1
|
Kuo JF, Wu HY, Tung CW, Huang WH, Lin CS, Wang CC. Induction of Thymus Atrophy and Disruption of Thymocyte Development by Fipronil through Dysregulation of IL-7-Associated Genes. Chem Res Toxicol 2024; 37:1488-1500. [PMID: 39141674 PMCID: PMC11409377 DOI: 10.1021/acs.chemrestox.4c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
The susceptibility of the immune system to immunotoxic chemicals is evident, particularly in the thymus, a vital primary immune organ prone to atrophy due to exposure to toxicants. Fipronil (FPN), a widely used insecticide, is of concern due to its potential neurotoxicity, hepatotoxicity, and immunotoxicity. Our previous study showed that FPN disturbed the antigen-specific T-cell functionality in vivo. As T-cell lineage commitment and thymopoiesis are closely interconnected with the normal function of the T-cell-mediated immune responses, this study aims to further examine the toxic effects of FPN on thymocyte development. In this study, 4-week-old BALB/c mice received seven doses of FPN (1, 5, 10 mg/kg) by gavage. Thymus size, medulla/cortex ratio, total thymocyte counts, double-positive thymocyte population, and IL-7-positive cells decreased dose-dependently. IL-7 aids the differentiation of early T-cell precursors into mature T cells, and several essential genes contribute to the maturation of T cells in the thymus. Foxn1 ensures that the thymic microenvironment is suitable for the maturation of T-cell precursors. Lyl1 is involved in specifying lymphoid cells and maintaining T-cell development in the thymus. The c-Kit/SCF collaboration fosters a supportive thymic milieu to promote the formation of functional T cells. The expression of IL-7, IL-7R, c-Kit, SCF, Foxn1, and Lyl1 genes in the thymus was significantly diminished in FPN-treated groups with the concordance with the reduction of IL-7 signaling proteins (IL-7, IL-7R, c-KIT, SCF, LYL1, FOXO3A, and GABPA), suggesting that the dysregulation of T-cell lineage-related genes may contribute to the thymic atrophy induced by FPN. In addition, FPN disturbed the functionality of thymocytes with an increase of IL-4 and IFN-γ production and a decrease of IL-2 secretion after T-cell mitogen stimulation ex vivo. Collectively, FPN significantly deregulated genes related to T-cell progenitor differentiation, survival, and expansion, potentially leading to impaired thymopoiesis.
Collapse
Affiliation(s)
- Jui-Fang Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Hsin-Ying Wu
- Laboratory Animal Center, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Wei-Hsiang Huang
- Graduate Institute of Molecular and Comparative Pathobiology, National Taiwan University, Taipei 106, Taiwan
| | - Chen-Si Lin
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Chia-Chi Wang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
2
|
Stewart CM, Siegler EL, Sakemura RL, Cox MJ, Huynh T, Kimball B, Mai L, Can I, Manriquez Roman C, Yun K, Sirpilla O, Girsch JH, Ogbodo E, Mohammed Ismail W, Gaspar-Maia A, Budka J, Kim J, Scholler N, Mattie M, Filosto S, Kenderian SS. IL-4 drives exhaustion of CD8 + CART cells. Nat Commun 2024; 15:7921. [PMID: 39266501 PMCID: PMC11393358 DOI: 10.1038/s41467-024-51978-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/22/2024] [Indexed: 09/14/2024] Open
Abstract
Durable response to chimeric antigen receptor T (CART) cell therapy remains limited in part due to CART cell exhaustion. Here, we investigate the regulation of CART cell exhaustion with three independent approaches including: a genome-wide CRISPR knockout screen using an in vitro model for exhaustion, RNA and ATAC sequencing on baseline and exhausted CART cells, and RNA and ATAC sequencing on pre-infusion CART cell products from responders and non-responders in the ZUMA-1 clinical trial. Each of these approaches identify interleukin (IL)-4 as a regulator of CART cell dysfunction. Further, IL-4-treated CD8+ CART cells develop signs of exhaustion independently of the presence of CD4+ CART cells. Conversely, IL-4 pathway editing or the combination of CART cells with an IL-4 monoclonal antibody improves antitumor efficacy and reduces signs of CART cell exhaustion in mantle cell lymphoma xenograft mouse models. Therefore, we identify both a role for IL-4 in inducing CART exhaustion and translatable approaches to improve CART cell therapy.
Collapse
Affiliation(s)
- Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - R Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Truc Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Brooke Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Long Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - James H Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ekene Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Justin Budka
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Jenny Kim
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | | | - Mike Mattie
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Simone Filosto
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Li S, Luo X, Sun M, Wang Y, Zhang Z, Jiang J, Hu D, Zhang J, Wu Z, Wang Y, Huang W, Xia L. Context-dependent T-BOX transcription factor family: from biology to targeted therapy. Cell Commun Signal 2024; 22:350. [PMID: 38965548 PMCID: PMC11225425 DOI: 10.1186/s12964-024-01719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
T-BOX factors belong to an evolutionarily conserved family of transcription factors. T-BOX factors not only play key roles in growth and development but are also involved in immunity, cancer initiation, and progression. Moreover, the same T-BOX molecule exhibits different or even opposite effects in various developmental processes and tumor microenvironments. Understanding the multiple roles of context-dependent T-BOX factors in malignancies is vital for uncovering the potential of T-BOX-targeted cancer therapy. We summarize the physiological roles of T-BOX factors in different developmental processes and their pathological roles observed when their expression is dysregulated. We also discuss their regulatory roles in tumor immune microenvironment (TIME) and the newly arising questions that remain unresolved. This review will help in systematically and comprehensively understanding the vital role of the T-BOX transcription factor family in tumor physiology, pathology, and immunity. The intention is to provide valuable information to support the development of T-BOX-targeted therapy.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| | - Wenjie Huang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Wu J, Lu Z, Zhao H, Lu M, Gao Q, Che N, Wang J, Ma T. The expanding Pandora's toolbox of CD8 +T cell: from transcriptional control to metabolic firing. J Transl Med 2023; 21:905. [PMID: 38082437 PMCID: PMC10714647 DOI: 10.1186/s12967-023-04775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
CD8+ T cells are the executor in adaptive immune response, especially in anti-tumor immunity. They are the subset immune cells that are of high plasticity and multifunction. Their development, differentiation, activation and metabolism are delicately regulated by multiple factors. Stimuli from the internal and external environment could remodel CD8+ T cells, and correspondingly they will also make adjustments to the microenvironmental changes. Here we describe the most updated progresses in CD8+ T biology from transcriptional regulation to metabolism mechanisms, and also their interactions with the microenvironment, especially in cancer and immunotherapy. The expanding landscape of CD8+ T cell biology and discovery of potential targets to regulate CD8+ T cells will provide new viewpoints for clinical immunotherapy.
Collapse
Affiliation(s)
- Jinghong Wu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Zhendong Lu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Hong Zhao
- Department of Pathology, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Nanying Che
- Department of Pathology, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
5
|
Chen HW, Zhou R, Cao BF, Liu K, Zhong Q, Huang YN, Liu HM, Zhao JQ, Wu XB. The predictive, preventive, and personalized medicine of insomnia: gut microbiota and inflammation. EPMA J 2023; 14:571-583. [PMID: 38094575 PMCID: PMC10713890 DOI: 10.1007/s13167-023-00345-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/21/2023] [Indexed: 12/05/2024]
Abstract
Background The human gut microbiota (GM) has been recognized as a significant factor in the development of insomnia, primarily through inflammatory pathways, making it a promising target for therapeutic interventions. Considering the principles of primary prediction, targeted prevention, and personalized treatment medicine (PPPM), identifying specific gut microbiota associated with insomnia and exploring the underlying mechanisms comprehensively are crucial steps towards achieving primary prediction, targeted prevention, and personalized treatment of insomnia. Working hypothesis and methodology We hypothesized that alterations in the composition of specific GM could induce insomnia through an inflammatory response, which postulates the existence of a GM-inflammation-insomnia pathway. Mendelian randomization (MR) analyses were employed to examine this pathway and explore the mediative effects of inflammation. We utilized genetic proxies representing GM, insomnia, and inflammatory indicators (including 41 circulating cytokines and C-reactive protein (CRP)), specifically identified from European ancestry. The primary method used to identify insomnia-related GM and examine the medicative effect of inflammation was the inverse variance weighted method, supplemented by the MR-Egger and weighted median methods. Our findings have the potential to identify individuals at risk of insomnia through screening for GM imbalances, leading to the development of targeted prevention and personalized treatment strategies for the condition. Results Nine genera and three circulating cytokines were identified to be associated with insomnia; only the associations of Clostridium (innocuum group) and β-NGF on insomnia remained significant after the FDR test, OR = 1.08 (95% CI = 1.04-1.12, P = 1.45 × 10-4, q = 0.02) and OR = 1.06 (95% CI = 1.02-1.10, P = 1.06 × 10-3, q = 0.04), respectively. CRP was associated with an increased risk of insomnia, OR = 1.05 (95% CI = 1.01-1.10, P = 6.42 × 10-3). CRP mediated the association of Coprococcus 1, Holdemania, and Rikenellaceae (RC9gut group) with insomnia. No heterogeneity or pleiotropy were detected. Conclusions Our study highlights the role of specific GM alterations in the development of insomnia and provides insights into the mediating effects of inflammation. Targeting these specific GM alterations presents a promising avenue for advancing the transition from reactive medicine to PPPM in managing insomnia, potentially leading to significant clinical benefits. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00345-1.
Collapse
Affiliation(s)
- Hao-Wen Chen
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Rui Zhou
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Bi-Fei Cao
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Kuan Liu
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Qi Zhong
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Yi-Ning Huang
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Hua-Min Liu
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin-Qing Zhao
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| | - Xian-Bo Wu
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, No. 1063-No. 1023, Shatai South Road, Baiyun District, Guangzhou, 510515 China
| |
Collapse
|
6
|
Rodríguez-Tomàs E, Arenas M, Baiges-Gaya G, Acosta J, Araguas P, Malave B, Castañé H, Jiménez-Franco A, Benavides-Villarreal R, Sabater S, Solà-Alberich R, Camps J, Joven J. Gradient Boosting Machine Identified Predictive Variables for Breast Cancer Patients Pre- and Post-Radiotherapy: Preliminary Results of an 8-Year Follow-Up Study. Antioxidants (Basel) 2022; 11:antiox11122394. [PMID: 36552602 PMCID: PMC9774765 DOI: 10.3390/antiox11122394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy (RT) is part of the standard treatment of breast cancer (BC) because of its effects on relapse reduction and survival. However, response to treatment is highly variable, and some patients may develop disease progression (DP), a second primary cancer, or may succumb to the disease. Antioxidant systems and inflammatory processes are associated with the onset and development of BC and play a role in resistance to treatment. Here, we report our investigation into the clinical evolution of BC patients, and the impact of RT on the circulating levels of the antioxidant enzyme paraoxonase-1 (PON1), cytokines, and other standard biochemical and hematological variables. Gradient Boosting Machine (GBM) algorithm was used to identify predictive variables. This was a retrospective study in 237 patients with BC. Blood samples were obtained pre- and post-RT, with samples of healthy women used as control subjects. Results showed that 24 patients had DP eight years post-RT, and eight patients developed a second primary tumor. The algorithm identified interleukin-4 and total lymphocyte counts as the most relevant indices discriminating between BC patients and control subjects, while neutrophils, total leukocytes, eosinophils, very low-density lipoprotein cholesterol, and PON1 activity were potential predictors of fatal outcome.
Collapse
Affiliation(s)
- Elisabet Rodríguez-Tomàs
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-977-310-300 (ext. 54132) (M.A.); +34-977-310-300 (ext. 55409) (J.C.)
| | - Gerard Baiges-Gaya
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Johana Acosta
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Pablo Araguas
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Bárbara Malave
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Rocío Benavides-Villarreal
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Sebastià Sabater
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Rosa Solà-Alberich
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-977-310-300 (ext. 54132) (M.A.); +34-977-310-300 (ext. 55409) (J.C.)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| |
Collapse
|
7
|
Viano ME, Baez NS, Savid-Frontera C, Lidon NL, Hodge DL, Herbelin A, Gombert JM, Barbarin A, Rodriguez-Galan MC. Virtual Memory CD8 + T Cells: Origin and Beyond. J Interferon Cytokine Res 2022; 42:624-642. [PMID: 36083273 PMCID: PMC9835308 DOI: 10.1089/jir.2022.0053] [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: 03/09/2022] [Accepted: 04/19/2022] [Indexed: 01/21/2023] Open
Abstract
The presence of CD8+ T cells with a memory phenotype in nonimmunized mice has been noted for decades, but it was not until about 2 decades ago that they began to be studied in greater depth. Currently called virtual memory CD8+ T cells, they consist of a heterogeneous group of cells with memory characteristics, without any previous contact with their specific antigens. These cells were identified in mice, but a few years ago, a cell type with characteristics equivalent to the murine ones was described in healthy humans. In this review, we address the different aspects of its biology mainly developed in murine models and what is currently known about its cellular equivalent in humans.
Collapse
Affiliation(s)
- Maria Estefania Viano
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia Soledad Baez
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolás Leonel Lidon
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - André Herbelin
- Inserm U1313, Poitiers, France
- Université de Poitiers, Poitiers, France
| | - Jean-Marc Gombert
- Inserm U1313, Poitiers, France
- Université de Poitiers, Poitiers, France
- Service d'Immunologie et Inflammation, CHU de Poitiers, Poitiers, France
| | - Alice Barbarin
- Inserm U1313, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | | |
Collapse
|
8
|
Bensussen A, Santana MA, Rodríguez-Jorge O. Metabolic alterations impair differentiation and effector functions of CD8+ T cells. Front Immunol 2022; 13:945980. [PMID: 35983057 PMCID: PMC9380903 DOI: 10.3389/fimmu.2022.945980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T lymphocytes are one of the main effector cells of the immune system, they protect the organism against intracellular threats such as viruses and bacteria, as well as neoplasms. It is currently well established that CD8+ T cells have distinct immune responses, given by their phenotypes Tc1, Tc2, Tc17, and TcReg. The cellular plasticity of such phenotypes depends on the presence of different combinations of cytokines in the extracellular medium. It is known that metabolic imbalances play an important role in immune response, but the precise role of metabolic disturbances on the differentiation and function of CD8+ T cells, however, has not been explored. In this work, we used a computational model to explore the potential effect of metabolic alterations such as hyperglycemia, high alcohol consumption, dyslipidemia, and diabetes on CD8+ T cell differentiation. Our model predicts that metabolic alterations preclude the effector function of all CD8+ T cell phenotypes except for TcReg cells. It also suggests that such inhibition originates from the increase of reactive oxygen species in response to metabolic stressors. Finally, we simulated the outcome of treating metabolic-inhibited CD8+ T cells with drugs targeting key molecules such as mTORC1, mTORC2, Akt, and others. We found that overstimulation of mTORC2 may restore cell differentiation and functions of all effector phenotypes, even in diabetic patients. These findings highlight the importance of our predictive model to find potential targets to strengthen immunosuppressed patients in chronic diseases, like diabetes.
Collapse
Affiliation(s)
- Antonio Bensussen
- Laboratorio de Dinámica de Redes Genéticas, Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Maria Angelica Santana
- Laboratorio de Inmunología, Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Otoniel Rodríguez-Jorge
- Laboratorio de Inmunología, Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
- *Correspondence: Otoniel Rodríguez-Jorge,
| |
Collapse
|
9
|
Kwesi-Maliepaard EM, Jacobs H, van Leeuwen F. Signals for antigen-independent differentiation of memory CD8 + T cells. Cell Mol Life Sci 2021; 78:6395-6408. [PMID: 34398252 PMCID: PMC8558200 DOI: 10.1007/s00018-021-03912-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 12/18/2022]
Abstract
Conventional CD8+ memory T cells develop upon stimulation with foreign antigen and provide increased protection upon re-challenge. Over the past two decades, new subsets of CD8+ T cells have been identified that acquire memory features independently of antigen exposure. These antigen-inexperienced memory T cells (TAIM) are described under several names including innate memory, virtual memory, and memory phenotype. TAIM cells exhibit characteristics of conventional or true memory cells, including antigen-specific responses. In addition, they show responsiveness to innate stimuli and have been suggested to provide additional levels of protection toward infections and cancer. Here, we discuss the current understanding of TAIM cells, focusing on extrinsic and intrinsic molecular conditions that favor their development, their molecular definitions and immunological properties, as well as their transcriptional and epigenetic regulation.
Collapse
Affiliation(s)
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands
| | - Fred van Leeuwen
- Division of Gene Regulation, Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands.
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Mammadli M, Harris R, Mahmudlu S, Verma A, May A, Dhawan R, Waickman AT, Sen JM, August A, Karimi M. Human Wnt/β-Catenin Regulates Alloimmune Signaling during Allogeneic Transplantation. Cancers (Basel) 2021; 13:cancers13153798. [PMID: 34359702 PMCID: PMC8345079 DOI: 10.3390/cancers13153798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is one of the most widely applied forms of adoptive immunotherapy for the treatment of hematological malignancies. Detrimental graft-versus-host disease (GVHD), but also beneficial graft-versus-leukemia (GVL) effects occurring after allo-HSCT are largely mediated by alloantigen-reactive donor T cells in the graft. Separating GVHD from GVL effects is a formidable challenge, and a greater understanding of donor T cell biology is required to accomplish the uncoupling of GVHD from GVL. Here, we evaluated the role of β-catenin in this process. Using a unique mouse model of transgenic overexpression of human β-catenin (Cat-Tg) in an allo-HSCT model, we show here that T cells from Cat-Tg mice did not cause GVHD, and surprisingly, Cat-Tg T cells maintained the GVL effect. Donor T cells from Cat-Tg mice exhibited significantly lower inflammatory cytokine production and reduced donor T cell proliferation, while upregulating cytotoxic mediators that resulted in enhanced cytotoxicity. RNA sequencing revealed changes in the expression of 1169 genes for CD4, and 1006 genes for CD8+ T cells involved in essential aspects of immune response and GVHD pathophysiology. Altogether, our data suggest that β-catenin is a druggable target for developing therapeutic strategies to reduce GVHD while preserving the beneficial GVL effects following allo-HSCT treatment.
Collapse
Affiliation(s)
- Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Sara Mahmudlu
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Anjali Verma
- Biomedical Research Center, National Institute on Aging-National Institutes of Health, 08C218, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA; (A.V.); (J.M.S.)
| | - Adriana May
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Rohan Dhawan
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Adam T. Waickman
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
| | - Jyoti Misra Sen
- Biomedical Research Center, National Institute on Aging-National Institutes of Health, 08C218, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA; (A.V.); (J.M.S.)
- Immunology Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA;
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (M.M.); (R.H.); (S.M.); (A.M.); (R.D.); (A.T.W.)
- Correspondence: ; Tel.: +315-464-2344 or +315-464-7652
| |
Collapse
|
11
|
Mammadli M, Huang W, Harris R, Xiong H, Weeks S, May A, Gentile T, Henty-Ridilla J, Waickman AT, August A, Bah A, Karimi M. Targeting SLP76:ITK interaction separates GVHD from GVL in allo-HSCT. iScience 2021; 24:102286. [PMID: 33851101 PMCID: PMC8024657 DOI: 10.1016/j.isci.2021.102286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/27/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for hematological malignancies, due to graft-versus-leukemia (GVL) activity mediated by alloreactive donor T cells. However, graft-versus-host disease (GVHD) is also mediated by these cells. Here, we assessed the effect of attenuating TCR-mediated SLP76:ITK interaction in GVL vs. GVHD effects after allo-HSCT. CD8+ and CD4+ donor T cells from mice expressing a Y145F mutation in SLP-76 did not cause GVHD but preserved GVL effects against B-ALL cells. SLP76Y145FKI CD8+ and CD4+ donor T cells also showed less inflammatory cytokine production and migration to GVHD target organs. We developed a novel peptide to specifically inhibit SLP76:ITK interactions, resulting in decreased phosphorylation of PLCγ1 and ERK, decreased cytokine production in human T cells, and separation of GVHD from GVL effects. Altogether, our data suggest that inhibiting SLP76:ITK interaction could be a therapeutic strategy to separate GVHD from GVL effects after allo-HSCT treatment.
Collapse
Affiliation(s)
- Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Hui Xiong
- Department of Radiology, Jiangxi Health Vocational College, Nanchang, 330052, China
| | - Samuel Weeks
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Adriana May
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Teresa Gentile
- Division of Hematology, translational research, SUNY Upstate Medical University, Syracuse NY 13210, USA
| | - Jessica Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Adam T. Waickman
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Alaji Bah
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| |
Collapse
|
12
|
Emerson DA, Rolig AS, Redmond WL. Enhancing the Generation of Eomes hi CD8 + T Cells Augments the Efficacy of OX40- and CTLA-4-Targeted Immunotherapy. Cancer Immunol Res 2021; 9:430-440. [PMID: 33593794 DOI: 10.1158/2326-6066.cir-20-0338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/10/2020] [Accepted: 02/12/2021] [Indexed: 11/16/2022]
Abstract
CTLA-4 blockade in combination with an agonist OX40-specific monoclonal antibody synergizes to augment antitumor immunity through enhanced T-cell effector function, leading to increased survival in preclinical cancer models. We have shown previously that anti-OX40/anti-CTLA-4 combination therapy synergistically enhances the expression of Eomesodermin (Eomes) in CD8+ T cells. Eomes is a critical transcription factor for the differentiation and memory function of CD8+ T cells. We hypothesized that EomeshiCD8+ T cells were necessary for anti-OX40/anti-CTLA-4 immunotherapy efficacy and that further enhancement of this population would improve tumor-free survival. Indeed, CD8+ T cell-specific deletion of Eomes abrogated the efficacy of anti-OX40/anti-CTLA-4 therapy. We also found that anti-OX40/anti-CTLA-4-induced EomeshiCD8+ T cells expressed lower levels of checkpoint receptors (PD1, Tim-3, and Lag-3) and higher levels of effector cytokines (IFNγ and TNFα) than their Eomeslo counterparts. Eomes expression is negatively regulated in T cells through interleukin-2-inducible T-cell kinase (ITK) signaling. We investigated the impact of modulating ITK signaling with ibrutinib, an FDA-approved tyrosine kinase inhibitor, and found that anti-OX40/anti-CTLA-4/ibrutinib therapy further enhanced CD8+ T cell-specific Eomes expression, leading to enhanced tumor regression and improved survival, both of which were associated with increased T-cell effector function across multiple tumor models. Taken together, these data demonstrate the potential of anti-OX40/anti-CTLA-4/ibrutinib as a triple therapy to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Dana A Emerson
- Cell, Developmental, and Cancer Biology Department, Oregon Health and Science University, Portland, Oregon.,Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Annah S Rolig
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon.
| |
Collapse
|
13
|
Mammadli M, Huang W, Harris R, Sultana A, Cheng Y, Tong W, Pu J, Gentile T, Dsouza S, Yang Q, Bah A, August A, Karimi M. Targeting Interleukin-2-Inducible T-Cell Kinase (ITK) Differentiates GVL and GVHD in Allo-HSCT. Front Immunol 2020; 11:593863. [PMID: 33324410 PMCID: PMC7726260 DOI: 10.3389/fimmu.2020.593863] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/29/2020] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is a potentially curative procedure for many malignant diseases. Donor T cells prevent disease recurrence via graft-versus-leukemia (GVL) effect. Donor T cells also contribute to graft-versus-host disease (GVHD), a debilitating and potentially fatal complication. Novel treatment strategies are needed which allow preservation of GVL effects without causing GVHD. Using murine models, we show that targeting IL-2-inducible T cell kinase (ITK) in donor T cells reduces GVHD while preserving GVL effects. Both CD8+ and CD4+ donor T cells from Itk-/- mice produce less inflammatory cytokines and show decrease migration to GVHD target organs such as the liver and small intestine, while maintaining GVL efficacy against primary B-cell acute lymphoblastic leukemia (B-ALL). Itk-/- T cells exhibit reduced expression of IRF4 and decreased JAK/STAT signaling activity but upregulating expression of Eomesodermin (Eomes) and preserve cytotoxicity, necessary for GVL effect. Transcriptome analysis indicates that ITK signaling controls chemokine receptor expression during alloactivation, which in turn affects the ability of donor T cells to migrate to GVHD target organs. Our data suggest that inhibiting ITK could be a therapeutic strategy to reduce GVHD while preserving the beneficial GVL effects following allo-HSCT treatment.
Collapse
Affiliation(s)
- Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States.,Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Aisha Sultana
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Ying Cheng
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jeffery Pu
- Department of Hematology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Teresa Gentile
- Department of Hematology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Shanti Dsouza
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Qi Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Alaji Bah
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
14
|
Abstract
Recent studies suggest that murine invariant natural killer T (iNKT) cell development culminates in three terminally differentiated iNKT cell subsets denoted as NKT1, 2, and 17 cells. Although these studies corroborate the significance of the subset division model, less is known about the factors driving subset commitment in iNKT cell progenitors. In this review, we discuss the latest findings in iNKT cell development, focusing in particular on how T-cell receptor signal strength steers iNKT cell progenitors toward specific subsets and how early progenitor cells can be identified. In addition, we will discuss the essential factors for their sustenance and functionality. A picture is emerging wherein the majority of thymic iNKT cells are mature effector cells retained in the organ rather than developing precursors.
Collapse
Affiliation(s)
- Kristin Hogquist
- Center for Immunology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hristo Georgiev
- Center for Immunology, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
15
|
Hussain T, Quinn KM. Similar but different: virtual memory CD8 T cells as a memory-like cell population. Immunol Cell Biol 2019; 97:675-684. [PMID: 31140625 DOI: 10.1111/imcb.12277] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/26/2019] [Accepted: 05/26/2019] [Indexed: 01/02/2023]
Abstract
Immunological memory is a phenomenon where the immune system can respond more rapidly to pathogens and immunological challenges that it has previously encountered. It is defined by several key hallmarks. After an initial encounter, immune cells (1) expand and (2) differentiate to form memory cell populations. Memory cells are (3) long-lived and (4) facilitate more rapid immune responses to subsequent infection because of (i) an increase in cell number, (ii) a decrease in the signaling threshold required for entry into cell cycle or effector function and (iii) localization of cells to tissue sites for surveillance. Classically, immunological memory has been antigen specific but it is becoming apparent that mechanisms of immunological memory can be co-opted by innate or antigen-inexperienced immune cells to generate heterogeneity in immune responses. One such cell is the virtual memory CD8 T (TVM ) cell, which is a semi-differentiated but antigen-naïve CD8 T-cell population. This review will summarize current knowledge of how TVM cells are generated, their memory-like hallmarks, how they are maintained during steady state, infection and aging, and propose a model to integrate key signaling pathways during their generation.
Collapse
Affiliation(s)
- Tabinda Hussain
- Monash University Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Kylie M Quinn
- Monash University Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,RMIT University School of Biomedical and Health Sciences, Bundoora, VIC, Australia
| |
Collapse
|
16
|
Baez NS, Cerbán F, Savid-Frontera C, Hodge DL, Tosello J, Acosta-Rodriguez E, Almada L, Gruppi A, Viano ME, Young HA, Rodriguez-Galan MC. Thymic expression of IL-4 and IL-15 after systemic inflammatory or infectious Th1 disease processes induce the acquisition of "innate" characteristics during CD8+ T cell development. PLoS Pathog 2019; 15:e1007456. [PMID: 30608984 PMCID: PMC6319713 DOI: 10.1371/journal.ppat.1007456] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 11/05/2018] [Indexed: 01/28/2023] Open
Abstract
Innate CD8+ T cells express a memory-like phenotype and demonstrate a strong cytotoxic capacity that is critical during the early phase of the host response to certain bacterial and viral infections. These cells arise in the thymus and depend on IL-4 and IL-15 for their development. Even though innate CD8+ T cells exist in the thymus of WT mice in low numbers, they are highly enriched in KO mice that lack certain kinases, leading to an increase in IL-4 production by thymic NKT cells. Our work describes that in C57BL/6 WT mice undergoing a Th1 biased infectious disease, the thymus experiences an enrichment of single positive CD8 (SP8) thymocytes that share all the established phenotypical and functional characteristics of innate CD8+ T cells. Moreover, through in vivo experiments, we demonstrate a significant increase in survival and a lower parasitemia in mice adoptively transferred with SP8 thymocytes from OT I—T. cruzi-infected mice, demonstrating that innate CD8+ thymocytes are able to protect against a lethal T. cruzi infection in an Ag-independent manner. Interestingly, we obtained similar results when using thymocytes from systemic IL-12 + IL-18-treated mice. This data indicates that cytokines triggered during the acute stage of a Th1 infectious process induce thymic production of IL-4 along with IL-15 expression resulting in an adequate niche for development of innate CD8+ T cells as early as the double positive (DP) stage. Our data demonstrate that the thymus can sense systemic inflammatory situations and alter its conventional CD8 developmental pathway when a rapid innate immune response is required to control different types of pathogens. Murine innate CD8+ T cells demonstrate strong cytotoxic capacity during the early phase of certain bacterial and viral infections. Such cells have been reported to be present in both mice and humans but many questions remain as to their differentiation and maturation process. Innate CD8+ T cells arise in the thymus and depend on IL-4 and IL-15 for their development. A description of the cellular and molecular mechanisms involved during their thymic development has been obtained from KO mice that lack kinases and transcription factors important for TCR signaling. In these mice, SP8 thymocytes with an innate phenotype are highly enriched over the conventional SP8 cells. Our work describes, for the first time, that in WT mice, thymic IL-4 and IL-15 expression triggered by Th1 infectious processes induce an adequate niche for development of innate rather than conventional CD8+ T cells. Our data show that the thymus is able to sense a systemic inflammatory response (probably mediated by systemic IL-12 and IL-18 production) and alter its ontogeny when pathogen control is needed.
Collapse
Affiliation(s)
- Natalia S. Baez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabio Cerbán
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Deborah L. Hodge
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Jimena Tosello
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Eva Acosta-Rodriguez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Almada
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Estefania Viano
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Howard A. Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Maria Cecilia Rodriguez-Galan
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
17
|
Gruarin P, Maglie S, Simone M, Häringer B, Vasco C, Ranzani V, Bosotti R, Noddings JS, Larghi P, Facciotti F, Sarnicola ML, Martinovic M, Crosti M, Moro M, Rossi RL, Bernardo ME, Caprioli F, Locatelli F, Rossetti G, Abrignani S, Pagani M, Geginat J. Eomesodermin controls a unique differentiation program in human IL‐10 and IFN‐γ coproducing regulatory T cells. Eur J Immunol 2018; 49:96-111. [DOI: 10.1002/eji.201847722] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/28/2018] [Accepted: 11/09/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Paola Gruarin
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Stefano Maglie
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Marco Simone
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Barbara Häringer
- German Rheumatology Research Center DRFZ Berlin Germany
- Charitè Research Center for Immunosciences RCIS Berlin Germany
| | - Chiara Vasco
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Valeria Ranzani
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Roberto Bosotti
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Johanna S. Noddings
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Paola Larghi
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Federica Facciotti
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Maria L. Sarnicola
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Martina Martinovic
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Mariacristina Crosti
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Monica Moro
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Riccardo L. Rossi
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Maria E. Bernardo
- Ospedale Pediatrico Bambino Gesù Dipartimento Onco–Ematologia e Medicina Trasfusionale Rome Italy
| | - Flavio Caprioli
- Unità Operativa di Gastroenterologia ed Endoscopia Fondazione Ca’ Granda Ospedale Maggiore Policlinico Milan Italy
| | - Franco Locatelli
- Ospedale Pediatrico Bambino Gesù Dipartimento Onco–Ematologia e Medicina Trasfusionale Rome Italy
| | - Grazisa Rossetti
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| | - Sergio Abrignani
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
- Department of Clinical Science and Community Health (DISCCO) University of Milan Milan Italy
| | - Massimiliano Pagani
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
- Department of Medical Biotechnology and Translational Medicine University of Milan Milan Italy
| | - Jens Geginat
- INGM‐National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi” Milan Italy
| |
Collapse
|
18
|
Helminth-induced IL-4 expands bystander memory CD8 + T cells for early control of viral infection. Nat Commun 2018; 9:4516. [PMID: 30375396 PMCID: PMC6207712 DOI: 10.1038/s41467-018-06978-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022] Open
Abstract
Infection with parasitic helminths can imprint the immune system to modulate bystander inflammatory processes. Bystander or virtual memory CD8+ T cells (TVM) are non-conventional T cells displaying memory properties that can be generated through responsiveness to interleukin (IL)-4. However, it is not clear if helminth-induced type 2 immunity functionally affects the TVM compartment. Here, we show that helminths expand CD44hiCD62LhiCXCR3hiCD49dlo TVM cells through direct IL-4 signaling in CD8+ T cells. Importantly, helminth-mediated conditioning of TVM cells provided enhanced control of acute respiratory infection with the murid gammaherpesvirus 4 (MuHV-4). This enhanced control of MuHV-4 infection could further be explained by an increase in antigen-specific CD8+ T cell effector responses in the lung and was directly dependent on IL-4 signaling. These results demonstrate that IL-4 during helminth infection can non-specifically condition CD8+ T cells, leading to a subsequently raised antigen-specific CD8+ T cell activation that enhances control of viral infection. Parasitic helminth infection is known to impact upon the host response to other bystander inflammatory processes. Here the authors show that IL4 production induced by helminth infection results in expansion of bystander CD8+ memory T cells and enhanced control to viral infection.
Collapse
|
19
|
Harland KL, Fox A, Nüssing S, Hensen L, Kedzierska K, Turner SJ, Kelso A. Limited Phenotypic and Functional Plasticity of Influenza Virus–Specific Memory CD8+T Cells during Activation in an Alternative Cytokine Environment. THE JOURNAL OF IMMUNOLOGY 2018; 201:3282-3293. [DOI: 10.4049/jimmunol.1701672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 09/17/2018] [Indexed: 12/21/2022]
|
20
|
Ni X, Fu B, Zhang J, Sun R, Tian Z, Wei H. Cytokine-Based Generation of CD49a +Eomes -/+ Natural Killer Cell Subsets. Front Immunol 2018; 9:2126. [PMID: 30319610 PMCID: PMC6167425 DOI: 10.3389/fimmu.2018.02126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/29/2018] [Indexed: 01/06/2023] Open
Abstract
Recent studies have identified CD49a+Eomes− and CD49a+Eomes+ subsets of tissue-resident NK (trNK) cells in different organs of the mouse. However, the characteristics of CD49a+Eomes−/+ NK cell development and the regulation of Eomes expression in NK cells remain unclear. Here, we established an in vitro cytokine-based feeder-free system in which bone marrow progenitor cells differentiate into CD49a+ NK cells. IL-15 was identified as being the key cytokine in this system that supported the development and maintenance of CD49a+ NK cells. The CD49a+ NK cells generated were Eomes−CD49b− and shared the same phenotype as hepatic trNK cells. IL-4 induced the expression of Eomes in generated NK cells and converted them into CD49a+Eomes+ cells, which were phenotypically and functionally similar to uterine trNK cells. Moreover, the IL-4/STAT6 axis was identified as being important in the generation of CD49a+Eomes+ induced NK cells. Collectively, these studies describe an approach to generate CD49a+Eomes−/+ subsets of NK cells and demonstrate important roles for IL-15 and IL-4 in the differentiation of these cells. These findings have potential for developmental research underlying the generation of different subsets of NK cells and the application of adoptive NK cell transfer therapies.
Collapse
Affiliation(s)
- Xiang Ni
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Binqing Fu
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Jinghe Zhang
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
21
|
Tuttle KD, Krovi SH, Zhang J, Bedel R, Harmacek L, Peterson LK, Dragone LL, Lefferts A, Halluszczak C, Riemondy K, Hesselberth JR, Rao A, O'Connor BP, Marrack P, Scott-Browne J, Gapin L. TCR signal strength controls thymic differentiation of iNKT cell subsets. Nat Commun 2018; 9:2650. [PMID: 29985393 PMCID: PMC6037704 DOI: 10.1038/s41467-018-05026-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/07/2018] [Indexed: 12/22/2022] Open
Abstract
During development in the thymus, invariant natural killer T (iNKT) cells commit to one of three major functionally different subsets, iNKT1, iNKT2, and iNKT17. Here, we show that T cell antigen receptor (TCR) signal strength governs the development of iNKT cell subsets, with strong signaling promoting iNKT2 and iNKT17 development. Altering TCR diversity or signaling diminishes iNKT2 and iNKT17 cell subset development in a cell-intrinsic manner. Decreased TCR signaling affects the persistence of Egr2 expression and the upregulation of PLZF. By genome-wide comparison of chromatin accessibility, we identify a subset of iNKT2-specific regulatory elements containing NFAT and Egr binding motifs that is less accessible in iNKT2 cells that develop from reduced TCR signaling. These data suggest that variable TCR signaling modulates regulatory element activity at NFAT and Egr binding sites exerting a determinative influence on the dynamics of gene enhancer accessibility and the developmental fate of iNKT cells. Invariant natural killer T (iNKT) cells can be subsetted by their cytokine profiles, but how they develop in the thymus is unclear. Here the authors show, by analysing mice carrying mutant Zap70 genes, that T cell receptor signaling strength induces epigenetic changes of genes to modulate iNKT lineages.
Collapse
Affiliation(s)
- Kathryn D Tuttle
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - S Harsha Krovi
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Jingjing Zhang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Romain Bedel
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.,Department of Oncology, University of Lausanne, Chemin des Boveresses 155, Epalinges, 1066, Switzerland
| | - Laura Harmacek
- Center for Genes, Environment, and Health, Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.,Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, 80206, CO, USA
| | - Lisa K Peterson
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, 80206, CO, USA.,ARUP Laboratories, Institute of Clinical and Experimental Pathology, 500 Chipeta Way, Salt Lake City, 84108, UT, Switzerland.,Department of Pathology, University of Utah, 30N 1900E, Salt Lake City, 84132, UT, USA
| | - Leonard L Dragone
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, 80206, CO, USA.,Merck Research Laboratories, San Francisco, CA, USA
| | - Adam Lefferts
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Catherine Halluszczak
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Kent Riemondy
- RNA Bioscience Initiative, University of Colorado School of Medicine, 12800 E. 19th Ave, Aurora, 80045, CO, USA
| | - Jay R Hesselberth
- RNA Bioscience Initiative, University of Colorado School of Medicine, 12800 E. 19th Ave, Aurora, 80045, CO, USA.,Department of Biochemistry & Molecular Genetics, University of Colorado School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA
| | - Anjana Rao
- La Jolla Institute, 9420 Athena Cir, La Jolla, 92037, CA, USA.,Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Dr, La Jolla, CA, 92037, USA.,University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Brian P O'Connor
- Center for Genes, Environment, and Health, Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.,Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, 80206, CO, USA
| | - Philippa Marrack
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA
| | - James Scott-Browne
- La Jolla Institute, 9420 Athena Cir, La Jolla, 92037, CA, USA.,Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Dr, La Jolla, CA, 92037, USA
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E. 19th Ave, Aurora, CO, 80045, USA. .,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA.
| |
Collapse
|
22
|
Qin Y, Shin JH, Yoon JH, Park SH. Embryonic Fibroblasts Promote Antitumor Cytotoxic Effects of CD8 + T Cells. Front Immunol 2018; 9:685. [PMID: 29706956 PMCID: PMC5908885 DOI: 10.3389/fimmu.2018.00685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
Adoptive CD8+ T cell therapy has emerged as an important modality for the treatment of cancers. However, the significant drawback of transfused T cells is their poor survival and functionality in response to tumors. To overcome this limitation, an important consideration is exploring a culture condition to generate superior antitumor cytotoxic T lymphocytes (CTLs) for adoptive therapy. Here, we provide a novel approach to generate potent CTL clones in mouse embryonic fibroblast-conditioned medium (MEF-CM). We found CTLs derived with MEF-CM have higher potential in long-term persistence in tumor bearing and non-tumor-bearing mice. Importantly, adoptive transfer of MEF-CM-cultured CTLs dramatically regressed tumor growth and prolonged mice survival. Characterization of MEF-CM-cultured CTLs (effector molecules, phenotypes, and transcription factors) suggests that MEF-CM enhances the effector functions of CD8+ T cells in part by the upregulation of the T-box transcription factor eomesodermin. Consequently, MEF-CM enhances the intrinsic qualities of effector CD8+ T cells to augment antitumor immunity.
Collapse
Affiliation(s)
- Yingyu Qin
- Department of Life Sciences, Korea University, Seoul, South Korea.,ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jung Hoon Shin
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jeong-Ho Yoon
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Se-Ho Park
- Department of Life Sciences, Korea University, Seoul, South Korea
| |
Collapse
|
23
|
Renkema KR, Lee JY, Lee YJ, Hamilton SE, Hogquist KA, Jameson SC. IL-4 sensitivity shapes the peripheral CD8+ T cell pool and response to infection. J Exp Med 2016; 213:1319-29. [PMID: 27298446 PMCID: PMC4925014 DOI: 10.1084/jem.20151359] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 04/28/2016] [Indexed: 11/15/2022] Open
Abstract
Homeostatic levels of IL-4 are necessary for normal development of memory phenotype CD8+ T cells and naive CD8+ T cells and for a robust CD8+ T cell response to LCMV infection. Previous studies have revealed that a population of innate memory CD8+ T cells is generated in response to IL-4, first appearing in the thymus and bearing high expression levels of Eomesodermin (Eomes) but not T-bet. However, the antigen specificity and functional properties of these cells is poorly defined. In this study, we show that IL-4 regulates not only the frequency and function of innate memory CD8+ T cells, but also regulates Eomes expression levels and functional reactivity of naive CD8+ T cells. Lack of IL-4 responsiveness attenuates the capacity of CD8+ T cells to mount a robust response to lymphocytic choriomeningitis virus infection, with both quantitative and qualitative effects on effector and memory antigen-specific CD8+ T cells. Unexpectedly, we found that, although numerically rare, memory phenotype CD8+ T cells in IL-4Rα–deficient mice exhibited enhanced reactivity after in vitro and in vivo stimulation. Importantly, our data revealed that these effects of IL-4 exposure occur before, not during, infection. Together, these data show that IL-4 influences the entire peripheral CD8+ T cell pool, influencing expression of T-box transcription factors, functional reactivity, and the capacity to respond to infection. These findings indicate that IL-4, a canonical Th2 cell cytokine, can sometimes promote rather than impair Th1 cell–type immune responses.
Collapse
Affiliation(s)
- Kristin R Renkema
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - June-Yong Lee
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - You Jeong Lee
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Sara E Hamilton
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| |
Collapse
|
24
|
Park HJ, Lee A, Lee JI, Park SH, Ha SJ, Jung KC. Effect of IL-4 on the Development and Function of Memory-like CD8 T Cells in the Peripheral Lymphoid Tissues. Immune Netw 2016; 16:126-33. [PMID: 27162529 PMCID: PMC4853498 DOI: 10.4110/in.2016.16.2.126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023] Open
Abstract
Unlike conventional T cells, innate CD8 T cells develop a memory-like phenotype in the thymus and immediately respond upon antigen stimulation, similar to memory T cells. The development of innate CD8 T cells in the thymus is known to require IL-4, which upregulates Eomesodermin (Eomes). These features are similar to that of virtual memory CD8 T cells and IL-4-induced memory-like CD8 T cells generated in the peripheral tissues. However, the relationship between these cell types has not been clearly documented. In the present study, IL-4-induced memory-like CD8 T cells generated in the peripheral tissues were compared with innate CD8 T cells in terms of phenotype and function. When an IL-4/anti-IL-4 antibody complex (IL-4C) was injected into C57BL/6 mice daily for 7 days, the EomeshiCXCR3 + CD8 T cell population was markedly increased in the peripheral lymphoid organs and blood. These cells were generated from naïve CD8 T cells or accumulated via the expansion of pre-existing CD44hiCXCR3 + CD8 T cells. Initially, the majority of these CXCR3 + CD8 T cells expressed low levels of CD44, which was followed by the conversion to the CD44hi phenotype. This conversion was associated with the acquisition of enhanced effector function. After discontinuation of IL-4C treatment, Eomes expression levels gradually decreased in CXCR3 + CD8 T cells. Taken together, the results of this study demonstrate that IL-4-induced memory-like CD8 T cells generated in the peripheral lymphoid tissues are phenotypically and functionally similar to the innate CD8 T cells generated in the thymus.
Collapse
Affiliation(s)
- Hi-Jung Park
- Graduate Course of Translational medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ara Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jae-Il Lee
- Graduate Course of Translational medicine, Seoul National University College of Medicine, Seoul 03080, Korea.; Transplantation Research Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Seong Hoe Park
- Graduate Course of Translational medicine, Seoul National University College of Medicine, Seoul 03080, Korea.; Transplantation Research Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Kyeong Cheon Jung
- Graduate Course of Translational medicine, Seoul National University College of Medicine, Seoul 03080, Korea.; Transplantation Research Institute, Seoul National University Medical Research Center, Seoul 03080, Korea.; Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
25
|
Regulation of IL-4 Expression in Immunity and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:31-77. [PMID: 27734408 DOI: 10.1007/978-94-024-0921-5_3] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-4 was first identified as a T cell-derived growth factor for B cells. Studies over the past several decades have markedly expanded our understanding of its cellular sources and function. In addition to T cells, IL-4 is produced by innate lymphocytes, such as NTK cells, and myeloid cells, such as basophils and mast cells. It is a signature cytokine of type 2 immune response but also has a nonimmune function. Its expression is tightly regulated at several levels, including signaling pathways, transcription factors, epigenetic modifications, microRNA, and long noncoding RNA. This chapter will review in detail the molecular mechanism regulating the cell type-specific expression of IL-4 in physiological and pathological type 2 immune responses.
Collapse
|
26
|
Type I interferons regulate eomesodermin expression and the development of unconventional memory CD8(+) T cells. Nat Commun 2015; 6:7089. [PMID: 25953241 PMCID: PMC4432629 DOI: 10.1038/ncomms8089] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/01/2015] [Indexed: 12/24/2022] Open
Abstract
CD8(+) T-cell memory phenotype and function are acquired after antigen-driven activation. Memory-like cells may also arise in absence of antigenic exposure in the thymus or in the periphery. Eomesodermin (Eomes) is a key transcription factor for the development of these unconventional memory cells. Herein, we show that type I interferon signalling in CD8(+) T cells directly activates Eomes gene expression. Consistent with this observation, the phenotype, function and age-dependent expansion of 'virtual memory' CD8(+) T cells are strongly affected in absence of type I interferon signalling. In addition, type I interferons induce a sustained expansion of 'virtual memory' CD8(+) T cells in an Eomes-dependent fashion. We further show that the development of 'innate thymic' CD8(+) T cells is dependent on the same pathway. In conclusion, we demonstrate that type I interferon signalling in CD8(+) T cells drives Eomes expression and thereby regulates the function and homeostasis of memory-like CD8(+) T cells.
Collapse
|
27
|
Huang W, August A. The signaling symphony: T cell receptor tunes cytokine-mediated T cell differentiation. J Leukoc Biol 2015; 97:477-85. [PMID: 25525115 PMCID: PMC4338847 DOI: 10.1189/jlb.1ri0614-293r] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 11/03/2014] [Accepted: 11/13/2014] [Indexed: 01/07/2023] Open
Abstract
T cell development, differentiation, and maintenance are orchestrated by 2 key signaling axes: the antigen-specific TCR and cytokine-mediated signals. The TCR signals the recognition of self- and foreign antigens to control T cell homeostasis for immune tolerance and immunity, which is regulated by a variety of cytokines to determine T cell subset homeostasis and differentiation. TCR signaling can synergize with or antagonize cytokine-mediated signaling to fine tune T cell fate; however, the latter is less investigated. Murine models with attenuated TCR signaling strength have revealed that TCR signaling can function as regulatory feedback machinery for T cell homeostasis and differentiation in differential cytokine milieus, such as IL-2-mediated Treg development; IL-7-mediated, naïve CD8(+) T cell homeostasis; and IL-4-induced innate memory CD8(+) T cell development. In this review, we discuss the symphonic cross-talk between TCR and cytokine-mediated responses that differentially control T cell behavior, with a focus on the negative tuning by TCR activation on the cytokine effects.
Collapse
Affiliation(s)
- Weishan Huang
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| |
Collapse
|