1
|
Pastwińska J, Karwaciak I, Karaś K, Bachorz RA, Ratajewski M. RORγT agonists as immune modulators in anticancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189021. [PMID: 37951483 DOI: 10.1016/j.bbcan.2023.189021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
RORγT is a transcription factor that directs the development of Th17 lymphocytes and other IL-17-expressing cells (e.g., Tc17 and ILC3 cells). These cells are involved in the body's defense against pathogenic bacteria and fungi, but they also participate in maintaining the proinflammatory environment in some autoimmune diseases and play a role in the immune system's response to cancer. Similar to other members of the nuclear receptor superfamily, the activity of RORγT is regulated by low-molecular-weight ligands. Therefore, extensive efforts have been dedicated to identifying inverse agonists that diminish the activity of this receptor and subsequently inhibit the development of autoimmune diseases. Unfortunately, in the pursuit of an ideal inverse agonist, the development of agonists has been overlooked. It is important to remember that these types of compounds, by stimulating lymphocytes expressing RORγT (Th17 and Tc17), can enhance the immune system's response to tumors. In this review, we present recent advancements in the biology of RORγT agonists and their potential application in anticancer therapy.
Collapse
Affiliation(s)
- Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Rafał A Bachorz
- Laboratory of Molecular Modeling, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland.
| |
Collapse
|
2
|
Picard FSR, Lutz V, Brichkina A, Neuhaus F, Ruckenbrod T, Hupfer A, Raifer H, Klein M, Bopp T, Pfefferle PI, Savai R, Prinz I, Waisman A, Moos S, Chang HD, Heinrich S, Bartsch DK, Buchholz M, Singh S, Tu M, Klein L, Bauer C, Liefke R, Burchert A, Chung HR, Mayer P, Gress TM, Lauth M, Gaida M, Huber M. IL-17A-producing CD8 + T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut 2023; 72:1510-1522. [PMID: 36759154 PMCID: PMC10359545 DOI: 10.1136/gutjnl-2022-327855] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 01/21/2023] [Indexed: 02/11/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant desmoplastic stroma composed of cancer-associated fibroblasts (CAF) and interspersed immune cells. A non-canonical CD8+ T-cell subpopulation producing IL-17A (Tc17) promotes autoimmunity and has been identified in tumours. Here, we evaluated the Tc17 role in PDAC. DESIGN Infiltration of Tc17 cells in PDAC tissue was correlated with patient overall survival and tumour stage. Wild-type (WT) or Il17ra-/- quiescent pancreatic stellate cells (qPSC) were exposed to conditional media obtained from Tc17 cells (Tc17-CM); moreover, co-culture of Tc17-CM-induced inflammatory (i)CAF (Tc17-iCAF) with tumour cells was performed. IL-17A/F-, IL-17RA-, RAG1-deficient and Foxn1nu/nu mice were used to study the Tc17 role in subcutaneous and orthotopic PDAC mouse models. RESULTS Increased abundance of Tc17 cells highly correlated with reduced survival and advanced tumour stage in PDAC. Tc17-CM induced iCAF differentiation as assessed by the expression of iCAF-associated genes via synergism of IL-17A and TNF. Accordingly, IL-17RA controlled the responsiveness of qPSC to Tc17-CM. Pancreatic tumour cells co-cultured with Tc17-iCAF displayed enhanced proliferation and increased expression of genes implicated in proliferation, metabolism and protection from apoptosis. Tc17-iCAF accelerated growth of mouse and human tumours in Rag1-/- and Foxn1nu/nu mice, respectively. Finally, Il17ra-expressed by fibroblasts was required for Tc17-driven tumour growth in vivo. CONCLUSIONS We identified Tc17 as a novel protumourigenic CD8+ T-cell subtype in PDAC, which accelerated tumour growth via IL-17RA-dependent stroma modification. We described a crosstalk between three cell types, Tc17, fibroblasts and tumour cells, promoting PDAC progression, which resulted in poor prognosis for patients.
Collapse
Affiliation(s)
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Anna Brichkina
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Felix Neuhaus
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Teresa Ruckenbrod
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Anna Hupfer
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Hartmann Raifer
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
- Core-Facility Flow Cytometry, Philipps-University Marburg, Marburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Petra Ina Pfefferle
- Comprehensive Biomaterial Bank Marburg (CBBMR), Philipps-Universitat Marburg, Marburg, Germany
| | - Rajkumar Savai
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Justus Liebig Universitat, Giessen, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Immo Prinz
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sonja Moos
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hyun-Dong Chang
- Institute of Biotechnology, Technische Universität, Berlin, Germany
- German Rheumatism Research Center (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Stefan Heinrich
- Department of Surgery, Johannes Gutenberg University, Mainz, Germany
| | - Detlef K Bartsch
- Division of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Malte Buchholz
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Shiv Singh
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Mengyu Tu
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Lukas Klein
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Goettingen, Germany
| | - Christian Bauer
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Robert Liefke
- Institute of Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Marburg, Germany
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, Philipps University Marburg Faculty of Medicine, Marburg, Germany
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Philipps-University Marburg, Marburg, Germany
| | - Philipp Mayer
- Department of Diagnostic and Interventional Radiology, Heidelberg University, Heidelberg, Germany
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Matthias Gaida
- Institute of Pathology, JGU Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, JGU-Mainz and TRON, Translational Oncology at the University Medical Center, JGU-Mainz, Mainz, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
3
|
Gamal W, Sahakian E, Pinilla-Ibarz J. The role of Th17 cells in chronic lymphocytic leukemia: friend or foe? Blood Adv 2023; 7:2401-2417. [PMID: 36574293 PMCID: PMC10238851 DOI: 10.1182/bloodadvances.2022008985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/18/2022] [Indexed: 12/28/2022] Open
Abstract
T helper 17 (Th17) cells have a prominent role in autoimmune diseases. In contrast, the nature of these cells in cancer is controversial, with either pro- or antitumorigenic activities depending on various cancer settings. Chronic lymphocytic leukemia (CLL), a B-cell malignancy, is characterized by an imbalance in T-cell immune responses that contributes to disease progression and increased mortality. Many clinical reports indicate an increase in Th17 cells and/or interleukin 17 serum cytokine levels in patients with CLL compared with healthy individuals, which correlates with various prognostic markers and significant changes in the tumor microenvironment. The exact mechanisms by which Th17 cells might contribute to CLL progression remain poorly investigated. In this review, we provide an updated presentation of the clinical information related to the significance of Th17 cells in CLL and their interaction with the complex leukemic microenvironment, including various mediators, immune cells, and nonimmune cells. We also address the available data regarding the effects of CLL-targeted therapies on Th17 cells and the potential of using these cells in adoptive cell therapies. Having a sound understanding of the role played by Th17 cells in CLL is crucial for designing novel therapies that can achieve immune homeostasis and maximize clinical benefits.
Collapse
Affiliation(s)
- Wael Gamal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
4
|
Lee YH, Chuah S, Nguyen PHD, Lim CJ, Lai HLH, Wasser M, Chua C, Lim TKH, Leow WQ, Loh TJ, Wan WK, Pang YH, Soon G, Cheow PC, Kam JH, Iyer S, Kow A, Bonney GK, Chan CY, Chung A, Goh BKP, Zhai W, Chow PKH, Albani S, Liu H, Chew V. IFNγ -IL-17 + CD8 T cells contribute to immunosuppression and tumor progression in human hepatocellular carcinoma. Cancer Lett 2023; 552:215977. [PMID: 36279983 DOI: 10.1016/j.canlet.2022.215977] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
IL-17-producing CD8 (Tc17) T cells have been shown to play an important role in infection and chronic inflammation, however their implications in hepatocellular carcinoma (HCC) remain elusive. In this study, we performed cytometry by time-of-flight (CyTOF) and revealed the distinctive immunological phenotypes of two IFNγ+ and IFNγ- Tc17 subsets that were preferentially enriched in human HCC. Single-cell RNA-sequencing analysis further revealed regulatory circuits governing the different phenotypes of these Tc17 subsets. In particular, we discovered that IFNγ- Tc17 subset demonstrated pro-tumoral characteristics and expressed higher levels of CCL20. This corresponded to increased tumor infiltration of T regulatory cells (Treg) validated by immunohistochemistry in another independent HCC cohort, demonstrating the immunosuppressive functions of IFNγ- Tc17 subset. Most importantly, higher intra-tumoral proportions of IFNγ- Tc17 were associated with poorer prognosis in patients with HCC and this was further validated in The Cancer Genome Atlas (TCGA) HCC cohort. Taken together, this compendium of transcriptomic and proteomic data of Tc17 subsets sheds light on the immunosuppressive phenotypes of IFNγ- Tc17 and its implications in HCC progression.
Collapse
Affiliation(s)
- Yun Hua Lee
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Samuel Chuah
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Phuong H D Nguyen
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Chun Jye Lim
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Hannah L H Lai
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Martin Wasser
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Camillus Chua
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Tony K H Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, 169856, Singapore
| | - Wei Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, 169856, Singapore
| | - Tracy Jiezhen Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, 169856, Singapore
| | - Wei Keat Wan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, 169856, Singapore
| | - Yin Huei Pang
- Department of Pathology, National University Hospital Singapore, 119074, Singapore
| | - Gwyneth Soon
- Department of Pathology, National University Hospital Singapore, 119074, Singapore
| | - Peng Chung Cheow
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Juinn Huar Kam
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Shridhar Iyer
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore, 119074, Singapore
| | - Alfred Kow
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore, 119074, Singapore
| | - Glenn K Bonney
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore, 119074, Singapore
| | - Chung Yip Chan
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Alexander Chung
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Brian K P Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore; Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100107, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunan, 650223, China
| | - Pierce K H Chow
- Department of Hepatopancreatobiliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, 169608, Singapore
| | - Salvatore Albani
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute, Immunology Translational Research Program and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, 169856, Singapore.
| |
Collapse
|
5
|
Heim M, Irondelle M, Duteil L, Cardot-Leccia N, Rocchi S, Passeron T, Tulic MK. Impact of topical emollient, steroids alone or combined with calcipotriol, on the immune infiltrate and clinical outcome in psoriasis. Exp Dermatol 2022; 31:1764-1778. [PMID: 36054319 DOI: 10.1111/exd.14657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/25/2022] [Accepted: 08/07/2022] [Indexed: 01/07/2023]
Abstract
Psoriasis is a chronic inflammatory disease whereby long-term disease control remains a challenge for the patients. Latest evidence suggests that combined topical treatment with steroids and vitamin D analogue foam (Calcipotriol/Betamethasone) is efficient in long-term management of the disease and reducing the number of relapses. Its effects on cellular inflammation and cytokine production remain to be explored. We set out to examine the effect of topical therapies on cellular infiltrate and cytokine profile in the lesional skin of psoriasis patients. This was a monocentric, double-blind, randomized trial with 30 patients. Patients were treated with the combined Calcipotriol/Betamethasone foam, Betamethasone foam alone, Clobetasol Propionate ointment or placebo. 4 mm skin biopsies from lesional and non-lesional sites were taken before and 4 weeks after treatment. Cellular infiltrate, IFNγ and IL-17 were studied by immunofluorescence. Each patient was their own control. Evolution in skin inflammation was studied in parallel with changes in patient's epidermal thickness and their tPASI clinical score. Lesional skin was characterized by increased epidermal thickness, increased number of IL-17 and IFNγ producing CD8+ T cells, NK cells and neutrophils. All treatment reduced epidermal thickness and improved patients tPASI scores. Only the combined Calcipotriol/Betamethasone foam completely abolished epidermal and dermal influx of CD8+ T cells, reduced number of CD8 + IFNγ+ cells (but not CD8 + IL-17+ cells) and significantly reduced the number of MPO+ neutrophils which were predominantly IL-17+. None of the treatments had effect on NK cells. We have shown the combined topical treatment with Calcipotriol/Betamethasone foam to be effective in reducing cellular influx into lesional skin of psoriasis patients and this effect to be superior to emollient or Betamethasone alone. Its previously described efficacy in the clinic may be attributed to its unique and rapid ability to inhibit both adaptive CD8+ T cell and innate immune neutrophilia influx into the skin, which was not observed for the other treatments.
Collapse
Affiliation(s)
- Marjorie Heim
- Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France.,Department of Dermatology, CHU Nice, Université Côte d'Azur, Nice, France
| | - Marie Irondelle
- Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France
| | - Luc Duteil
- CHU Nice, Centre de Pharmacologie Clinique Appliquée à la Dermatologie (CPCAD), Université Côte d'Azur, Nice, France
| | | | - Stéphane Rocchi
- Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France
| | - Thierry Passeron
- Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France.,Department of Dermatology, CHU Nice, Université Côte d'Azur, Nice, France
| | - Meri K Tulic
- Centre Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, Nice, France
| |
Collapse
|
6
|
Kartasheva-Ebertz D, Gaston J, Lair-Mehiri L, Mottez E, Buivan TP, Massault PP, Scatton O, Gaujoux S, Vaillant JC, Pol S, Lagaye S. IL-17A in Human Liver: Significant Source of Inflammation and Trigger of Liver Fibrosis Initiation. Int J Mol Sci 2022; 23:ijms23179773. [PMID: 36077175 PMCID: PMC9456490 DOI: 10.3390/ijms23179773] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
IL-17A is considered to guide liver inflammation and fibrosis. From twenty-two human liver samples of different fibrosis stages (F0 to F4), IL-17A, IL-22, and TGFβ1 protein expression in liver tissue lysates were analyzed. Ten paired samples of liver tissue (F0–F1 stage) and blood from the same patient were used to analyze intrahepatic and blood T-lymphoid IL-17A+ cells by flow cytometry. The analyses have been performed regardless of pathology, considering the stage of fibrosis. Human liver tissue was used for the primary human liver slice cultures, followed by subsequent cytokine stimulation and fibrotic markers’ analysis by ELISA. IL-17A production in human liver tissue was significantly higher in the early fibrotic stage compared with the advanced stage. Th17 T cells and, to a lesser extent, MAIT cells were the main sources of IL-17A in both compartments, the liver and the blood. Moreover, the presence of liver Th17IL-17A+INFγ+ cells was detected in the liver. IL-17A stimulation of human liver slice culture increased the expression of profibrotic and pro-inflammatory markers. IL-17A, secreted by Th17 and MAIT cells in the liver, triggered fibrosis by inducing the expression of IL-6 and profibrotic markers and could be a target for antifibrotic treatment. Further amplitude studies are needed to confirm the current results.
Collapse
Affiliation(s)
- Daria Kartasheva-Ebertz
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
- Correspondence:
| | - Jesintha Gaston
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
| | - Loriane Lair-Mehiri
- Department of Hepatology and Addictology, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Estelle Mottez
- Institut Pasteur, Centre de Recherche Translationnelle, F-75015 Paris, France
| | - Tan-Phuc Buivan
- Institut Pasteur, Centre de Recherche Translationnelle, F-75015 Paris, France
| | - Pierre-Philippe Massault
- Department of Digestive Surgery, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Olivier Scatton
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Sebastien Gaujoux
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Jean-Christophe Vaillant
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Stanislas Pol
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
- Department of Hepatology and Addictology, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Sylvie Lagaye
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Centre de Recherche (CDR) Saint-Antoine, INSERM—UMR_S 938/Sorbonne Université, F-75012 Paris, France
| |
Collapse
|
7
|
Hailemichael Y, Johnson DH, Abdel-Wahab N, Foo WC, Bentebibel SE, Daher M, Haymaker C, Wani K, Saberian C, Ogata D, Kim ST, Nurieva R, Lazar AJ, Abu-Sbeih H, Fa'ak F, Mathew A, Wang Y, Falohun A, Trinh V, Zobniw C, Spillson C, Burks JK, Awiwi M, Elsayes K, Soto LS, Melendez BD, Davies MA, Wargo J, Curry J, Yee C, Lizee G, Singh S, Sharma P, Allison JP, Hwu P, Ekmekcioglu S, Diab A. Interleukin-6 blockade abrogates immunotherapy toxicity and promotes tumor immunity. Cancer Cell 2022; 40:509-523.e6. [PMID: 35537412 PMCID: PMC9221568 DOI: 10.1016/j.ccell.2022.04.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 01/21/2022] [Accepted: 04/11/2022] [Indexed: 12/17/2022]
Abstract
Immune checkpoint blockade (ICB) therapy frequently induces immune-related adverse events. To elucidate the underlying immunobiology, we performed a deep immune analysis of intestinal, colitis, and tumor tissue from ICB-treated patients with parallel studies in preclinical models. Expression of interleukin-6 (IL-6), neutrophil, and chemotactic markers was higher in colitis than in normal intestinal tissue; T helper 17 (Th17) cells were more prevalent in immune-related enterocolitis (irEC) than T helper 1 (Th1). Anti-cytotoxic T-lymphocyte-associated antigen 4 (anti-CTLA-4) induced stronger Th17 memory in colitis than anti-program death 1 (anti-PD-1). In murine models, IL-6 blockade associated with improved tumor control and a higher density of CD4+/CD8+ effector T cells, with reduced Th17, macrophages, and myeloid cells. In an experimental autoimmune encephalomyelitis (EAE) model with tumors, combined IL-6 blockade and ICB enhanced tumor rejection while simultaneously mitigating EAE symptoms versus ICB alone. IL-6 blockade with ICB could de-couple autoimmunity from antitumor immunity.
Collapse
Affiliation(s)
- Yared Hailemichael
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel H Johnson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Precision Cancer Therapies Program, Department of Hematology and Medical Oncology, Ochsner Health, New Orleans, LA, USA
| | - Noha Abdel-Wahab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Section of Rheumatology & Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Rheumatology and Rehabilitation, Assiut University Hospitals, Faculty of Medicine, Assiut University, Egypt
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Salah-Eddine Bentebibel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - May Daher
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chantal Saberian
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dai Ogata
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sang T Kim
- Section of Rheumatology & Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roza Nurieva
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hamzah Abu-Sbeih
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faisal Fa'ak
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antony Mathew
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adewunmi Falohun
- Section of Rheumatology & Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van Trinh
- Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chrystia Zobniw
- Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine Spillson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Awiwi
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khaled Elsayes
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brenda D Melendez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jonathan Curry
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Lizee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shalini Singh
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Han W, Li J, Zhou H, Qian J, Tong Z, Wang W, Zhong J, Xue T, Chen Q, Yao Y, Shao S. Identification of the association of CD28 + CD244 + Tc17/IFN-γ cells with chronic hepatitis C virus infection. J Med Virol 2020; 92:3534-3544. [PMID: 32568409 DOI: 10.1002/jmv.26205] [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: 11/08/2019] [Accepted: 06/16/2020] [Indexed: 11/08/2022]
Abstract
CD8+ T cells play multiple and complex immunological roles including antiviral, regulatory, and exhaustive effects in hepatitis C virus (HCV) infected patients. Some CD8+ T-cell subsets were confirmed to be closely related to HCV infection such as TCM , TEM , TEM RA, Tc17, and CD8+ Treg. Herein, we report a new subset of interleukin (IL)-17/interferon (IFN)-γ producing CD8+ T (Tc17/IFN-γ) cells that markedly correlate with CD28+ CD244+ cells, IL-17 levels, and HCV RNA in HCV patients. During early treatment with peg-IFN-a2a plus ribavirin, the imbalance of these Tc17/IFN-γ cells could be partially restored, together with normalized serum alanine aminotransferase but not aspartate transaminase. Also, we analyzed the dynamic change of the percentage of this T cells subset in patients with different outcome after 4-week course of treatment with peg-IFN-a2a plus ribavirin and found that the percentage of CD8+ CD28+ CD244+ T cells significantly decreased in recovered patients but not in nonrecovered patients. In vitro, CD28+ CD244+ T cells were the only CD8+ T-cell group that secreted both IL-17 and IFN-γ in this axis and blockade with anti-CD244 antibodies significantly reduced cytokine production. Taken together, this study demonstrates that the frequency and regulatory functions of CD28+ CD244+ Tc17/IFN-γ cells may play an important role in persistent HCV infection.
Collapse
Affiliation(s)
- Wenzheng Han
- Clinical Laboratory, First Affiliated Hospital of Wanan Medical College, Wuhu, Anhui, China
| | - Jiajia Li
- Clinical Laboratory, First Affiliated Hospital of Wanan Medical College, Wuhu, Anhui, China
| | - Hongchang Zhou
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou Central Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Jing Qian
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou Central Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Zhaowei Tong
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Weihong Wang
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jianfeng Zhong
- Department of Infectious Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Tao Xue
- Department of Medical Therapeutics, First People's Hospital Affiliated to Huzhou University, Huzhou, Zhejiang, China
| | - Qing Chen
- Clinical Laboratory, First Affiliated Hospital of Wanan Medical College, Wuhu, Anhui, China
| | - Yunliang Yao
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou Central Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Shengwen Shao
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou Central Hospital, Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
9
|
Liang J, Nong S, Jiang L, Chi X, Bi D, Cao J, Mo L, Luo X, Huang H. Correlations of disease severity and age with hematology parameter variations in patients with COVID-19 pre- and post-treatment. J Clin Lab Anal 2020; 35:e23609. [PMID: 33222271 PMCID: PMC7744896 DOI: 10.1002/jcla.23609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 01/08/2023] Open
Abstract
Background For better understanding of the pathological changes of COVID‐19, benefiting clinical management of the disease and the preparation for future waves of similar pandemics. Methods Hematology parameters from a total of 52 cases of COVID‐19 admitted for treatment in a designated hospital were retrospectively analyzed. Data were analyzed by SPSS statistical software. Results Pre‐treatment T‐cell subsets, total lymphocytes, red blood cell distribution width (RDW), eosinophils, and basophils were significantly lower than that of post‐treatment, while the inflammatory indexes neutrophils, neutrophil to lymphocyte ratio (NLR), and C‐reactive protein (CRP) levels, as well as red blood cell (RBC) and hemoglobin, were significantly reduced after treatment. The T‐cell subsets, total lymphocytes, and basophils in severely and critically ill patients were significantly lower than those in moderately ill patients. Neutrophils, NLR, eosinophils, procalcitonin (PCT), and CRP was significantly higher in severely and critically ill patients than in moderately ill patients. CD3+, CD8+, total lymphocytes, platelets, and basophils in patients older than 50 were lower than that of those younger than 50, while neutrophils, NLR, CRP, and RDW in patients older than 50 were higher than that of younger than 50. There was a positive correlation among prothrombin time (PT), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) in severely and critically ill patients. Conclusions T‐cell subsets, lymphocyte count, RDW, neutrophils, eosinophils, NLR, CRP, PT, ALT, and AST are important indicators in the management especially for severely and critically ill patients with COVID‐19.
Collapse
Affiliation(s)
- Juanying Liang
- Department of Laboratory Medicine, The Forth People's Hospital of Nanning City, Nanning, China
| | - Shaoyun Nong
- Department of Laboratory Medicine, Guangxi Nationalities Hospital, Nanning, China
| | - Liejun Jiang
- Department of Laboratory Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaowei Chi
- Department of Laboratory Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dewu Bi
- Department of Laboratory Medicine, The Forth People's Hospital of Nanning City, Nanning, China
| | - Jun Cao
- Department of Laboratory Medicine, The Forth People's Hospital of Nanning City, Nanning, China
| | - Lida Mo
- Department of Laboratory Medicine, The Forth People's Hospital of Nanning City, Nanning, China
| | - Xiaolu Luo
- Department of Laboratory Medicine, The Forth People's Hospital of Nanning City, Nanning, China
| | - Huayi Huang
- School of Medical Laboratory, Youjiang Medical University for Nationalities, Baise, China.,Mindray North America, Mahwah, NJ, USA.,Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
10
|
Corgnac S, Malenica I, Mezquita L, Auclin E, Voilin E, Kacher J, Halse H, Grynszpan L, Signolle N, Dayris T, Leclerc M, Droin N, de Montpréville V, Mercier O, Validire P, Scoazec JY, Massard C, Chouaib S, Planchard D, Adam J, Besse B, Mami-Chouaib F. CD103 +CD8 + T RM Cells Accumulate in Tumors of Anti-PD-1-Responder Lung Cancer Patients and Are Tumor-Reactive Lymphocytes Enriched with Tc17. CELL REPORTS MEDICINE 2020; 1:100127. [PMID: 33205076 PMCID: PMC7659589 DOI: 10.1016/j.xcrm.2020.100127] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 07/24/2020] [Accepted: 09/24/2020] [Indexed: 12/23/2022]
Abstract
Accumulation of CD103+CD8+ resident memory T (TRM) cells in human lung tumors has been associated with a favorable prognosis. However, the contribution of TRM to anti-tumor immunity and to the response to immune checkpoint blockade has not been clearly established. Using quantitative multiplex immunofluorescence on cohorts of non-small cell lung cancer patients treated with anti-PD-(L)1, we show that an increased density of CD103+CD8+ lymphocytes in immunotherapy-naive tumors is associated with greatly improved outcomes. The density of CD103+CD8+ cells increases during immunotherapy in most responder, but not in non-responder, patients. CD103+CD8+ cells co-express CD49a and CD69 and display a molecular profile characterized by the expression of PD-1 and CD39. CD103+CD8+ tumor TRM, but not CD103−CD8+ tumor-infiltrating counterparts, express Aiolos, phosphorylated STAT-3, and IL-17; demonstrate enhanced proliferation and cytotoxicity toward autologous cancer cells; and frequently display oligoclonal expansion of TCR-β clonotypes. These results explain why CD103+CD8+ TRM are associated with better outcomes in anti-PD-(L)1-treated patients. A high density of CD103+CD8+ cells in tumors correlates with response to anti-PD-(L)1 The density of CD103+CD8+ cells increases after anti-PD-1 in most responder patients CD103+CD8+ TRM cells are enriched with tumor-specific T cells A subset of CD103+CD8+ TRM cells display a Tc17 differentiation program
Collapse
Affiliation(s)
- Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Ines Malenica
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Laura Mezquita
- Department of Cancer Medicine, Gustave Roussy, Institut d'Oncologie Thoracique, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Edouard Auclin
- Gastrointestinal and Medical Oncology Department, Hôpital Européen Georges Pompidou, Paris, France
| | - Elodie Voilin
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Jamila Kacher
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Heloise Halse
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Laetitia Grynszpan
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Nicolas Signolle
- INSERM Unit U981, Department of Experimental Pathology, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Thibault Dayris
- Department of Biology and Medical Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Marine Leclerc
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Nathalie Droin
- Department of Biology and Medical Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Vincent de Montpréville
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France.,Hôpital Marie-Lannelongue, Service d'Anatomie Pathologique, 92350 Le-Plessis-Robinson, France
| | - Olaf Mercier
- Hôpital Marie-Lannelongue, Service d'Anatomie Pathologique, 92350 Le-Plessis-Robinson, France
| | - Pierre Validire
- Institut Mutualiste Montsouris, Service d'Anatomie Pathologique, 75014 Paris, France
| | - Jean-Yves Scoazec
- Department of Biology and Medical Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Christophe Massard
- Drug Development Department, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - David Planchard
- Department of Cancer Medicine, Gustave Roussy, Institut d'Oncologie Thoracique, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Julien Adam
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Institut d'Oncologie Thoracique, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| |
Collapse
|
11
|
Pereira Neto TA, Gonçalves-Pereira MH, de Queiroz CP, Ramos MF, de Oliveira FDFS, Oliveira-Prado R, do Nascimento VA, Abdalla LF, Santos JHA, Martins-Filho OA, Naveca FG, Teixeira-Carvalho A, Santiago HDC. Multifunctional T cell response in convalescent patients two years after ZIKV infection. J Leukoc Biol 2020; 108:1265-1277. [PMID: 32726884 DOI: 10.1002/jlb.4ma0520-708r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Zika is an important emerging infectious disease in which the role of T cells remains elusive. This study aimed to evaluate the phenotype of multifunctional T cells in individuals 2 yr after exposure to Zika virus (ZIKV). We used a library of 671 synthetic peptides covering the whole polyprotein of ZIKV in pools corresponding to each viral protein (i.e., capsid, membrane precursor or prM, envelope, NS1 [nonstructural protein], NS2A + NS2B, NS3, NS4A + NS4B, and NS5) to stimulate PBMCs from individuals previously exposed to ZIKV. We observed an increased frequency of ZIKV-specific IFNγ, IL-17A, TNF, and IL-10 production by T cell populations. IFNγ and TNF production were especially stimulated by prM, capsid, or NS1 in CD8+ T cells and by capsid or prM in CD4+ T cells. In addition, there was an increase in the frequency of IL-10+ CD8+ T cells after stimulation with prM, capsid, NS1, NS3, or NS5. Multifunctional properties were observed in ZIKV-specific T cells responding especially to prM, capsid, NS1 or, to a smaller extent, NS3 antigens. For example, we found a consistent IFNγ + TNF+ CD8+ T cell population in response to most virus antigens and CD4+ and CD8+ T cells that were IFNγ + IL-17A+ and IL-17A+IL-10+, which could also produce TNF, in response to capsid, prM, NS1, or NS3 stimulation. Interestingly, CD8+ T cells were more prone to a multifunctional phenotype than CD4+ T cells, and multifunctional T cells were more efficient at producing cytokines than single-function cells. This work provides relevant insights into the quality of ZIKV-specific T cell responses and ZIKV immunity.
Collapse
Affiliation(s)
| | | | - Camila Pereira de Queiroz
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michele Faria Ramos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | - Felipe Gomes Naveca
- Leonidas e Maria Deane Institute, Oswaldo Cruz Foundation, Manaus, Amazonas, Brazil
| | | | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
12
|
LRCH1 deficiency enhances LAT signalosome formation and CD8 + T cell responses against tumors and pathogens. Proc Natl Acad Sci U S A 2020; 117:19388-19398. [PMID: 32727906 DOI: 10.1073/pnas.2000970117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CD8+ T cells play pivotal roles in eradicating pathogens and tumor cells. T cell receptor (TCR) signaling is vital for the optimal activation of CD8+ T cells. Upon TCR engagement, the transmembrane adapter protein LAT (linker for activation of T cells) recruits other key signaling molecules and forms the "LAT signalosome" for downstream signal transduction. However, little is known about which functional partners could restrain the formation of the LAT signalosome and inhibit CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity. Here we have demonstrated that LRCH1 (leucine-rich repeats and calponin homology domain containing 1) directly binds LAT, reduces LAT phosphorylation and interaction with GRB2, and also promotes the endocytosis of LAT. Lrch1 -/- mice display better protection against influenza virus and Listeria infection, with enhanced CD8+ T cell proliferation and cytotoxicity. Adoptive transfer of Lrch1 -/- CD8+ CTLs leads to increased B16-MO5 tumor clearance in vivo. Furthermore, knockout of LRCH1 in human chimeric antigen receptor (CAR) T cells that recognize the liver tumor-associated antigen glypican-3 could improve CAR T cell migration and proliferation in vitro. These findings suggest LRCH1 as a potential translational target to improve T cell immunotherapy against infection and tumors.
Collapse
|
13
|
St Paul M, Ohashi PS. The Roles of CD8 + T Cell Subsets in Antitumor Immunity. Trends Cell Biol 2020; 30:695-704. [PMID: 32624246 DOI: 10.1016/j.tcb.2020.06.003] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/04/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
Effector CD8+ T cells are typically thought to be a homogenous group of cytotoxic cells that produce interferon-(IFN) γ. However, recent findings have challenged this notion because multiple subsets of CD8+ T cells have been described, each with distinct effector functions and cytotoxic potential. These subsets, referred to as the Tc subsets, have also been detected in tumor microenvironments (TMEs), where they potentially influence the antitumor response and patient outcomes. In this review, we highlight the prevalence and roles of Tc subsets in the TME. We also discuss their therapeutic applications in the context of adoptive immunotherapy to treat cancer.
Collapse
Affiliation(s)
- Michael St Paul
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2C1, Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1C1, Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2C1, Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1C1, Canada.
| |
Collapse
|
14
|
Kannan N, Haug M, Steigedal M, Flo TH. Mycobacterium smegmatis Vaccine Vector Elicits CD4+ Th17 and CD8+ Tc17 T Cells With Therapeutic Potential to Infections With Mycobacterium avium. Front Immunol 2020; 11:1116. [PMID: 32582196 PMCID: PMC7296097 DOI: 10.3389/fimmu.2020.01116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/07/2020] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium avium (Mav) complex is increasingly reported to cause non-tuberculous infections in individuals with a compromised immune system. Treatment is complicated and no vaccines are available. Previous studies have shown some potential of using genetically modified Mycobacterium smegmatis (Msm) as a vaccine vector to tuberculosis since it is non-pathogenic and thus would be tolerated by immunocompromised individuals. In this study, we used a mutant strain of Msm disrupted in EspG3, a component of the ESX-3 secretion system. Infection of macrophages and dendritic cells with Msm ΔespG3 showed increased antigen presentation compared to cells infected with wild-type Msm. Vaccination of mice with Msm ΔespG3, expressing the Mav antigen MPT64, provided equal protection against Mav infection as the tuberculosis vaccine, Mycobacterium bovis BCG. However, upon challenge with Mav, we observed a high frequency of IL-17-producing CD4+ (Th17 cells) and CD8+ (Tc17 cells) T cells in mice vaccinated with Msm ΔespG3::mpt64 that was not seen in BCG-vaccinated mice. Adoptive transfer of cells from Msm ΔespG3-vaccinated mice showed that cells from the T cell compartment contributed to protection from Mav infection. Further experiments revealed Tc17-enriched T cells did not provide prophylactic protection against subsequent Mav infection, but a therapeutic effect was observed when Tc17-enriched cells were transferred to mice already infected with Mav. These initial findings are important, as they suggest a previously unknown role of Tc17 cells in mycobacterial infections. Taken together, Msm ΔespG3 shows promise as a vaccine vector against Mav and possibly other (myco)bacterial infections.
Collapse
Affiliation(s)
- Nisha Kannan
- Center of Molecular Inflammation Research and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Markus Haug
- Center of Molecular Inflammation Research and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Infectious Diseases, St. Olavs University Hospital, Trondheim, Norway
| | - Magnus Steigedal
- Center of Molecular Inflammation Research and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Infectious Diseases, St. Olavs University Hospital, Trondheim, Norway
| | - Trude Helen Flo
- Center of Molecular Inflammation Research and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
15
|
Kuen DS, Kim BS, Chung Y. IL-17-Producing Cells in Tumor Immunity: Friends or Foes? Immune Netw 2020; 20:e6. [PMID: 32158594 PMCID: PMC7049578 DOI: 10.4110/in.2020.20.e6] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
IL-17 is produced by RAR-related orphan receptor gamma t (RORγt)-expressing cells including Th17 cells, subsets of γδT cells and innate lymphoid cells (ILCs). The biological significance of IL-17-producing cells is well-studied in contexts of inflammation, autoimmunity and host defense against infection. While most of available studies in tumor immunity mainly focused on the role of T-bet-expressing cells, including cytotoxic CD8+ T cells and NK cells, and their exhaustion status, the role of IL-17-producing cells remains poorly understood. While IL-17-producing T-cells were shown to be anti-tumorigenic in adoptive T-cell therapy settings, mice deficient in type 17 genes suggest a protumorigenic potential of IL-17-producing cells. This review discusses the features of IL-17-producing cells, of both lymphocytic and myeloid origins, as well as their suggested pro- and/or anti-tumorigenic functions in an organ-dependent context. Potential therapeutic approaches targeting these cells in the tumor microenvironment will also be discussed.
Collapse
Affiliation(s)
- Da-Sol Kuen
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,BK21 Plus Program, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Byung-Seok Kim
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,BK21 Plus Program, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
16
|
Song L, Ma S, Chen L, Miao L, Tao M, Liu H. Long-term prognostic significance of interleukin-17-producing T cells in patients with non-small cell lung cancer. Cancer Sci 2019; 110:2100-2109. [PMID: 31100180 PMCID: PMC6609818 DOI: 10.1111/cas.14068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 12/16/2022] Open
Abstract
The presence of interleukin (IL)‐17‐producing T cells has recently been reported in non‐small cell lung cancer (NSCLC) patients. However, the long‐term prognostic significance of these populations in NSCLC patients remains unknown. In the present study, we collected peripheral blood from 82 NSCLC patients and 22 normal healthy donors (NC). Percentages of IL‐17‐producing CD4+T (Th17), CD8+T (Tc17) and γδT cells (γδT17) were measured to determine their association with clinical outcomes and overall survival (OS) in NSCLC. All NSCLC patients were followed up until July 2018. Median follow‐up time was 13.5 months (range 1‐87 months). The 3‐ and 5‐year survival rate was 27% and 19.6%, respectively. We found that Th17 cells and γδT17 cells were significantly increased, whereas Tc17 cells were markedly decreased in patients with NSCLC compared with those in NC. In addition, Th17 cells were significantly positively associated with T helper type 1 cells (Th1), whereas γδT17 cells were significantly negatively associated with γδT + interferon (IFN)‐γ+ cells. High percentages of peripheral Tc17 cells were significantly associated with favorable 5‐year OS (P = .025), especially in patients with early TNM stage (P = .016). Furthermore, high percentages of peripheral Th17 cells were positively associated with favorable 5‐year OS in patients with late TNM stage (P = .002). However, no significant association was observed between γδT17 cells and OS, regardless of the TNM stage. In conclusion, our findings suggest that enhanced Th17 and reduced Tc17 cells in the peripheral blood could be a significant predictor of a favorable prognosis for NSCLC patients.
Collapse
Affiliation(s)
- Li Song
- Department of Oncology, Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Shoubao Ma
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Longpei Chen
- Department of Oncology, Shanghai Changhai Hospital, Shanghai, China
| | - Liyan Miao
- Department of Oncology, Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore
| |
Collapse
|
17
|
Liu X, Zawidzka EM, Li H, Lesch CA, Dunbar J, Bousley D, Zou W, Hu X, Carter LL. RORγ Agonists Enhance the Sustained Antitumor Activity through Intrinsic Tc17 Cytotoxicity and Tc1 Recruitment. Cancer Immunol Res 2019; 7:1054-1063. [PMID: 31064778 DOI: 10.1158/2326-6066.cir-18-0714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/06/2019] [Accepted: 05/02/2019] [Indexed: 11/16/2022]
Abstract
Activation of RORγ with synthetic small-molecule agonists has been shown to enhance type 17 effector (CD4+ Th17 and CD8+ Tc17 cells) cell functions and decrease immunosuppressive mechanisms, leading to improved antitumor efficacy in adoptive cell transfer and syngeneic murine tumor models. However, whether Tc17 cells possess intrinsic cytotoxicity and the mechanism they use to lyse target cells is controversial. We report here that Tc17 cells were lytic effectors dependent on perforin and granzyme A. In contrast to Tc1 cells, Tc17 cells resisted activation-induced cell death and maintained granzyme A levels, which conferred the ability to lyse target cells in serial encounters. Thus, although the acute lytic capacity of Tc17 cells could be inferior to Tc1 cells, comparable lysis was achieved over time. In addition to direct lytic activity, Tc17 cells infiltrated early into the tumor mass, recruited other CD8+ T cells to the tumor, and enhanced the survival and lytic capability of these cells during repeated target encounters. Synthetic RORγ agonists further augmented Tc17 survival and lytic activity in vitro and in vivo, controlling tumor growth not only through direct cytotoxicity, but also through recruitment and improved function of other effector cells in the tumor microenvironment, which suggests complementary and cooperate activities for effective immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Weiping Zou
- University of Michigan Medical School, Ann Arbor, Michigan
| | - Xiao Hu
- Lycera Corp. Ann Arbor, Michigan
| | | |
Collapse
|
18
|
Rezalotfi A, Ahmadian E, Aazami H, Solgi G, Ebrahimi M. Gastric Cancer Stem Cells Effect on Th17/Treg Balance; A Bench to Beside Perspective. Front Oncol 2019; 9:226. [PMID: 31024835 PMCID: PMC6464032 DOI: 10.3389/fonc.2019.00226] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 03/13/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer stem cells (GCSCs), a small population among tumor cells, are responsible for tumor initiation, development, metastasis, and recurrence. They play a crucial role in immune evasion, immunomodulation, and impairment of effector immunity and believed to be emerged to change the balance of the immune system, importantly CD4+ T cells in the chronic inflamed tumor site. However, different subtypes of innate and adaptive immune cells are involved in the formation of the immune system in the tumor microenvironment, we would look at T cells in this study. Tumor microenvironment induces differentiation of CD4+ T cells into different subsets of T cells, mainly suppressive regulatory T cells (Treg), and T helper 17 (Th17) cells, although their exact role in tumor immunity is still under debate depending on tumor types and stages. Counterbalance between Th17 and Treg cells in the gastrointestinal system result in the homeostasis and normal function of the immune system, particularly mucosal immunity. Recent data demonstrated a high infiltration of Th17 and Treg cells into the gastric tumor site and proved that tumor microenvironment might disturb the balance between Th17 and Treg. It is possible to assume an association between activation of CSCs which contribute to metastasis in late stages, and the imbalanced Th17/Treg cells observed in advanced gastric cancer patients. This review intends to clarify the importance of gastric tumor microenvironment specifically CSCs in relation to Th17/Tregs balance firstly and to highlight the relevance of imbalanced Th17/Treg subsets in determining the stages and behavior of the tumor secondly. Finally, the present study suggests a clinical approach looking at the plasticity of T cells with a focus on Th17 as a promising dedicated arm in cancer immunotherapy.
Collapse
Affiliation(s)
- Alaleh Rezalotfi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elmira Ahmadian
- Faculty of Biological Sciences and Technology, Department of Animal Sciences, Shahid Beheshti University, Tehran, Iran
| | - Hossein Aazami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- *Correspondence: Ghasem Solgi
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Marzieh Ebrahimi
| |
Collapse
|
19
|
Perdomo-Celis F, Feria MG, Taborda NA, Rugeles MT. A Low Frequency of IL-17-Producing CD8 + T-Cells Is Associated With Persistent Immune Activation in People Living With HIV Despite HAART-Induced Viral Suppression. Front Immunol 2018; 9:2502. [PMID: 30420859 PMCID: PMC6215827 DOI: 10.3389/fimmu.2018.02502] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Immune activation is the hallmark of HIV infection, even in patients with highly active anti-retroviral therapy (HAART)-induced viral suppression. A major cause of immune activation during HIV infection is the intestinal microbial translocation as a consequence, among other factors, of the decrease and/or dysfunction of interleukin (IL)-17-producing T-cells, due to their role promoting the integrity of the intestinal barrier. A population of IL-17-producing CD8+ T-cells (Tc17 cells), characterized by the expression of CD161, has been described, but its relation with the persistent immune activation in non-viremic people living with HIV (PLWH) on HAART is unclear. By flow cytometry, we characterized the activation phenotype (evaluated by the expression of HLA-DR and CD38) of circulating CD161-expressing CD8+ T-cells; in addition, we explored the functionality of polyclonally-stimulated Tc17 cells in PLWH under HAART-induced viral suppression, and in healthy individuals. Finally, we determined the association of Tc17 cells with the expression of cellular and soluble activation markers. Circulating CD161-expressing CD8+ T-cells were decreased in PLWH compared with healthy individuals, despite their similar basal activation state. After polyclonal stimulation, IL-17 production was higher in CD8+ T-cells co-expressing HLA-DR and CD38 in healthy individuals. In contrast, although PLWH had a higher frequency of HLA-DR+ CD38+ CD8+ T-cells after stimulation, they had a lower production of IL-17. Interferon (IFN)-γ-producing CD8+ T-cells (Tc1 cells) were increased in PLWH. The low Tc17 cells response was associated with a high expression of CD38 and programmed death 1 protein, high levels of soluble CD14 and the treatment duration. Finally, to explore potential immunomodulatory strategies, the in vitro effect of the anti-inflammatory agent sulfasalazine was assessed on Tc17 cells. Interestingly, a decreased inflammatory environment, death of activated CD8+ T-cells, and an increased frequency of Tc17 cells were observed with sulfasalazine treatment. Thus, our findings suggest that activated CD8+ T-cells have a marked capacity to produce IL-17 in healthy individuals, but not in PLWH, despite HAART. This dysfunction of Tc17 cells is associated with the persistent immune activation observed in these patients, and can be partially restored by anti-inflammatory agents.
Collapse
Affiliation(s)
- Federico Perdomo-Celis
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Manuel G Feria
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Natalia A Taborda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Maria T Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
20
|
Kostic M, Zivkovic N, Cvetanovic A, Stojanovic I. Granulocyte-macrophage colony-stimulating factor as a mediator of autoimmunity in multiple sclerosis. J Neuroimmunol 2018; 323:1-9. [DOI: 10.1016/j.jneuroim.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/18/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022]
|
21
|
Alves JJP, De Medeiros Fernandes TAA, De Araújo JMG, Cobucci RNO, Lanza DCF, Bezerra FL, Andrade VS, Fernandes JV. Th17 response in patients with cervical cancer. Oncol Lett 2018; 16:6215-6227. [PMID: 30405758 PMCID: PMC6202464 DOI: 10.3892/ol.2018.9481] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022] Open
Abstract
Persistent infection by high-risk human papillomavirus (HR-HPV) is the main risk factor for uterine cervical cancer (UCC). However, viral infection alone is not sufficient for the development and progression of premalignant cervical lesions for cancer. In previous years it has been suggested that the adaptive immune response triggered by the differentiation of naïve helper T cells in Th17 cells may serve an important role in disease development. It has been hypothesized that Th17 cells may be involved in the promotion of UCC, as high levels of interleukin 17 (IL17) expression have been detected in the mucosa of the uterine cervix of patients affected by the disease. However, the role of Th17 cells in the tumor development and progression remains unclear. It is believed that the immune response of the Th17 type during persistent infection of the genital tract with HR-HPV triggers chronic inflammation with a long duration with the production of IL17 and other pro-inflammatory cytokines, creating a favorable environment for tumor development. These cytokines are produced by immune system cells in addition to tumor cells and appear to function by modulating the host immune system, resulting in an immunosuppressive response as opposed to inducing an effective protective immune response, thus contributing to the growth and progression of the tumor. In the present review, the latest advances are presented about the function of Th17 cells and the cytokines produced by them in the development and progression of UCC.
Collapse
Affiliation(s)
- Jayra Juliana Paiva Alves
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | | | | | | | | | - Fabiana Lima Bezerra
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | - Vânia Sousa Andrade
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| |
Collapse
|
22
|
Cheon SY, Kim JM, Kim EJ, Kim SY, Kam EH, Ho CC, Lee SK, Koo BN. Intranuclear delivery of synthetic nuclear factor-kappa B p65 reduces inflammasomes after surgery. Biochem Pharmacol 2018; 158:141-152. [PMID: 30096289 DOI: 10.1016/j.bcp.2018.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Patients undergoing surgery can suffer from various complications, including post-operative bleeding, local or systematic infection, and neurologic disorders. Major surgery can initiate innate immune responses and trigger overproduction of inflammatory mediators, which can contribute to organ dysfunction. Inflammasomes are innate immune complexes, which are connected to the pathogenesis of various diseases, including atherosclerosis, hemorrhagic brain injury, and Alzheimer's disease. In the present study, we hypothesized that nucleotide-binding oligomerization domain-containing-like receptor protein (NLRP) inflammasomes may have a role in the pathological effects of surgery. Therefore, we designed a protein inhibitor of nuclear factor kappa B (NF-κB) p65 transcripts, called nt-p65-TMD (nuclear transducible (nt) transcription modulated domain (TMD) of RelA (p65)), that can penetrate the nucleus, and evaluated its therapeutic efficacy for dampening surgery-induced inflammasome activation. It was found that the nt-p65-TMD significantly reduced the NLRP1 inflammasome complex components (NLRP1, ASC, and Caspase-1) and interleukin (IL)-1β and IL-18 productions in the spleen after surgery. In the spleen, specific cell population and selective mediators were altered after surgery with/without nt-p65-TMD treatment. Also, we found that treatment of nt-p65-TMD decreased cell death in the spleen after surgery. Therefore, nt-p65-TMD is a potential novel strategy for reducing surgery-induced NLRP1 inflammasome and complications.
Collapse
Affiliation(s)
- So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Min Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Kam
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chun-Chang Ho
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Bon-Nyeo Koo
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Hogg A, Sui Y, Ben-Sasson SZ, Paul WE, Berzofsky JA. Role of CD4 T cell helper subsets in immune response and deviation of CD8 T cells in mice. Eur J Immunol 2017; 47:2059-2069. [PMID: 28741316 DOI: 10.1002/eji.201747091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/15/2017] [Accepted: 07/19/2017] [Indexed: 02/02/2023]
Abstract
The ability of different CD4+ T cell subsets to help CD8+ T-cell response is not fully understood. Here, we found using the murine system that Th17 cells induced by IL-1β, unlike Th1, were not effective helpers for antiviral CD8 responses as measured by IFNγ-producing cells or protection against virus infection. However, they skewed CD8 responses to a Tc17 phenotype. Thus, the apparent lack of help was actually immune deviation. This skewing depended on both IL-21 and IL-23. To overcome this effect, we inhibited Th17 induction by blocking TGF-β. Anti-TGF-β allowed the IL-1β adjuvant to enhance CD8+ T-cell responses without skewing the phenotype to Tc17, thereby providing an approach to harness the benefit of common IL-1-inducing adjuvants like alum without immune deviation.
Collapse
Affiliation(s)
- Alison Hogg
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| | - Shlomo Z Ben-Sasson
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Lautenberg Center for General and Tumor Immunology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - William E Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
24
|
Nanjappa SG, McDermott AJ, Fites JS, Galles K, Wüthrich M, Deepe GS, Klein BS. Antifungal Tc17 cells are durable and stable, persisting as long-lasting vaccine memory without plasticity towards IFNγ cells. PLoS Pathog 2017; 13:e1006356. [PMID: 28542595 PMCID: PMC5456400 DOI: 10.1371/journal.ppat.1006356] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 06/02/2017] [Accepted: 04/14/2017] [Indexed: 01/01/2023] Open
Abstract
Our understanding of persistence and plasticity of IL-17A+ memory T cells is clouded by conflicting results in models analyzing T helper 17 cells. We studied memory IL-17A+ CD8+ T-cell (Tc17) homeostasis, persistence and plasticity during fungal vaccine immunity. We report that vaccine-induced memory Tc17 cells persist with high fidelity to the type 17 phenotype. Tc17 cells persisted durably for a year as functional IL-17A+ memory cells without converting to IFNγ+ (Tc1) cells, although they produced multiple type I cytokines in the absence of residual vaccine antigen. Memory Tc17 cells were canonical CD8+ T cells with phenotypic features distinct from Tc1 cells, and were Ror(γ)thi, TCF-1hi, T-betlo and EOMESlo. In investigating the bases of Tc17 persistence, we observed that memory Tc17 cells had much higher levels of basal homeostatic proliferation than did Tc1 cells. Conversely, memory Tc17 cells displayed lower levels of anti-apoptotic molecules Bcl-2 and Bcl-xL than Tc1 cells, yet were resistant to apoptosis. Tc1 cells required Bcl-2 for their survival, but Bcl-2 was dispensable for the maintenance of Tc17 cells. Tc17 and Tc1 cells displayed different requirements for HIF-1α during effector differentiation and sustenance and memory persistence. Thus, antifungal vaccination induces durable and stable memory Tc17 cells with distinct requirements for long-term persistence that distinguish them from memory Tc1 cells. CD4+ T-cell deficient patients such as those with AIDS and idiopathic CD4+ T-cell lymphopenia are vulnerable to systemic fungal infections. We previously showed that CD8+ T cells can be exploited in CD4+ T cell deficient hosts for vaccine immunity against lethal fungal pneumonia in mice and that IL-17A production by these cells (Tc17) is essential. Existing dogma holds that IL-17A producing CD4+ T cells (Th17) are highly plastic, unstable, and convert into IFNγ producing cells, losing the capacity to produce IL-17A, which is the signature feature of Tc17 cells. Here, we show that vaccine-elicited antifungal Tc17 cells are maintained as stable and long-lasting memory cells that resist conversion into IFNγ cells (Tc1) and protect CD4+ T cell deficient hosts against lethal pulmonary fungal infection. Antifungal Tc17 cells displayed features that define classical memory cells. However, memory Tc17 exhibited different requirements than Tc1 cells in the factors that promote T cell survival, including anti-apoptotic molecules Bcl-2 and Bcl-xl, and HIF-1α, which aids survival of cells in lower oxygen conditions found during inflammation. Thus, our study reveals that fungal vaccination elicits a durable, stable population of Tc17 cells with distinct features of survival needed for preventing infection in immunocompromised hosts.
Collapse
Affiliation(s)
- Som Gowda Nanjappa
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- * E-mail: (SGN); (BSK)
| | - Andrew J. McDermott
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - J. Scott Fites
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Kevin Galles
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Marcel Wüthrich
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - George S. Deepe
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati, College of Medicine, Cincinnati, OH, United States of America
| | - Bruce S. Klein
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- * E-mail: (SGN); (BSK)
| |
Collapse
|
25
|
Xu WH, Hu XL, Liu XF, Bai P, Sun YC. Peripheral Tc17 and Tc17/Interferon-γ Cells are Increased and Associated with Lung Function in Patients with Chronic Obstructive Pulmonary Disease. Chin Med J (Engl) 2017; 129:909-16. [PMID: 27064034 PMCID: PMC4831524 DOI: 10.4103/0366-6999.179798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Chronic obstructive pulmonary disease (COPD) is characterized by progressive loss of lung function and local and systemic inflammation, in which CD8+ T-cells are believed to play a key role. Activated CD8+ T-cells differentiate into distinct subpopulations, including interferon-γ (IFN-γ)-producing Tc1 and interleukin (IL)-17-producing Tc17 cells. Recent evidence indicates that Tc17 cells exhibit considerable plasticity and may convert into IL-17/IFN-γ-double producing (Tc17/IFN-γ) cells when driven by inflammatory conditions. The aim of this study was to investigate the Tc17/IFN-γ subpopulation in peripheral blood of patients with COPD and to evaluate their potential roles in this disease. Methods: Peripheral blood samples were collected from 15 never-smokers, 23 smokers with normal lung function, and 25 patients with COPD (Global Initiative for Chronic Obstructive Lung Disease 2–4). Proportions of the IL-17/IFN-γ-double expressing subpopulation were assessed using flow cytometry. Plasma concentrations of cytokines favoring Tc17/IFN-γ differentiation were measured by enzyme-linked immunosorbent assay. Results: Patients with COPD had higher proportions of Tc17 cells and Tc17/IFN-γ cells in the peripheral blood than smokers and never-smokers. The plasticity of Tc17 cells was higher than that of Th17 cells. The percentages of Tc17 cells and Tc17/IFN-γ cells showed negative correlations with forced expiratory volume in 1 s % predicted value (r = −0.418, P = 0.03; r = −0.596, P = 0.002, respectively). The plasma concentrations of IL-6, transforming growth factor-β1, and IL-12 were significantly higher in patients with COPD compared with smokers and never-smokers. Conclusions: Peripheral Tc17 cells are increased and more likely to convert to Tc17/IFN-γ cells in COPD, suggesting that Tc17 cell plasticity may be involved in persistent inflammation of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Yong-Chang Sun
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing 100730; Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
26
|
Th17 plasticity and transition toward a pathogenic cytokine signature are regulated by cyclosporine after allogeneic SCT. Blood Adv 2017; 1:341-351. [PMID: 29296949 DOI: 10.1182/bloodadvances.2016002980] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/08/2016] [Indexed: 11/20/2022] Open
Abstract
T-helper 17 (Th17) cells have been widely implicated as drivers of autoimmune disease. In particular, Th17 cytokine plasticity and acquisition of an interleukin-17A+(IL-17A+)interferon γ(IFNγ)+ cytokine profile is associated with increased pathogenic capacity. Donor Th17 polarization is known to exacerbate graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (allo-SCT); however, donor Th17 cytokine coexpression and plasticity have not been fully characterized. Using IL-17 "fate-mapping" mice, we identified IL-6-dependent Th17 cells early after allo-SCT, characterized by elevated expression of proinflammatory cytokines, IL-17A, IL-22, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor. This population did not maintain lineage fidelity, with a marked loss of IL-17A and IL-22 expression late posttransplant. Th17 cells were further segregated based on IFNγ coexpression, and IL-17A+IFNγ+ Th17 displayed an enhanced proinflammatory phenotype. Th17 cytokine plasticity and IFNγ production were critically dependent upon donor-derived IL-12p40, and cyclosporine (CsA) treatment regulated this differentiation pathway. This observation was highly concordant with clinical samples from allo-SCT recipients receiving CsA-based immune suppression where although the IFNγ-negative-Th17 subset predominated, IFNγ+-Th17 cells were also present. In sum, Th17 polarization and ensuing differentiation are mediated by sequential inflammatory signals, which are modulated by immunosuppressive therapy, leading to distinct phenotypes within this lineage.
Collapse
|
27
|
Arra A, Lingel H, Kuropka B, Pick J, Schnoeder T, Fischer T, Freund C, Pierau M, Brunner-Weinzierl MC. The differentiation and plasticity of Tc17 cells are regulated by CTLA-4-mediated effects on STATs. Oncoimmunology 2017; 6:e1273300. [PMID: 28344884 DOI: 10.1080/2162402x.2016.1273300] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/08/2016] [Accepted: 12/10/2016] [Indexed: 10/20/2022] Open
Abstract
As the blockade of inhibitory surface-molecules such as CTLA-4 on T cells has led to recent advances in antitumor immune therapy, there is great interest in identifying novel mechanisms of action of CD8+ T cells to evoke effective cytotoxic antitumor responses. Using in vitro and in vivo models, we investigated the molecular pathways underlying the CTLA-4-mediated differentiation of IL-17-producing CD8+ T cells (Tc17 cells) that strongly impairs cytotoxicity. Our studies demonstrate that Tc17 cells lacking CTLA-4 signaling have limited production of STAT3-target gene products such as IL-17, IL-21, IL-23R and RORγt. Upon re-stimulation with IL-12, these cells display fast downregulation of Tc17 hallmarks and acquire Tc1 characteristics such as IFNγ and TNF-α co-expression, which is known to correlate with tumor control. Indeed, upon adoptive transfer, these cells were highly efficient in the antigen-specific rejection of established OVA-expressing B16 melanoma in vivo. Mechanistically, in primary and re-stimulated Tc17 cells, STAT3 binding to the IL-17 promoter was strongly augmented by CTLA-4, associated with less binding of STAT5 and reduced relative activation of STAT1 which is known to block STAT3 activity. Inhibiting CTLA-4-induced STAT3 activity reverses enhancement of signature Tc17 gene products, rendering Tc17 cells susceptible to conversion to Tc1-like cells with enhanced cytotoxic potential. Thus, CTLA-4 critically shapes the characteristics of Tc17 cells by regulating relative STAT3 activation, which provides new perspectives to enhance cytotoxicity of antitumor responses.
Collapse
Affiliation(s)
- Aditya Arra
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Holger Lingel
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Benno Kuropka
- Institut für Chemie und Biochemie, Protein Biochemistry Group, Freie Universität, Berlin, Germany; Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Jonas Pick
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Tina Schnoeder
- Department of Hematology and Oncology, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Christian Freund
- Institut für Chemie und Biochemie, Protein Biochemistry Group, Freie Universität, Berlin, Germany; Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Mandy Pierau
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Monika C Brunner-Weinzierl
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| |
Collapse
|
28
|
Chellappa S, Hugenschmidt H, Hagness M, Subramani S, Melum E, Line PD, Labori KJ, Wiedswang G, Taskén K, Aandahl EM. CD8+ T Cells That Coexpress RORγt and T-bet Are Functionally Impaired and Expand in Patients with Distal Bile Duct Cancer. THE JOURNAL OF IMMUNOLOGY 2017; 198:1729-1739. [PMID: 28053236 DOI: 10.4049/jimmunol.1600061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 12/03/2016] [Indexed: 01/15/2023]
Abstract
CD8+ T cells that express retinoic acid-related orphan receptor (ROR)γt (TC17 cells) have been shown to promote procarcinogenic inflammation and contribute to a tolerogenic microenvironment in tumors. We investigated their phenotype and functional properties in relationship to the pathogenesis of human distal bile duct cancer (DBDC). DBDC patients had an elevated level of type 17 immune responses and the frequency of CD8+RORγt+ T cells (TC17 cells) was increased in peripheral blood. The CD8+RORγt+ T cells represented a highly activated subset and produced IL-17A in equal amount as CD4+RORγt+ T cells (TH17 cells). Most CD8+RORγt+ T cells coexpressed T-bet, a lineage transcription factor for TH1 and TC1 development, suggesting that CD8+RORγt+ T cells undergo plasticity toward a TC17/1-like phenotype with coproduction of IL-17A and INF-γ. In comparison with CD8+RORγt- T cells, the CD8+RORγt+ T cells had a higher level of TCR signaling and were terminally differentiated and exhausted. These cells also had impaired ability to re-express perforin after degranulation and reduced cytotoxic immune function. A subset of CD8+RORγt+ T cells expressing a low level of programmed cell death protein 1 and a high level of OX40 were associated with reduced patient survival. In conclusion, CD8+RORγt+ T cells are proinflammatory and functionally impaired and may contribute to the pathogenesis of DBDC.
Collapse
Affiliation(s)
- Stalin Chellappa
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway.,Biotechnology Center, University of Oslo, 0316 Oslo, Norway.,K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway.,K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, 0379 Oslo, Norway
| | - Harald Hugenschmidt
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway.,Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Morten Hagness
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway.,Biotechnology Center, University of Oslo, 0316 Oslo, Norway.,K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway.,Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Saranya Subramani
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Espen Melum
- K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway.,Norwegian Primary Sclerosing Cholangitis Research Center, Research Institute of Internal Medicine, Section of Gastroenterology, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, 0424 Oslo, Norway
| | - Pål Dag Line
- Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Knut-Jørgen Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Gro Wiedswang
- Department of Gastrointestinal Surgery, Oslo University Hospital, 0317 Oslo, Norway; and
| | - Kjetil Taskén
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway.,Biotechnology Center, University of Oslo, 0316 Oslo, Norway.,K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway.,K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, 0379 Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Einar Martin Aandahl
- Center for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway; .,Biotechnology Center, University of Oslo, 0316 Oslo, Norway.,K.G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway.,Section for Transplantation Surgery, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
29
|
Salehi Z, Doosti R, Beheshti M, Janzamin E, Sahraian MA, Izad M. Differential Frequency of CD8+ T Cell Subsets in Multiple Sclerosis Patients with Various Clinical Patterns. PLoS One 2016; 11:e0159565. [PMID: 27467597 PMCID: PMC4965085 DOI: 10.1371/journal.pone.0159565] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/04/2016] [Indexed: 12/05/2022] Open
Abstract
Recent evidence points to a pathogenic role for CD8+ cytotoxic T (Tc) cells in Multiple sclerosis (MS). Based on cytokine profile, Tc cells can be divided into different subsets: IFN-γ (Tc1), IL-4 (Tc2), IL-10 (Tc10), IL-17 (Tc17), IL-21 (Tc21), IL-22 (Tc22) and TNF-α producing cells. In this study we evaluated the frequency of Tc cell subsets and the serum level of Tc17 differentiation cytokines in MS patients with different clinical patterns. We analyzed Tc cell subsets percentage in peripheral blood of relapsing-remitting (RRMS) (n = 28), secondary-progressive (SPMS) (n = 10) and primary-progressive (PPMS) (n = 4) MS patients in comparison to healthy controls (n = 15) using flow cytometry. Serum level of TGF-β, IL-6 and IL-23 were measured by ELISA. We showed elevated levels of Tc1 and Tc17 cells in SPMS and RRMS patients in relapse phase, respectively (P = 0.04). Interestingly, the percentage of TNF-α producing CD8+ T cells in relapse and remission phase of RRMS and SPMS patients were higher than controls (P = 0.01, P = 0.004, P = 0.01, respectively) and Tc21 increased in remission phase of RRMS compared to SPMS (P = 0.03). We also found higher frequency of CD8+ IFN-γ+ TNF-α+ IL-17+ T cells in relapse phase of RRMS compared to remission phase, SPMS patients and controls (P = 0.01, P = 0.004 and P = 0.02, respectively). TGF- β increased in sera of RRMS patients in remission phase (P = 0.03) and SPMS (P = 0.05) compared to healthy subjects. Increased level of Tc17 and CD8+ IFN-γ+ TNF-α+ IL-17+ T cells in relapse phase highlights the critical role of IL-17 in RRMS pathogenesis.
Collapse
Affiliation(s)
- Zahra Salehi
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rozita Doosti
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Beheshti
- Pathophysiology laboratory, Sina hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Janzamin
- Flow Cytometry laboratory, Department of Stem Cell and Developmental Biology, Royan Institute, Tehran, Iran
| | - Mohammad Ali Sahraian
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail: (MI); (M-AS)
| | - Maryam Izad
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail: (MI); (M-AS)
| |
Collapse
|
30
|
Abstract
BACKGROUND Distinct CD8+ T-cell subsets such as interleukin-17-expressing Tc17 and Foxp3-expressing Tcreg are functionally similar to CD4+ T cells. Though CD4+ T cells are dysregulated in patients with inflammatory bowel disease (IBD), CD8+ T cells are not well investigated. Vitamin D is an environmental factor which influences T-cell subsets. We assessed the prevalence of CD8+ T-cell subsets among peripheral blood mononuclear cells (PBMC) and lamina propria mononuclear cells (LPMC) of patients with Crohn's disease, patients with ulcerative colitis, and healthy controls. We then tested the effect of 1α,25-dihydroxyvitamin D3 on CD8+ T-cell subsets. METHODS A total of 73 patients with Crohn's disease, 49 patients with ulcerative colitis, and 47 healthy controls were studied. LPMC or PBMC were isolated and flow cytometry was performed. CD3+ T cells, isolated from PBMC, were cultured with or without 1α,25-dihydroxyvitamin D3, before flow cytometry. RESULTS In LPMC, the prevalence of Tcreg was higher in patients with IBD (P < 0.05), whereas Tc17 were higher in patients with ulcerative colitis compared with patients with Crohn's disease and healthy controls (P < 0.05). In PBMC, both Tcreg and Tc17 were higher in patients with IBD (P < 0.01). Double-expressing interferon-γ+ interleukin-17+ and Foxp3+ interleukin-17+ CD8+ T cells were also identified indicating possible CD8+ plasticity. 1α,25-dihydroxyvitamin D3 decreased interferon-γ-expressing Tc1 (P < 0.05), but had no effect on Tc17 or Tcreg. CONCLUSIONS The prevalence of novel CD8+ T-cell subsets is altered in patients with IBD. Double-expressing cells indicate plasticity and were identified in patients with IBD. Vitamin D may have a limited effect on CD8+ T cells by decreasing interferon-γ expression.
Collapse
|
31
|
IL-17+ CD8+ T cells: Differentiation, phenotype and role in inflammatory disease. Immunol Lett 2016; 178:20-6. [PMID: 27173097 PMCID: PMC5046976 DOI: 10.1016/j.imlet.2016.05.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/06/2016] [Indexed: 01/01/2023]
Abstract
IL-17A (IL-17) is produced by multiple cell subsets, including CD8+ T cells. The presence of IL-17+ CD8+ T cells in human inflammatory diseases suggests these cells may contribute to immunopathology. Increased knowledge of human IL-17+ CD8+ T cells will enhance our overall understanding of their role in human disease.
The pro-inflammatory cytokine interleukin-17A (IL-17) has been the subject of research by many groups worldwide. IL-17 expression is often associated with a specific subset of CD4+ T cells (the so-called Th17 cells); however various other immune cell subsets can also synthesise and express IL-17, including CD8+ T cells. Here we review recent data regarding the presence of IL-17+ CD8+ T cells (also known as Tc17 cells) in human inflammatory disease, discuss current knowledge regarding the culture conditions required for the differentiation of these cells in humans and mice, and describe key phenotypic and functional features. Collectively, this information may shed light on the potential pathogenic role that IL-17+ CD8+ T cells may play in human inflammatory disease.
Collapse
|
32
|
Lau MC, Keith P, Costello ME, Bradbury LA, Hollis KA, Thomas R, Thomas GP, Brown MA, Kenna TJ. Genetic association of ankylosing spondylitis with TBX21 influences T-bet and pro-inflammatory cytokine expression in humans and SKG mice as a model of spondyloarthritis. Ann Rheum Dis 2016; 76:261-269. [PMID: 27125523 DOI: 10.1136/annrheumdis-2015-208677] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 04/01/2016] [Accepted: 04/09/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Ankylosing spondylitis (AS) is a highly heritable immune-mediated arthropathy. Inflammation in AS is poorly understood. TBX21 encodes T-bet, a transcription factor, lying within a locus with genome-wide significant association with AS. T-bet is implicated in innate and adaptive immunity. However, the role of T-bet in AS pathogenesis is unclear. METHODS We assessed the importance of T-bet in disease development and progression in peripheral blood mononuclear cells from 172 AS cases and 83 healthy controls carrying either risk or protective alleles of the peak AS-associated TBX21 single nucleotide polymorphism. Kinetics and localisation of T-bet expression in the SKG mouse model of spondyloarthropathy was examined, along with the impact of Tbx21 knockout on arthritis development in SKG mice. RESULTS Patients with AS had higher T-bet expression than healthy individuals, driven predominantly by natural killer and CD8+ T cells, with expression levels in CD8+ T cells completely distinguishing AS cases from healthy controls. T-bet expression was increased in AS cases carrying risk compared with protective alleles of rs11657479. In curdlan-treated SKG mice, T-bet expression increased early after disease initiation and persisted throughout the course of disease. There was marked reduction in gut and peripheral joint inflammation, and less IFNγ-producing and IL-17-producing CD8+ T cells, in Tbx21-/- compared with wild-type SKG mice. CONCLUSIONS AS-associated variants in TBX21 influence T-bet expression. T-bet+ innate and adaptive immune cells have altered IL-17 and IFNγ, and early activation marker CD69 expression than T-bet cells. This indicates that T-bet is a major component of inflammatory pathways of spondyloarthropathy in humans and mice.
Collapse
Affiliation(s)
- Max C Lau
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Patricia Keith
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Mary-Ellen Costello
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Linda A Bradbury
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kelly A Hollis
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Gethin P Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Matthew A Brown
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Tony J Kenna
- Queensland University of Technology, Institute for Health and Biomedical Innovation, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Chen HW, Tsai JP, Yao TY, Hsieh CL, Chen IH, Liu SJ. TGF-β and IL-21 cooperatively stimulate activated CD8(+) T cells to differentiate into Tc17 cells. Immunol Lett 2016; 174:23-7. [PMID: 27085379 DOI: 10.1016/j.imlet.2016.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/22/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022]
Abstract
TGF-β together with IL-21 or IL-6 can drive the differentiation of naïve CD8(+) T cells into IL-17-producing CD8(+) T cells. These IL-17-producing CD8(+) T cells are termed Tc17 cells. Tc17 cells preserve plasticity under various conditions in vitro and in vivo. IFN-γ-producing CD8(+) T cells are termed Tc1 cells. However, Tc1 cells are considered relatively stable. In the present study, we show that the combination of TGF-β plus IL-21, but not IL-6, converts Tc1 cells into Tc17 cells; this conversion is associated with elevated RORα, RORγt, and Batf mRNA levels. These results indicate that Tc1 cells are skewed to the Tc17 cell phenotype under TGF-β plus IL-21-polarizing conditions. Furthermore, IL-6R is expressed on naïve, but not activated, CD8(+) T cells. In contrast, IL-21R is expressed on both naïve and activated CD8(+) T cells. Thus, differential expression profiles of IL-6R and IL-21R on naïve and activated CD8(+) T cells may be one mechanism by which TGF-β plus IL-21, but not IL-6, can drive activated CD8(+) T cells to differentiate into IL-17-producing cells. Taken together, these results provide a novel viewpoint for the plasticity of Tc1 cells.
Collapse
Affiliation(s)
- Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC; Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, ROC.
| | - Jy-Ping Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC
| | - Tsung-You Yao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC
| | - Chia-Ling Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC
| | - I-Hua Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC
| | - Shin-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli 350, Taiwan, ROC; Graduate Institute of Immunology, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
34
|
Durham LE, Kirkham BW, Taams LS. Contribution of the IL-17 Pathway to Psoriasis and Psoriatic Arthritis. Curr Rheumatol Rep 2016. [PMID: 26209291 DOI: 10.1007/s11926-015-0529-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Investigators have accrued compelling evidence that the IL-17 pathway is central to the pathogenesis of psoriasis and psoriatic arthritis. The evidence comprises genome-wide association studies (GWAS), data from experimental murine models and findings from in vitro studies on patients' cells or tissue biopsies. More recently, the success of drugs blocking the IL-17 pathway in treating both psoriasis (PsO) and psoriatic arthritis (PsA) confirms that IL-17 is a clinically relevant therapeutic target. However, there remain many unanswered questions: is PsA simply an extension of PsO from the skin to the synovial tissue or are there differences in the underlying pathogenesis of these diseases? Which cell type represents the primary source of IL-17 in PsO and PsA? And how are these cells regulated? This review outlines the IL-17 pathway, summarises the evidence supporting its role in PsO and PsA and discusses recent data that may help to address these yet unresolved questions.
Collapse
Affiliation(s)
- L E Durham
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), Division of Immunology, Infection and Inflammatory Disease, King's College London, London, SE1 1UL, UK,
| | | | | |
Collapse
|
35
|
Yousefi M, Movassaghpour AA, Shamsasenjan K, Ghalamfarsa G, Sadreddini S, Jadidi-Niaragh F, Hojjat-Farsangi M. The skewed balance between Tregs and Th17 in chronic lymphocytic leukemia. Future Oncol 2016; 11:1567-82. [PMID: 25963433 DOI: 10.2217/fon.14.298] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
While Tregs maintain self-tolerance and inhibit antitumor responses, T helper (Th)17 cells may enhance inflammatory and antitumor responses. The balance between these two important T-cell subsets has been skewed in many immunopathologic conditions such as autoimmune and cancer diseases. B-cell chronic lymphocytic leukemia (CLL) is the most common form of leukemia in the western world and is characterized with monoclonal expansion of B lymphocytes. There is evidence which implies that the progression of CLL is associated with expansion of Treg and downregulation of Th17 cells. In this review, we will discuss about immunobiology of Treg and Th17 cells and their role in immunopathogenesis of CLL as well as their reciprocal changes during disease progression.
Collapse
Affiliation(s)
- Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | | | |
Collapse
|
36
|
Tc17 cells are a proinflammatory, plastic lineage of pathogenic CD8+ T cells that induce GVHD without antileukemic effects. Blood 2015. [DOI: 10.1182/blood-2015-01-622662] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Key Points
Donor-derived Tc17 cells differentiate early after allogeneic transplant in response to IL-6 and alloantigen presentation by host DCs. Tc17 are highly proinflammatory and pathogenic posttransplant, but exert limited or no GVL activity.
Collapse
|
37
|
Larochelle C, Lécuyer MA, Alvarez JI, Charabati M, Saint-Laurent O, Ghannam S, Kebir H, Flanagan K, Yednock T, Duquette P, Arbour N, Prat A. Melanoma cell adhesion molecule-positive CD8 T lymphocytes mediate central nervous system inflammation. Ann Neurol 2015; 78:39-53. [PMID: 25869475 DOI: 10.1002/ana.24415] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 03/23/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Although Tc17 lymphocytes are enriched in the central nervous system (CNS) of multiple sclerosis (MS) subjects and of experimental autoimmune encephalomyelitis (EAE) animals, limited information is available about their recruitment into the CNS and their role in neuroinflammation. Identification of adhesion molecules used by autoaggressive CD8(+) T lymphocytes to enter the CNS would allow further characterization of this pathogenic subset and could provide new therapeutic targets in MS. We propose that melanoma cell adhesion molecule (MCAM) is a surface marker and adhesion molecule used by pathogenic CD8(+) T lymphocytes to access the CNS. METHODS Frequency, phenotype, and function of MCAM(+) CD8(+) T lymphocytes was characterized using a combination of ex vivo, in vitro, in situ, and in vivo approaches in humans and mice, including healthy controls, MS subjects, and EAE animals. RESULTS Herein, we report that MCAM is expressed by human effector CD8(+) T lymphocytes and it is strikingly upregulated during MS relapses. We further demonstrate that MCAM(+) CD8(+) T lymphocytes express more interleukin 17, interferon γ, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor than MCAM(-) lymphocytes, and exhibit an enhanced killing capacity toward oligodendrocytes. MCAM blockade restricts the transmigration of CD8(+) T lymphocytes across human blood-brain barrier endothelial cells in vitro, and blocking or depleting MCAM in vivo reduces chronic neurological deficits in active, transfer, and spontaneous progressive EAE models. INTERPRETATION Our data demonstrate that MCAM identifies encephalitogenic CD8(+) T lymphocytes, suggesting that MCAM could represent a biomarker of MS disease activity and a valid target for the treatment of neuroinflammatory conditions.
Collapse
Affiliation(s)
- Catherine Larochelle
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM)-Notre Dame Hospital, Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montreal, Montreal, Quebec, Canada
| | - Marc-André Lécuyer
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Jorge Ivan Alvarez
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marc Charabati
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Olivia Saint-Laurent
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Soufiane Ghannam
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Hania Kebir
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Ken Flanagan
- Prothena Biosciences, South San Francisco, CA, USA
| | - Ted Yednock
- Prothena Biosciences, South San Francisco, CA, USA
| | - Pierre Duquette
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM)-Notre Dame Hospital, Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montreal, Montreal, Quebec, Canada
| | - Nathalie Arbour
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montreal, Montreal, Quebec, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM)-Notre Dame Hospital, Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montreal, Montreal, Quebec, Canada
| |
Collapse
|
38
|
SUMO2 overexpression enhances the generation and function of interleukin-17-producing CD8⁺ T cells in mice. Cell Signal 2015; 27:1246-52. [PMID: 25762490 DOI: 10.1016/j.cellsig.2015.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
Abstract
Small ubiquitin-like modifier (SUMO) 2 is a small protein that controls the activity and stability of other proteins by SUMOylation. In this study, T cell-specific SUMO2 overexpressing transgenic mice were generated to study the effect of SUMO2 on T lymphocytes. SUMO2 overexpression promoted differentiation of interleukin (IL)-17-producing CD8(+) T cells, and significantly suppressed the growth of EL4 tumor cells in vivo. Moreover, the tumor tissue from SUMO2-overexpressing mice had higher interferon (IFN)-γ and granzyme B mRNA levels. Although SUMO2 overexpression did not increase IFN-γ or granzyme B production in cytotoxic T lymphocytes, IL-12 treatment restored and increased IFN-γ secretion in IL-17-producing CD8(+) T cells. SUMO2 overexpression also increased gene expression of chemokines, CCL4, and CXCL10, which attract cytotoxic T lymphocytes to tumor tissues. Additionally, SUMO2-overexpressing T cells exhibited increased STAT3 phosphorylation, implying a SUMO2 target which up-regulates STAT3 activity governing IL-17A-producing CD8(+) T cell differentiation and antitumor immune responses.
Collapse
|
39
|
Liang Y, Pan HF, Ye DQ. IL-17A-producing CD8(+)T cells as therapeutic targets in autoimmunity. Expert Opin Ther Targets 2015; 19:651-61. [PMID: 25611933 DOI: 10.1517/14728222.2014.997710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The involvement of IL-17-producing CD8(+)T cells (TC17) in various conditions, such as infection, cancer and autoimmune inflammation, has been documented in both humans and mice; however, TC17 cells have received only marginal attention. AREAS COVERED Here, we provide an overview of the cytokines, chemokines, and cytokine and chemokine receptors that characterize the murine and human TC17 cell phenotype. We also discuss signaling pathways, molecular interactions, and transcriptional and epigenetic events that contribute to TC17 differentiation and acquisition of effector functions. Heterogeneity and inherent phenotypic instability of TC17 cells were shown both in humans and murine models. Aberrant expression of TC17 cells was observed in many autoimmune conditions. Moreover, functional analysis demonstrated in vivo plasticity of TC17 cells may be a key feature of TC17 cell biology in autoimmune diseases. EXPERT OPINION Due to its important roles in inflammation and autoimmunity, TC17 cell pathway may have promise as a potential therapeutic target for autoimmune diseases. The strategies include the suppression of TC17 cell generation and migration and the blockade of TC17 cell instability and heterogeneity. TMP778 may open an avenue to novel therapeutic strategies.
Collapse
Affiliation(s)
- Yan Liang
- Anhui Medical University, School of Public Health, Department of Epidemiology and Biostatistics , 81 Meishan Road, Hefei, Anhui, 230032 , PR China . +86 551 65167726 ; +86 551 65161171 ;
| | | | | |
Collapse
|
40
|
Abstract
Tc17 cells-a subset of CD8(+)T cells-have recently been identified that are characterized by the production of interleukin (IL)-17. Cytokines IL-6 and transforming growth factor-beta 1 (TGF-β1) and transcription factors signaling transducers and activators of transcription (STAT)3, retinoic acid receptor-related orphan nuclear receptor gamma (RORγt), and interferon regulatory factor (IRF)4 are necessary for differentiation of Tc17 cells, controlling expression of molecules essential for Tc17 cell trafficking and function. Current human researches have determined the significance of CD161 expression as either a marker of Tc17 cells or as an effector and regulator of Tc17 cell function. Noncytotoxic Tc17 cells possess a high plasticity to convert into IFN-γ producing cells, which exhibit strong cytotoxic activity. The importance of in vivo plasticity of Tc17 cells for the induction of autoimmune diseases has been demonstrated and Tc17 cells potentially represent novel therapeutic targets in autoimmune diseases. The involvement of interleukin (IL)-17-producing CD8(+)T cells (Tc17) in various conditions, such as infection, cancer, and autoimmune inflammation, has been documented in both humans and mice; however, Tc17 cells have received only marginal attention. Here, we provide an overview of the cytokines and chemokines that characterize the murine and human Tc17 cells. Moreover, we discuss signaling pathways, molecular interactions, and transcriptional events that contribute to Tc17 differentiation and acquisition of effector functions. Also considered is the basis of Tc17 cell plasticity toward the Tc1 lineage, and we suggest that in vivo plasticity of Tc17 cells may be a key feature of Tc17 cell biology in autoimmune diseases. Furthermore, current human researches have revealed that Tc17 cells are different than that in mice because all of them express CD161 and exclusively originate from CD161 precursors present in umbilical cord blood. Finally, we focus on the recent evidence for long-lived Tc17 memory cell populations in mouse models and humans, and their functional roles in mediating disease memory. Hopefully, the information obtained will benefit for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yan Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University , Hefei, Anhui , China
| | | | | |
Collapse
|
41
|
Keenan BP, Saenger Y, Kafrouni MI, Leubner A, Lauer P, Maitra A, Rucki AA, Gunderson AJ, Coussens LM, Brockstedt DG, Dubensky TW, Hassan R, Armstrong TD, Jaffee EM. A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice. Gastroenterology 2014; 146:1784-94.e6. [PMID: 24607504 PMCID: PMC4035450 DOI: 10.1053/j.gastro.2014.02.055] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/09/2014] [Accepted: 02/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Premalignant lesions and early stage tumors contain immunosuppressive microenvironments that create barriers for cancer vaccines. Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which express an activated form of Kras in pancreatic tissues, develop pancreatic intraepithelial neoplasms (PanIN) that progress to pancreatic ductal adenocarcinoma (PDA). We used these mice to study immune suppression in PDA. METHODS We immunized KPC and Kras(G12D/+);Pdx-1-Cre mice with attenuated intracellular Listeria monocytogenes (which induces CD4(+) and CD8(+) T-cell immunity) engineered to express Kras(G12D) (LM-Kras). The vaccine was given alone or in sequence with an anti-CD25 antibody (PC61) and cyclophosphamide to deplete T-regulatory (Treg) cells. Survival times were measured; pancreatic and spleen tissues were collected and analyzed by histologic, flow cytometry, and immunohistochemical analyses. RESULTS Interferon γ-mediated, CD8(+) T-cell responses were observed in KPC and Kras(G12D/+);Pdx-1-Cre mice given LM-Kras, but not in unvaccinated mice. Administration of LM-Kras to KPC mice 4-6 weeks old (with early stage PanINs), depleted of Treg cells, significantly prolonged survival and reduced PanIN progression (median survival, 265 days), compared with unvaccinated mice (median survival, 150 days; P = .002), mice given only LM-Kras (median survival, 150 days; P = .050), and unvaccinated mice depleted of Treg cells (median survival, 170 days; P = .048). In 8- to 12-week-old mice (with late-stage PanINs), LM-Kras, alone or in combination with Treg cell depletion, did not increase survival time or slow PanIN progression. The combination of LM-Kras and Treg cell depletion reduced numbers of Foxp3(+)CD4(+) T cells in pancreatic lymph nodes, increased numbers of CD4(+) T cells that secrete interleukin 17 and interferon γ, and caused CD11b(+)Gr1(+) cells in the pancreas to acquire an immunostimulatory phenotype. CONCLUSIONS Immunization of KPC mice with Listeria monocytogenes engineered to express Kras(G12D), along with depletion of Treg cells, reduces progression of early stage, but not late-stage, PanINs. This approach increases infiltration of the lesion with inflammatory cells. It might be possible to design immunotherapies against premalignant pancreatic lesions to slow or prevent progression to PDA.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- CD11b Antigen/metabolism
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Carcinoma in Situ/drug therapy
- Carcinoma in Situ/genetics
- Carcinoma in Situ/immunology
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cyclophosphamide/pharmacology
- Disease Models, Animal
- Disease Progression
- Forkhead Transcription Factors/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Inflammation Mediators/metabolism
- Integrases/genetics
- Integrases/metabolism
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/metabolism
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- Receptors, Chemokine/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Bridget P Keenan
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland; Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yvonne Saenger
- Division of Hematology and Oncology, Tisch Cancer Institute and Department of Dermatology, Mount Sinai School of Medicine, New York, New York
| | - Michel I Kafrouni
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Ashley Leubner
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | | | - Anirban Maitra
- Department of Pathology and Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Agnieszka A Rucki
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Andrew J Gunderson
- Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon
| | - Lisa M Coussens
- Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon
| | | | | | - Raffit Hassan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Todd D Armstrong
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Elizabeth M Jaffee
- The Sidney Kimmel Comprehensive Cancer Center, the Skip Viragh Center for Clinical Pancreatic Cancer Research, and the Sol Goldman Pancreatic Cancer Center at Johns Hopkins, Baltimore, Maryland.
| |
Collapse
|
42
|
Pick J, Arra A, Lingel H, Hegel JK, Huber M, Nishanth G, Jorch G, Fischer KD, Schlüter D, Tedford K, Brunner-Weinzierl MC. CTLA-4 (CD152) enhances the Tc17 differentiation program. Eur J Immunol 2014; 44:2139-52. [PMID: 24723371 DOI: 10.1002/eji.201343497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 02/21/2014] [Accepted: 03/28/2014] [Indexed: 01/24/2023]
Abstract
Although CD8(+) T cells that produce IL-17 (Tc17 cells) have been linked to host defense, Tc17 cells show reduced cytotoxic activity, which is the characteristic function of CD8(+) T cells. Here, we show that CTLA-4 enhances the frequency of IL-17 in CD8(+) T cells, indicating that CTLA-4 (CD152) specifically promotes Tc17 differentiation. Simultaneous stimulation of CTLA-4(+/+) and CTLA-4(-/-) T cells in cocultures and agonistic CTLA-4 stimulation unambiguously revealed a cell-intrinsic mechanism for IL-17 control by CTLA-4. The quality of CTLA-4-induced Tc17 cells was tested in vivo, utilizing infection with the facultative intracellular bacterium Listeria monocytogenes (LM). Unlike CTLA-4(+/+) Tc17 cells, CTLA-4(-/-) were nearly as efficient as Tc1 CTLA-4(+/+) cells in LM clearance. Additionally, adoptively transferred CTLA-4(-/-) Tc17 cells expressed granzyme B after rechallenge, and produced Tc1 cytokines such as IFN-γ and TNF-α, which strongly correlate with bacterial clearance. CTLA-4(+/+) Tc17 cells demonstrated a high-quality Tc17 differentiation program ex vivo, which was also evident in isolated IL-17-secreting Tc17 cells, with CTLA-4-mediated enhanced upregulation of Tc17-related molecules such as IL-17A, RORγt, and IRF-4. Our results show that CTLA-4 promotes Tc17 differentiation that results in robust Tc17 responses. Its inactivation might therefore represent a central therapeutic target to enhance clearance of infection.
Collapse
Affiliation(s)
- Jonas Pick
- Department of Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ryba-Stanisławowska M, Stanisławowski M, Myśliwska J. Effector and regulatory T cell subsets in diabetes-associated inflammation. Is there a connection with ST2/IL-33 axis? Perspective. Autoimmunity 2014; 47:361-71. [PMID: 24547981 DOI: 10.3109/08916934.2014.886198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Type 1 diabetes (DM1) is a chronic inflammatory disease, which when progresses leads to the development of late vascular complications. The disease involves impairments in regulatory and effector subsets of T lymphocytes, which suppress and maintain inflammatory response, respectively. ST2/IL-33 pathway is involved in T-cell-mediated immune response and might regulate the inflammatory process in several diseases. This review presents the latest research findings regarding effector and regulatory T cell subsets in the context of inflammation accompanying DM1 with particular focus on the ST2/IL-33 network and its possible association with T cell-mediated immunity.
Collapse
|
44
|
Changes of Th17/Tc17 and Th17/Treg cells in endometrial carcinoma. Gynecol Oncol 2014; 132:599-605. [PMID: 24388919 DOI: 10.1016/j.ygyno.2013.12.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/04/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVES T helper 17 (Th17), T cytotoxic 17 (Tc17) and regulatory T (Treg) cells are important factors in the pathogenesis of inflammatory and autoimmune diseases. However, information concerning the roles of these cells in antitumor immunity or endometrial tumorigenesis remains limited. In this study, we aimed to describe the distribution of Th17, Tc17 and Treg cells in endometrial carcinoma patients, and elucidate the probable role of these effector T cells. METHODS We assessed the expression of interleukin (IL)-17 and Foxp3 in the peripheral blood of endometrial carcinoma patients and healthy controls by flow cytometry to determine the relative numbers of Th17, Tc17 and Treg cells. Th17 cells and Tc17 cells were counted as percentages of the total number of CD3(+) T cells; Treg cells were counted as a percentage of the total number of CD4(+) T cells. We also evaluated Th17 and Tc17 cells in tumor tissue by immunohistochemical staining. IL-17 and IL-10, dominant products of these three cell types, were detected by using enzyme-linked immunosorbent assays. RESULTS The frequencies of Th17, Tc17 and Treg cells, as well as the serum level of IL-10, were significantly elevated in endometrial carcinoma patients compared to healthy controls. The Th17/Tc17 and Th17/Treg ratios were also observed to change significantly. However, there was no significant difference on the IL-17 levels in the serum. Additionally, immunohistochemistry performed on tumor tissues indicated that the amounts of Th17 and Tc17 increased in the cancer patients. CONCLUSIONS Our data suggests a probable involvement of Th17, Tc17 and Treg cells in the pathogenesis of endometrial carcinoma. Restoring the balance of these cells may help with the research and development of immunotherapies for endometrial carcinoma.
Collapse
|
45
|
Terhune J, Berk E, Czerniecki BJ. Dendritic Cell-Induced Th1 and Th17 Cell Differentiation for Cancer Therapy. Vaccines (Basel) 2013; 1:527-49. [PMID: 26344346 PMCID: PMC4494209 DOI: 10.3390/vaccines1040527] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/18/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023] Open
Abstract
The success of cellular immunotherapies against cancer requires the generation of activated CD4+ and CD8+ T-cells. The type of T-cell response generated (e.g., Th1 or Th2) will determine the efficacy of the therapy, and it is generally assumed that a type-1 response is needed for optimal cancer treatment. IL-17 producing T-cells (Th17/Tc17) play an important role in autoimmune diseases, but their function in cancer is more controversial. While some studies have shown a pro-cancerous role for IL-17, other studies have shown an anti-tumor function. The induction of polarized T-cell responses can be regulated by dendritic cells (DCs). DCs are key regulators of the immune system with the ability to affect both innate and adaptive immune responses. These properties have led many researchers to study the use of ex vivo manipulated DCs for the treatment of various diseases, such as cancer and autoimmune diseases. While Th1/Tc1 cells are traditionally used for their potent anti-tumor responses, mounting evidence suggests Th17/Tc17 cells should be utilized by themselves or for the induction of optimal Th1 responses. It is therefore important to understand the factors involved in the induction of both type-1 and type-17 T-cell responses by DCs.
Collapse
Affiliation(s)
- Julia Terhune
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Erik Berk
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Brian J Czerniecki
- Department of Surgery and Harrison Department of Surgical Research, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Rena Rowan Breast Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Yu CR, Kim SH, Mahdi RM, Egwuagu CE. SOCS3 deletion in T lymphocytes suppresses development of chronic ocular inflammation via upregulation of CTLA-4 and expansion of regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:5036-43. [PMID: 24101549 DOI: 10.4049/jimmunol.1301132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Suppressors of cytokine signaling (SOCS) proteins are negative-feedback regulators of the JAK/STAT pathway, and SOCS3 contributes to host immunity by regulating the intensity and duration of cytokine signals and inflammatory responses. Mice with Socs3 deletion in myeloid cells exhibit enhanced STAT3 signaling, expansion of Th1 and Th17 cells, and develop severe experimental autoimmune encephalomyelitis. Interestingly, development of the unique IL-17/IFN-γ double-producing (Th17/IFN-γ and Tc17/IFN-γ) subsets that exhibit strong cytotoxic activities and are associated with pathogenesis of several autoimmune diseases has recently been shown to depend on epigenetic suppression of SOCS3 expression, further suggesting involvement of SOCS3 in autoimmunity and tumor immunity. In this study, we generated mice with Socs3 deletion in the CD4 T cell compartment (CD4-SOCS3 knockout [KO]) to determine in vivo effects of the loss of Socs3 in the T cell-mediated autoimmune disease, experimental autoimmune uveitis (EAU). In contrast to the exacerbation of experimental autoimmune encephalomyelitis in myeloid-specific SOCS3-deleted mice, CD4-SOCS3KO mice were protected from acute and chronic uveitis. Protection from EAU correlated with enhanced expression of CTLA-4 and expansion of IL-10-producing regulatory T cells with augmented suppressive activities. We further show that SOCS3 interacts with CTLA-4 and negatively regulates CTLA-4 levels in T cells, providing a mechanistic explanation for the expansion of regulatory T cells in CD4-SOCS3 during EAU. Contrary to in vitro epigenetic studies, Th17/IFN-γ and Tc17/IFN-γ populations were markedly reduced in CD4-SOCS3KO, suggesting that SOCS3 promotes expansion of the Th17/IFN-γ subset associated with development of severe uveitis. Thus, SOCS3 is a potential therapeutic target in uveitis and other autoinflammatory diseases.
Collapse
Affiliation(s)
- Cheng-Rong Yu
- Molecular Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | | |
Collapse
|
47
|
Wu B, Zou Q, Hu Y, Wang B. Interleukin-22 as a molecular adjuvant facilitates IL-17-producing CD8+ T cell responses against a HBV DNA vaccine in mice. Hum Vaccin Immunother 2013; 9:2133-41. [PMID: 23941891 PMCID: PMC3906397 DOI: 10.4161/hv.26047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/19/2013] [Accepted: 08/05/2013] [Indexed: 01/06/2023] Open
Abstract
Interleukin-22 (IL-22) is mainly produced by activated Th1 cells, Th17 cells and NK cells and promotes anti-microbial defense, pro-inflammatory and tissue remodeling responses. However, its potential use as a vaccine adjuvant has not been tested. In this study, we tested if a DNA construct expressing IL-22 (pVAX-IL-22) could be used as a molecular adjuvant to enhance host immune responses induced by HBV DNA vaccination (pcD-S2). After immunizing mice with pcD-S2 combined with pVAX-IL-22, we didn't find enhancement of HBsAg-specific antibody responses in comparison to mice immunized with pcD-S2 alone. However, there was an enhancement of the level of IL-17 expression in antigen specific CD8(+) cytotoxic T lymphocytes (Tc17). By using CD8 T-cell knockout (KO) and IL-17 KO mice, Tc17 cells were found to be a dominant population driving cytotoxicity. Importantly, there was a correlation between pVAX-IL-22 enhancement of T lymphocytes and a reduction of HBsAg-positive hepatocytes in HBsAg transgenic mice. These results demonstrate that IL-22 might be used as an effective adjuvant to enhance cellular immune responses during HBsAg DNA vaccination since it can induce Tc17 cells to break tolerance in HBsAg transgenic mice.
Collapse
Affiliation(s)
- Bing Wu
- State Key Laboratory for Agro-Biotechnology; College of Biological Science; China Agricultural University; Beijing, P.R. China
| | - Qiang Zou
- State Key Laboratory for Agro-Biotechnology; College of Biological Science; China Agricultural University; Beijing, P.R. China
| | - Yanxin Hu
- College of Veterinary Medicine; China Agricultural University; Beijing, P.R. China
| | - Bin Wang
- State Key Laboratory for Agro-Biotechnology; College of Biological Science; China Agricultural University; Beijing, P.R. China
- Key Laboratory of Medical Molecular Virology of MOH and MOE; Fudan University Shanghai Medical College; Shanghai, P.R. China
| |
Collapse
|
48
|
Kumawat AK, Strid H, Tysk C, Bohr J, Hörnquist EH. Microscopic colitis patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile. Mol Immunol 2013; 55:355-64. [PMID: 23566938 DOI: 10.1016/j.molimm.2013.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Microscopic colitis (MC) is a chronic inflammatory bowel disorder of unknown aetiology comprising collagenous colitis (CC) and lymphocytic colitis (LC). Data on the local cytokine profile in MC is limited. This study investigated the T helper (Th) cell and cytotoxic T lymphocyte (CTL) mucosal cytokine profile at messenger and protein levels in MC patients. METHODS Mucosal biopsies from CC (n=10), LC (n=5), and CC or LC patients in histopathological remission (CC-HR, n=4), (LC-HR, n=6), ulcerative colitis (UC, n=3) and controls (n=10) were analysed by real-time PCR and Luminex for expression/production of IL-1β, -4, -5, -6, -10, -12, -17, -21, -22, -23, IFN-γ, TNF-α, T-bet and RORC2. RESULTS Mucosal mRNA but not protein levels of IFN-γ and IL-12 were significantly up regulated in CC, LC as well as UC patients compared to controls. Transcription of the Th1 transcription factor T-bet was significantly enhanced in CC but not LC patients. mRNA levels for IL-17A, IL-21, IL-22 and IL-6 were significantly up regulated in CC and LC patients compared to controls, albeit less than in UC patients. Significantly enhanced IL-21 protein levels were noted in both CC and LC patients. IL-6 protein and IL-1β mRNA levels were increased in CC and UC but not LC patients. Increased mucosal mRNA levels of IFN-γ, IL-21 and IL-22 were correlated with higher clinical activity, recorded as the number of bowel movements per day, in MC patients. Although at lower magnitude, IL-23A mRNA was upregulated in CC and LC, whereas TNF-α protein was increased in CC, LC as well as in UC patients. Neither mRNA nor protein levels of IL-4, IL-5 or IL-10 were significantly changed in any of the colitis groups. LC-HR and especially CC-HR patients had normalized mRNA and protein levels of the above cytokines compared to LC and CC patients. No significant differences were found between LC and CC in cytokine expression/production. CONCLUSION LC and CC patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Case-Control Studies
- Colitis, Collagenous/genetics
- Colitis, Collagenous/immunology
- Colitis, Collagenous/pathology
- Colitis, Lymphocytic/genetics
- Colitis, Lymphocytic/immunology
- Colitis, Lymphocytic/pathology
- Colitis, Microscopic/genetics
- Colitis, Microscopic/immunology
- Colitis, Microscopic/pathology
- Cytokines/biosynthesis
- Cytokines/genetics
- Female
- Humans
- Immunity, Mucosal/genetics
- Male
- Middle Aged
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Th1 Cells/immunology
- Th1 Cells/pathology
- Th17 Cells/immunology
- Th17 Cells/pathology
- Young Adult
Collapse
|
49
|
Rubino SJ, Geddes K, Magalhaes JG, Streutker C, Philpott DJ, Girardin SE. Constitutive induction of intestinal Tc17 cells in the absence of hematopoietic cell-specific MHC class II expression. Eur J Immunol 2013; 43:2896-906. [PMID: 23881368 DOI: 10.1002/eji.201243028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 06/13/2013] [Accepted: 07/19/2013] [Indexed: 12/30/2022]
Abstract
The enteric pathogen Citrobacter rodentium induces a mucosal IL-17 response in CD4(+) T helper (Th17) cells that is dependent on the Nod-like receptors Nod1 and Nod2. Here, we sought to determine whether this early Th17 response required antigen presentation by major histocompatibility complex class II (MHCII) for full induction. At early phases of C. rodentium infection, we observed that the intestinal mucosal Th17 response was fully blunted in irradiated mice reconstituted with MHCII-deficient (MHCII(-/-) →WT) hematopoietic cells. Surprisingly, we also observed a substantial increase in the relative frequency of IL-17(+) CD8(+) CD4(-) TCR-β(+) cells (Tc17 cells) and FOXP3(+) CD8(+) CD4(-) TCR-β(+) cells in the lamina propria and intraepithelial lymphocyte compartment of MHCII(-/-) →WT mice compared with that in WT→WT counterparts. Moreover, MHCII(-/-) →WT mice displayed increased susceptibility, increased bacterial translocation to deeper organs, and more severe colonic histopathology after infection with C. rodentium. Finally, a similar phenotype was observed in mice deficient for CIITA, a transcriptional regulator of MHCII expression. Together, these results indicate that MHCII is required to mount early mucosal Th17 responses to an enteric pathogen, and that MHCII regulates the induction of atypical CD8(+) T-cell subsets, such as Tc17 cells and FOXP3(+) CD8(+) cells, in vivo.
Collapse
Affiliation(s)
- Stephen J Rubino
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Expansion of Memory-Type CD8+ T Cells Correlates With the Failure of Early Immunosuppression Withdrawal After Cadaver Liver Transplantation Using High-Dose ATG Induction and Rapamycin. Transplantation 2013; 96:306-15. [DOI: 10.1097/tp.0b013e3182985414] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|