CD47 was observed in the Treg cells of AD mice and AD patients than in those of normal mice and healthy controls. Furthermore, Treg cells from the spleen showed a significantly higher expression of CD47 than those from the thymus. CONCLUSION We found that CD47 is highly expressed in the Treg cells of AD mice, particularly in the spleen. Based on our results, we propose that CD47(high) Treg cells are likely induced Treg cells and that upregulated CD47 in the Treg cells of AD patients may play a role in the increased population of Treg cells in AD.
Collapse
Affiliation(s)
- Nara Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jung U Shin
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Shan Jin
- Department of Dermatology, Yanbian University Hospital, Yanji, Jilin, China
| | - Ki Na Yun
- Biomedical Group, Korea Basic Science Institute, Cheongju, Korea
| | - Jin Young Kim
- Biomedical Group, Korea Basic Science Institute, Cheongju, Korea
| | - Chang Ook Park
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seo Hyeong Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Yeon Noh
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang Hoon Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
50
|
Activin A programs the differentiation of human TFH cells. Nat Immunol 2016; 17:976-84. [PMID: 27376469 PMCID: PMC4955732 DOI: 10.1038/ni.3494] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/24/2016] [Indexed: 12/22/2022]
Abstract
Follicular helper T (TFH) cells are CD4+ T cells specialized in helping B cells and are associated both with protective antibody responses and autoimmune diseases. The promise of targeting TFH cells therapeutically has been limited by fragmentary understanding of extrinsic signals regulating human TFH cell differentiation. A screen of a human protein library identified activin A as new regulator of TFH cell differentiation. Activin A orchestrated expression of multiple TFH-associated genes, independently or in concert with additional signals. TFH programming by activin A was antagonized by the cytokine IL-2. Activin A’s capacity to drive TFH cell differentiation in vitro was conserved for non-human primates but not mice. Finally, activin A-induced TFH programming was dependent on SMAD2 and SMAD3 signaling and blocked by pharmacological inhibitors.
Collapse
|