1
|
Matar AM, Shehata WA, Kora MA, Shendi SS. Tissue and circulating levels of IL-17A and FoxP3 + in patients with scabies: Correlation with clinical features. Mol Biochem Parasitol 2024; 260:111652. [PMID: 39209219 DOI: 10.1016/j.molbiopara.2024.111652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/31/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The scabies mite is known to induce a complicated immune response that involves both innate and long-term adaptive immunity. Many immune effectors and pathways are involved. Th17/Treg balance can influence the complex immune response to scabies. The immunological effectors including IL-17A, as a pro-inflammatory cytokine, and Treg cells, anti-inflammatory regulatory T cells, are essential for preserving cutaneous immunological homeostasis. So, evaluating these immune effectors may help in comprehending the pathophysiology of scabies and facilitate the development of new treatment approaches. This study examined the expression of IL-17A and FoxP3+ in the skin and serum of 50 scabies patients and 25 healthy controls. An assessment of their correlation with clinical features was performed. Regarding tissue response, scabietic patients exhibited a significant increase in IL-17A and FoxP3+ expression in their epidermis and dermis compared to controls (P<0.001), but the correlation between these factors was not significant in either area (P>0.05). Also, patients showed a significant increase in serum IL-17A levels compared to controls (P<0.001), with a significant association between serum IL-17A levels and lesion severity, but no significant correlation was observed between skin and serum responses (P>0.05). In conclusion, there was increased expression of both IL-17A and FoxP3+, with FoxP3+ being significantly more abundant than IL-17A in the skin of scabies patients. Skin FoxP3+ up-regulation has been linked to the severity of the condition.
Collapse
Affiliation(s)
- Amira M Matar
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Menoufia 6132720, Egypt.
| | - Wafaa A Shehata
- Department of Dermatology and Andrology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Menoufia 6132720, Egypt.
| | - Mona A Kora
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Menoufia 6132720, Egypt.
| | - Sawsan S Shendi
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin al-Kom, Menoufia 6132720, Egypt.
| |
Collapse
|
2
|
Yi P, Yu W, Xiong Y, Dong Y, Huang Q, Lin Y, Du Y, Hua F. IL-35: New Target for Immunotherapy Targeting the Tumor Microenvironment. Mol Cancer Ther 2024; 23:148-158. [PMID: 37988561 DOI: 10.1158/1535-7163.mct-23-0242] [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: 04/26/2023] [Revised: 08/15/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Interleukin 35(IL-35) is a newly discovered inhibitory cytokine of the IL12 family. More recently, IL-35 was found to be increased in the tumor microenvironment (TME) and peripheral blood of many patients with cancer, indicating that it plays an important role in the TME. Tumors secrete cytokines that recruit myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) into the TME to promote malignant progression, which is a great challenge for cancer treatment. Radiotherapy causes serious adverse effects, and tumor resistance to immune checkpoint inhibitors is still an unsolved challenge. Thus, new cancer therapy approaches are urgently needed. Numerous studies have shown that IL-35 can recruit immunosuppressive cells to enable tumor immune escape by promoting the conversion of immune cells into a tumor growth-promoting phenotype as well as facilitating tumor angiogenesis. IL-35-neutralizing antibodies were found to boost the chemotherapeutic effect of gemcitabine and considerably reduce the microvascular density of pancreatic cancer in mice. Therefore, targeting IL-35 in the TME provides a promising cancer treatment target. In addition, IL-35 may be used as an independent prognostic factor for some tumors in the near future. This review intends to reveal the interplay of IL-35 with immune cells in the TME, which may provide new options for the treatment of cancer.
Collapse
Affiliation(s)
- Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wenjun Yu
- Fuzhou First People's Hospital of Jiangxi Province, Fuzhou City, Jiangxi Province, P.R. China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Qiang Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yunfei Du
- Department of Anesthesiology, Nanchang Central Hospital, Nanchang, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
3
|
Wu Q, Pan C, Zhou Y, Wang S, Xie L, Zhou W, Ding L, Chen T, Qian J, Su R, Gao X, Mei Z, Qiao Y, Yin S, Wu Y, Wang J, Zhou L, Zheng S. Targeting neuropilin-1 abolishes anti-PD-1-upregulated regulatory T cells and synergizes with 4-1BB agonist for liver cancer treatment. Hepatology 2023; 78:1402-1417. [PMID: 36811396 DOI: 10.1097/hep.0000000000000320] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/09/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND AIMS Regulatory T cells (Tregs) are an obstacle to PD-1 blockade-mediated antitumor efficacy. However, the behaviors of Tregs response to anti-PD-1 in HCC and the characteristics of Tregs tissue adaptation from peripheral lymphoid tissues to the tumor are still unclear. APPROACH RESULTS Here, we determine that PD-1 monotherapy potentially augments the accumulation of tumor CD4 + Tregs. Mechanistically, anti-PD-1 mediates Tregs proliferation in lymphoid tissues rather than in the tumor. Increased peripheral Tregs burden replenishes intratumoral Tregs, raising the ratio of intratumoral CD4 + Tregs to CD8 + T cells. Subsequently, single-cell transcriptomics revealed that neuropilin-1 (Nrp-1) supports Tregs migration behavior, and the genes of Crem and Tnfrsf9 regulate the behaviors of the terminal suppressive Tregs. Nrp-1 + 4-1BB - Tregs stepwise develop to the Nrp-1 - 4-1BB + Tregs from lymphoid tissues into the tumor. Moreover, Treg-restricted Nrp1 depletion abolishes anti-PD-1-upregulated intratumoral Tregs burden and synergizes with the 4-1BB agonist to enhance the antitumor response. Finally, a combination of the Nrp-1 inhibitor and the 4-1BB agonist in humanized HCC models showed a favorable and safe outcome and evoked the antitumor effect of the PD-1 blockade. CONCLUSION Our findings elucidate the potential mechanism of anti-PD-1-mediated intratumoral Tregs accumulation in HCC and uncover the tissue adaptation characteristics of Tregs and identify the therapeutic potential of targeting Nrp-1 and 4-1BB for reprogramming the HCC microenvironment.
Collapse
Affiliation(s)
- Qinchuan Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Caixu Pan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yuan Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Liting Xie
- Department of Ultrasound, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wuhua Zhou
- Department of Hepatobiliary Pancreatic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Limin Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Tianchi Chen
- Department of vascular surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Rong Su
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Xingxing Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Zhibin Mei
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Shengyong Yin
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yi Wu
- Lyvgen Biopharma, Shanghai, China
| | | | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Wang Y, Sun P, Hao X, Cao D, Liu J, Zhang D. Decreased DIO3OS Expression Predicts Poor Prognosis in Hepatocellular Carcinoma and is Associated with Immune Infiltration. Biochem Genet 2023; 61:1791-1806. [PMID: 36802306 DOI: 10.1007/s10528-023-10345-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/02/2023] [Indexed: 02/23/2023]
Abstract
Hepatocellular carcinoma has become one of the most shared cancers in the whole world because of its high morbidity, poor survival rate, and low recovery rate. LncRNA DIO3 opposite strand upstream RNA (DIO3OS) has been reported to be obviously important in several human cancers, while its biological function in hepatocellular carcinoma (HCC) remains unclear. Here, DIO3OS gene expression data and clinical information of HCC patients were extracted from the Cancer Genome Atlas (TCGA) database and the university of California Santa Cruz (UCSC) Xena database. In our study, the Wilcoxon rank sum test was used to compare DIO3OS expression between healthy individuals and HCC patients. It was found that patients with HCC had significantly lower DIO3OS expression than healthy individuals. Furthermore, Kaplan-Meier curves and Cox regression analysis showed that high DIO3OS expression tended to predict better prognosis and higher survival rate in HCC patients. In addition, the gene set enrichment analysis (GSEA) assay was used to annotate the biological function of DIO3OS. It was found that DIO3OS was significantly correlated with immune invasion in HCC. This was also aided by the subsequent ESTIMATE assay. Our study provides a novel biomarker and therapeutic strategy for patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yunhan Wang
- Department of Pathology and Pathophysiology, Weifang Medical University, Weifang, 261053, Shandong Province, China
| | - Ping Sun
- Department of Immunology, Weifang Medical University, Weifang, 261053, Shandong Province, China
| | - Xinping Hao
- Department of Intensive Care Unit, Weifang Traditional Chinese Medicine Hospital, Weifang, 261041, Shandong Province, China
| | - Daihong Cao
- Dpartment of Pathology, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, 030000, Shanxi Province, China
| | - Jiangyue Liu
- Department of Pathology and Pathophysiology, Weifang Medical University, Weifang, 261053, Shandong Province, China.
| | - Daijuan Zhang
- Department of Pathology and Pathophysiology, Weifang Medical University, Weifang, 261053, Shandong Province, China.
| |
Collapse
|
5
|
Park SH, Eun R, Heo J, Lim YT. Nanoengineered drug delivery in cancer immunotherapy for overcoming immunosuppressive tumor microenvironment. Drug Deliv Transl Res 2023; 13:2015-2031. [PMID: 36581707 DOI: 10.1007/s13346-022-01282-8] [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] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
Almost like a living being in and of itself, tumors actively interact with and modify their environment to escape immune responses. Owing to the pre-formation of cancer-favorable microenvironment prior to anti-cancer treatment, the numerous attempts that followed propose limited efficacy in oncology. Immunogenicity by activation of immune cells within the tumor microenvironment or recruitment of immune cells from nearby lymph nodes is quickly offset as the immunosuppressive environment, rapidly converting immunogenic cells into immune suppressive cells, overriding the immune system. Tumor cells, as well as regulatory cells, namely M2 macrophages, Treg cells, and MDSCs, derived by the immunosuppressive environment, also cloak from potential anti-tumoral factors by directly or indirectly secreting cytokines, such as IL-10 and TGF-β, related to immune regulation. Enzymes and other metabolic or angiogenetic constituents - VEGF, IDO1, and iNOS - are also employed directed for anti-cancer immune cell malfunctioning. Therefore, the conversion of "cold" immunosuppressive environment into "hot" immune responsive environment is of paramount importance, bestowing the advances in the field of cancer immunotherapy the opportunity to wholly fulfill its intended purpose. This paper reviews the mechanisms by which tumors wield to exercise immune suppression and the nanoengineered delivery strategies being developed to overcome this suppression.
Collapse
Affiliation(s)
- Sei Hyun Park
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Ryounho Eun
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Janghun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea.
| |
Collapse
|
6
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
7
|
McGee MC, Zhang T, Magazine N, Islam R, Carossino M, Huang W. PD-1 and ICOS counter-regulate tissue resident regulatory T cell development and IL-10 production during flu. Front Immunol 2022; 13:984476. [PMID: 36159872 PMCID: PMC9492985 DOI: 10.3389/fimmu.2022.984476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022] Open
Abstract
Regulatory T cells that express the transcription factor Foxp3 (Treg cells) are a highly heterogenous population of immunoregulatory cells critical for maintaining immune homeostasis and preventing immunopathology during infections. Tissue resident Treg (TR-Treg) cells are maintained within nonlymphoid tissues and have been shown to suppress proinflammatory tissue resident T cell responses and promote tissue repair. Human populations are repetitively exposed to influenza infections and lung tissue resident effector T cell responses are associated with flu-induced long-term pulmonary sequelae. The kinetics of TR-Treg cell development and molecular features of TR-Treg cells during repeated and/or long-term flu infections are unclear. Utilizing a Foxp3RFP/IL-10GFP dual reporter mouse model along with intravascular fluorescent in vivo labeling, we characterized the TR-Treg cell responses to repetitive heterosubtypic influenza infections. We found lung tissue resident Treg cells accumulated and expressed high levels of co-inhibitory and co-stimulatory receptors post primary and secondary infections. Blockade of PD-1 or ICOS signaling reveals that PD-1 and ICOS signaling pathways counter-regulate TR-Treg cell expansion and IL-10 production, during secondary influenza infection. Furthermore, the virus-specific TR-Treg cell response displayed distinct kinetics, when compared to conventional CD4+ tissue resident memory T cells, during secondary flu infection. Our results provide insight into the tissue resident Foxp3+ regulatory T cell response during repetitive flu infections, which may be applicable to other respiratory infectious diseases such as tuberculosis and COVID.
Collapse
Affiliation(s)
- Michael C. McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Tianyi Zhang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Nicholas Magazine
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Rezwanul Islam
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 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
- *Correspondence: Weishan Huang,
| |
Collapse
|
8
|
Hippen KL, Hefazi M, Larson JH, Blazar BR. Emerging translational strategies and challenges for enhancing regulatory T cell therapy for graft-versus-host disease. Front Immunol 2022; 13:926550. [PMID: 35967386 PMCID: PMC9366169 DOI: 10.3389/fimmu.2022.926550] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 02/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for many types of cancer. Genetic disparities between donor and host can result in immune-mediated attack of host tissues, known as graft versus host disease (GVHD), a major cause of morbidity and mortality following HSCT. Regulatory CD4+ T cells (Tregs) are a rare cell type crucial for immune system homeostasis, limiting the activation and differentiation of effector T cells (Teff) that are self-reactive or stimulated by foreign antigen exposure. Adoptive cell therapy (ACT) with Treg has demonstrated, first in murine models and now in patients, that prophylactic Treg infusion can also suppress GVHD. While clinical trials have demonstrated Treg reduce severe GVHD occurrence, several impediments remain, including Treg variability and practical need for individualized Treg production for each patient. Additionally, there are challenges in the use of in vitro expansion techniques and in achieving in vivo Treg persistence in context of both immune suppressive drugs and in lymphoreplete patients being treated for GVHD. This review will focus on 3 main translational approaches taken to improve the efficacy of tTreg ACT in GVHD prophylaxis and development of treatment options, following HSCT: genetic modification, manipulating TCR and cytokine signaling, and Treg production protocols. In vitro expansion for Treg ACT presents a multitude of approaches for gene modification to improve efficacy, including: antigen specificity, tissue targeting, deletion of negative regulators/exhaustion markers, resistance to immunosuppressive drugs common in GVHD treatment. Such expansion is particularly important in patients without significant lymphopenia that can drive Treg expansion, enabling a favorable Treg:Teff ratio in vivo. Several potential therapeutics have also been identified that enhance tTreg stability or persistence/expansion following ACT that target specific pathways, including: DNA/histone methylation status, TCR/co-stimulation signaling, and IL-2/STAT5 signaling. Finally, this review will discuss improvements in Treg production related to tissue source, Treg subsets, therapeutic approaches to increase Treg suppression and stability during tTreg expansion, and potential for storing large numbers of Treg from a single production run to be used as an off-the-shelf infusion product capable of treating multiple recipients.
Collapse
Affiliation(s)
- Keli L. Hippen
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Jemma H. Larson
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Bruce R. Blazar
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| |
Collapse
|
9
|
Han Z, Ma K, Tao H, Liu H, Zhang J, Sai X, Li Y, Chi M, Nian Q, Song L, Liu C. A Deep Insight Into Regulatory T Cell Metabolism in Renal Disease: Facts and Perspectives. Front Immunol 2022; 13:826732. [PMID: 35251009 PMCID: PMC8892604 DOI: 10.3389/fimmu.2022.826732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Kidney disease encompasses a complex set of diseases that can aggravate or start systemic pathophysiological processes through their complex metabolic mechanisms and effects on body homoeostasis. The prevalence of kidney disease has increased dramatically over the last two decades. CD4+CD25+ regulatory T (Treg) cells that express the transcription factor forkhead box protein 3 (Foxp3) are critical for maintaining immune homeostasis and preventing autoimmune disease and tissue damage caused by excessive or unnecessary immune activation, including autoimmune kidney diseases. Recent studies have highlighted the critical role of metabolic reprogramming in controlling the plasticity, stability, and function of Treg cells. They are also likely to play a vital role in limiting kidney transplant rejection and potentially promoting transplant tolerance. Metabolic pathways, such as mitochondrial function, glycolysis, lipid synthesis, glutaminolysis, and mammalian target of rapamycin (mTOR) activation, are involved in the development of renal diseases by modulating the function and proliferation of Treg cells. Targeting metabolic pathways to alter Treg cells can offer a promising method for renal disease therapy. In this review, we provide a new perspective on the role of Treg cell metabolism in renal diseases by presenting the renal microenvironment、relevant metabolites of Treg cell metabolism, and the role of Treg cell metabolism in various kidney diseases.
Collapse
Affiliation(s)
- Zhongyu Han
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hongxia Tao
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongli Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiong Zhang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xiyalatu Sai
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Yunlong Li
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qing Nian
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Department of Blood Transfusion Sicuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
10
|
Saini N, Lakshminarayanan S, Kundu P, Sarin A. Notch1 Modulation of Cellular Calcium Regulates Mitochondrial Metabolism and Anti-Apoptotic Activity in T-Regulatory Cells. Front Immunol 2022; 13:832159. [PMID: 35222416 PMCID: PMC8866856 DOI: 10.3389/fimmu.2022.832159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/21/2022] [Indexed: 01/04/2023] Open
Abstract
As the major hub of metabolic activity and an organelle sequestering pro-apoptogenic intermediates, mitochondria lie at the crossroads of cellular decisions of death and survival. Intracellular calcium is a key regulator of these outcomes with rapid, uncontrolled uptake into mitochondria, activating pro-apoptotic cascades that trigger cell death. Here, we show that calcium uptake and mitochondrial metabolism in murine T-regulatory cells (Tregs) is tuned by Notch1 activity. Based on analysis of Tregs and the HEK cell line, we present evidence that modulation of cellular calcium dynamics underpins Notch1 regulation of mitochondrial homeostasis and consequently anti-apoptotic activity. Targeted siRNA-mediated ablations reveal dependency on molecules controlling calcium release from the endoplasmic reticulum (ER) and the chaperone, glucose-regulated protein 75 (Grp75), the associated protein Voltage Dependent Anion Channel (VDAC)1 and the Mitochondrial Calcium Uniporter (MCU), which together facilitate ER calcium transfer and uptake into the mitochondria. Endogenous Notch1 is detected in immune-complexes with Grp75 and VDAC1. Deficits in mitochondrial oxidative and survival in Notch1 deficient Tregs, were corrected by the expression of recombinant Notch1 intracellular domain, and in part by recombinant Grp75. Thus, the modulation of calcium dynamics and consequently mitochondrial metabolism underlies Treg survival in conditions of nutrient stress. This work positions a key role for Notch1 activity in these outcomes.
Collapse
Affiliation(s)
- Neetu Saini
- Regulation of Cell Fate, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, India.,Department of Biology, Manipal Academy of Higher Education, Manipal, India
| | - Sowmya Lakshminarayanan
- National Centre for Biological Science, TATA Institute of Fundamental Research (TIFR), Bengaluru, India
| | - Priyanka Kundu
- National Centre for Biological Science, TATA Institute of Fundamental Research (TIFR), Bengaluru, India
| | - Apurva Sarin
- Regulation of Cell Fate, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, India
| |
Collapse
|
11
|
Grover P, Goel PN, Greene MI. Regulatory T Cells: Regulation of Identity and Function. Front Immunol 2021; 12:750542. [PMID: 34675933 PMCID: PMC8524049 DOI: 10.3389/fimmu.2021.750542] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
T regulatory cells suppress a variety of immune responses to self-antigens and play a role in peripheral tolerance maintenance by limiting autoimmune disorders, and other pathological immune responses such as limiting immune reactivity to oncoprotein encoded antigens. Forkhead box P3 (FOXP3) expression is required for Treg stability and affects functional activity. Mutations in the master regulator FOXP3 and related components have been linked to autoimmune diseases in humans, such as IPEX, and a scurfy-like phenotype in mice. Several lines of evidence indicate that Treg use a variety of immunosuppressive mechanisms to limit an immune response by targeting effector cells, including secretion of immunoregulatory cytokines, granzyme/perforin-mediated cell cytolysis, metabolic perturbation, directing the maturation and function of antigen-presenting cells (APC) and secretion of extracellular vesicles for the development of immunological tolerance. In this review, several regulatory mechanisms have been highlighted and discussed.
Collapse
Affiliation(s)
- Payal Grover
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peeyush N Goel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Nicholls J, Cao B, Le Texier L, Xiong LY, Hunter CR, Llanes G, Aguliar EG, Schroder WA, Phipps S, Lynch JP, Cao H, Heazlewood SY, Williams B, Clouston AD, Nefzger CM, Polo JM, Nilsson SK, Blazar BR, MacDonald KPA. Bone Marrow Regulatory T Cells Are a Unique Population, Supported by Niche-Specific Cytokines and Plasmacytoid Dendritic Cells, and Required for Chronic Graft-Versus-Host Disease Control. Front Cell Dev Biol 2021; 9:737880. [PMID: 34631716 PMCID: PMC8493124 DOI: 10.3389/fcell.2021.737880] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Regulatory T cell (Treg) reconstitution is essential for reestablishing tolerance and maintaining homeostasis following stem-cell transplantation. We previously reported that bone marrow (BM) is highly enriched in autophagy-dependent Treg and autophagy disruption leads to a significant Treg loss, particularly BM-Treg. To correct the known Treg deficiency observed in chronic graft-versus-host disease (cGVHD) patients, low dose IL-2 infusion has been administered, substantially increasing peripheral Treg (pTreg) numbers. However, as clinical responses were only seen in ∼50% of patients, we postulated that pTreg augmentation was more robust than for BM-Treg. We show that BM-Treg and pTreg have distinct characteristics, indicated by differential transcriptome expression for chemokine receptors, transcription factors, cell cycle control of replication and genes linked to Treg function. Further, BM-Treg were more quiescent, expressed lower FoxP3, were highly enriched for co-inhibitory markers and more profoundly depleted than splenic Treg in cGVHD mice. In vivo our data are consistent with the BM and not splenic microenvironment is, at least in part, driving this BM-Treg signature, as adoptively transferred splenic Treg that entered the BM niche acquired a BM-Treg phenotype. Analyses identified upregulated expression of IL-9R, IL-33R, and IL-7R in BM-Treg. Administration of the T cell produced cytokine IL-2 was required by splenic Treg expansion but had no impact on BM-Treg, whereas the converse was true for IL-9 administration. Plasmacytoid dendritic cells (pDCs) within the BM also may contribute to BM-Treg maintenance. Using pDC-specific BDCA2-DTR mice in which diptheria toxin administration results in global pDC depletion, we demonstrate that pDC depletion hampers BM, but not splenic, Treg homeostasis. Together, these data provide evidence that BM-Treg and splenic Treg are phenotypically and functionally distinct and influenced by niche-specific mediators that selectively support their respective Treg populations. The unique properties of BM-Treg should be considered for new therapies to reconstitute Treg and reestablish tolerance following SCT.
Collapse
Affiliation(s)
- Jemma Nicholls
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Laetitia Le Texier
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Laura Yan Xiong
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christopher R. Hunter
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Genesis Llanes
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ethan G. Aguliar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Wayne A. Schroder
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Simon Phipps
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jason P. Lynch
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Huimin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Shen Y. Heazlewood
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Brenda Williams
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | | | - Christian M. Nefzger
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jose M. Polo
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Susan K. Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kelli P. A. MacDonald
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Kawai K, Uchiyama M, Hester J, Issa F. IL-33 drives the production of mouse regulatory T cells with enhanced in vivo suppressive activity in skin transplantation. Am J Transplant 2021; 21:978-992. [PMID: 33314772 PMCID: PMC7613121 DOI: 10.1111/ajt.16266] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 01/25/2023]
Abstract
Regulatory T cells (Tregs) are crucial mediators of immune homeostasis with the ability to modulate allogeneic response and control transplant rejection. Although Treg-based cell therapies have shown immense promise, methods to optimize current strategies are critical for successful implementation within the clinic. IL-33 is a cytokine with pleiotropic properties and effects on Treg function and development. In this study, we explored the unique properties of Treg populations activated through the IL-33/ST2 pathway, aiming to exploit their tolerogenic properties for cell therapy. We show that treatment with exogenous IL-33 results in a generalized downregulation of genes critical to T cell biology together with an upregulation of Treg-associated genes. Tregs that develop in response to IL-33 upregulate critical Treg-associated markers, yet without developing enhanced in vitro suppressive capacity. Conversely, these Tregs display potent regulatory activity in vivo, promoting long-term skin allograft survival in a stringent transplantation model. Detailed transcriptomic and immunophenotypic analyses of IL-33-expanded Tregs reveal an enhancement in graft-homing chemokine receptors, which may be partly responsible for their superior in vivo activity that is not reflected in vitro. IL-33 treatment is therefore an attractive adjunctive strategy for patients receiving Treg cell therapeutics.
Collapse
Affiliation(s)
- Kento Kawai
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Masateru Uchiyama
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK,Department of Surgery, Teikyo University, Tokyo, Japan
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
Thorsen SU, Collier F, Pezic A, O'Hely M, Clarke M, Tang MLK, Burgner D, Vuillermin P, Ponsonby AL. Maternal and Cord Blood 25-Hydroxyvitamin D 3 Are Associated with Increased Cord Blood and Naive and Activated Regulatory T Cells: The Barwon Infant Study. THE JOURNAL OF IMMUNOLOGY 2021; 206:874-882. [PMID: 33431661 DOI: 10.4049/jimmunol.2000515] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
Vitamin D has shown immune-modulatory effects but mostly in in vitro and animal studies. Regulatory T cells (Treg) are important for a balanced immune system. The relationship between vitamin D on the number of circulating neonatal Treg is unclear. We sought to investigate the association between maternal and neonatal vitamin D metabolites and cord blood (CB) Treg subsets. In a cohort of Australian infants (n = 1074), recruited using an unselected antenatal sampling frame, 158 mother-infant pairs had data on the following: 1) 25-hydroxyvitamin D3 (25(OH)D3) measures in both maternal peripheral blood (28- to 32-wk gestation) and infant CB; 2) proportions (percentage of CD4+ T cells) of CB Treg subsets (CD4+CD45RA+ FOXP3low naive Treg, and CD4+CD45RA- FOXP3high activated Treg [aTreg]); and 3) possible confounders, including maternal personal UV radiation. Multiple regression analyses were used. The median 25(OH)D3 was 85.4 and 50.7 nmol/l for maternal and CB samples, respectively. Higher maternal 25(OH)D3 levels were associated with increased CB naive Treg (relative adjusted mean difference [AMD] per 25 nmol/l increase: 5%; 95% confidence interval [CI]: 1-9%), and aTreg (AMD per 25 nmol/l increase: 17%; 95% CI: 6-28%). Furthermore, a positive association between CB 25(OH)D3 levels and CB aTreg (AMD per 25 nmol/l increase: 29%; 95% CI: 13-48%) was also evident. These results persisted after adjustment for other factors such as maternal personal UV radiation and season of birth. 25(OH)D3, may play a role in the adaptive neonatal immune system via induction of FOXP3+ Tregs. Further studies of immune priming actions of antenatal 25(OH)D3 are warranted.
Collapse
Affiliation(s)
- Steffen U Thorsen
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev and Gentofte University Hospital, 2730 Herlev, Denmark.,Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Fiona Collier
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia.,Geelong Center for Emerging Infectious Diseases Laboratory, University Hospital, Barwon Health, Geelong, Victoria 3220, Australia.,Child Research Unit, University Hospital, Barwon Health, Geelong, Victoria 3220, Australia.,School of Medicine, Deakin University, Geelong, Victoria 3220, Australia
| | - Angela Pezic
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Martin O'Hely
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Michael Clarke
- Biological and Molecular Mass Spectrometry Facility, Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth, Western Australia 6009, Australia; and
| | - Mimi L K Tang
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Peter Vuillermin
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia.,Child Research Unit, University Hospital, Barwon Health, Geelong, Victoria 3220, Australia.,School of Medicine, Deakin University, Geelong, Victoria 3220, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia; .,The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | | |
Collapse
|
15
|
Alperin M. Deeper dive into tissue-level events that incite and promote pelvic organ prolapse. Am J Obstet Gynecol 2021; 224:1-2. [PMID: 33386102 DOI: 10.1016/j.ajog.2020.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 10/22/2022]
|
16
|
Tissue-Resident Lymphocytes: Implications in Immunotherapy for Hepatocellular Carcinoma. Int J Mol Sci 2020; 22:ijms22010232. [PMID: 33379384 PMCID: PMC7796120 DOI: 10.3390/ijms22010232] [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: 11/15/2020] [Revised: 12/16/2020] [Accepted: 12/25/2020] [Indexed: 12/29/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a hard-to-treat cancer. The recent introduction of immune checkpoint inhibitors (ICIs) provided viable options to treat HCC, but the response rate is currently not sufficient. Thus, a better understanding of ICI-responding cells within tumors is needed to improve outcomes of ICI treatment in HCC. Recently, tissue-resident memory T (TRM) cells were defined as a subset of the memory T cell population; this cell population is actively under investigation to elucidate its role in anti-tumor immunity. In addition, the role of other tissue-resident populations such as tissue resident regulatory T (Treg) cells, mucosal associated invariant T (MAIT) cells, γδ T cells, and invariant natural killer T (iNKT) cells in anti-tumor immunity is also actively being investigated. However, there is no study that summarizes recent studies and discusses future perspectives in terms of tissue resident lymphocytes in HCC. In this review, we summarize key features of tissue-resident lymphocytes and their role in the anti-tumor immunity. Additionally, we review recent studies regarding the characteristics of tissue-resident lymphocytes in HCC and their role in ICI treatment and other immunotherapeutic strategies.
Collapse
|
17
|
Camacho V, Matkins VR, Patel SB, Lever JM, Yang Z, Ying L, Landuyt AE, Dean EC, George JF, Yang H, Ferrell PB, Maynard CL, Weaver CT, Turnquist HR, Welner RS. Bone marrow Tregs mediate stromal cell function and support hematopoiesis via IL-10. JCI Insight 2020; 5:135681. [PMID: 33208555 PMCID: PMC7710301 DOI: 10.1172/jci.insight.135681] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
The nonimmune roles of Tregs have been described in various tissues, including the BM. In this study, we comprehensively phenotyped marrow Tregs, elucidating their key features and tissue-specific functions. We show that marrow Tregs are migratory and home back to the marrow. For trafficking, marrow Tregs use S1P gradients, and disruption of this axis allows for specific targeting of the marrow Treg pool. Following Treg depletion, the function and phenotype of both mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) was impaired. Transplantation also revealed that a Treg-depleted niche has a reduced capacity to support hematopoiesis. Finally, we found that marrow Tregs are high producers of IL-10 and that Treg-secreted IL-10 has direct effects on MSC function. This is the first report to our knowledge revealing that Treg-secreted IL-10 is necessary for stromal cell maintenance, and our work outlines an alternative mechanism by which this cytokine regulates hematopoiesis.
Collapse
Affiliation(s)
| | | | | | - Jeremie M. Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, and
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li Ying
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Ashley E. Landuyt
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emma C. Dean
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Henry Yang
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Paul Brent Ferrell
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
18
|
Lymphocyte Immunosuppression and Dysfunction Contributing to Persistent Inflammation, Immunosuppression, and Catabolism Syndrome (PICS). Shock 2020; 55:723-741. [PMID: 33021569 DOI: 10.1097/shk.0000000000001675] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
ABSTRACT Persistent Inflammation, Immune Suppression, and Catabolism Syndrome (PICS) is a disease state affecting patients who have a prolonged recovery after the acute phase of a large inflammatory insult. Trauma and sepsis are two pathologies after which such an insult evolves. In this review, we will focus on the key clinical determinants of PICS: Immunosuppression and cellular dysfunction. Currently, relevant immunosuppressive functions have been attributed to both innate and adaptive immune cells. However, there are significant gaps in our knowledge, as for trauma and sepsis the immunosuppressive functions of these cells have mostly been described in acute phase of inflammation so far, and their clinical relevance for the development of prolonged immunosuppression is mostly unknown. It is suggested that the initial immune imbalance determines the development of PCIS. Additionally, it remains unclear what distinguishes the onset of immune dysfunction in trauma and sepsis and how this drives immunosuppression in these cells. In this review, we will discuss how regulatory T cells (Tregs), innate lymphoid cells, natural killer T cells (NKT cells), TCR-a CD4- CD8- double-negative T cells (DN T cells), and B cells can contribute to the development of post-traumatic and septic immunosuppression. Altogether, we seek to fill a gap in the understanding of the contribution of lymphocyte immunosuppression and dysfunction to the development of chronic immune disbalance. Further, we will provide an overview of promising diagnostic and therapeutic interventions, whose potential to overcome the detrimental immunosuppression after trauma and sepsis is currently being tested.
Collapse
|
19
|
Mannie MD, DeOca KB, Bastian AG, Moorman CD. Tolerogenic vaccines: Targeting the antigenic and cytokine niches of FOXP3 + regulatory T cells. Cell Immunol 2020; 355:104173. [PMID: 32712270 PMCID: PMC7444458 DOI: 10.1016/j.cellimm.2020.104173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
FOXP3+ regulatory T cells (Tregs) constitute a critical barrier that enforces tolerance to both the self-peptidome and the extended-self peptidome to ensure tissue-specific resistance to autoimmune, allergic, and other inflammatory disorders. Here, we review intuitive models regarding how T cell antigen receptor (TCR) specificity and antigen recognition efficiency shape the Treg and conventional T cell (Tcon) repertoires to adaptively regulate T cell maintenance, tissue-residency, phenotypic stability, and immune function in peripheral tissues. Three zones of TCR recognition efficiency are considered, including Tcon recognition of specific low-efficiency self MHC-ligands, Treg recognition of intermediate-efficiency agonistic self MHC-ligands, and Tcon recognition of cross-reactive high-efficiency agonistic foreign MHC-ligands. These respective zones of TCR recognition efficiency are key to understanding how tissue-resident immune networks integrate the antigenic complexity of local environments to provide adaptive decisions setting the balance of suppressive and immunogenic responses. Importantly, deficiencies in the Treg repertoire appear to be an important cause of chronic inflammatory disease. Deficiencies may include global deficiencies in Treg numbers or function, subtle 'holes in the Treg repertoire' in tissue-resident Treg populations, or simply Treg insufficiencies that are unable to counter an overwhelming molecular mimicry stimulus. Tolerogenic vaccination and Treg-based immunotherapy are two therapeutic modalities meant to restore dominance of Treg networks to reverse chronic inflammatory disease. Studies of these therapeutic modalities in a preclinical setting have provided insight into the Treg niche, including the concept that intermediate-efficiency TCR signaling, high IFN-β concentrations, and low IL-2 concentrations favor Treg responses and active dominant mechanisms of immune tolerance. Overall, the purpose here is to assimilate new and established concepts regarding how cognate TCR specificity of the Treg repertoire and the contingent cytokine networks provide a foundation for understanding Treg suppressive strategy.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| | - Kayla B DeOca
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Alexander G Bastian
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| |
Collapse
|
20
|
Kaufman CL, Kanitakis J, Weissenbacher A, Brandacher G, Mehra MR, Amer H, Zelger BG, Zelger B, Pomahac B, McDiarmid S, Cendales L, Morelon E. Defining chronic rejection in vascularized composite allotransplantation-The American Society of Reconstructive Transplantation and International Society of Vascularized Composite Allotransplantation chronic rejection working group: 2018 American Society of Reconstructive Transplantation meeting report and white paper Research goals in defining chronic rejection in vascularized composite allotransplantation. SAGE Open Med 2020; 8:2050312120940421. [PMID: 32704373 PMCID: PMC7361482 DOI: 10.1177/2050312120940421] [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: 06/15/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives: This report summarizes a collaborative effort between the American Society of Reconstructive Transplantation and the International Society of Vascularized Composite Allotransplantation to establish what is known about chronic rejection in recipients of vascularized composite allografts, with an emphasis on upper extremity and face transplants. As a picture of chronic rejection in hand and face vascularized composite allografts emerges, the results will be applied to other types of vascularized composite allografts, such as uterine transplantation. Methods: The overall goal is to develop a definition of chronic rejection in vascularized composite allografts so that we can establish longitudinal correlates of factors such as acute rejection, immunosuppressive therapy, de novo donor-specific antibody and trauma/infection and other external factors on the development of chronic rejection. As Dr Kanitakis eloquently stated at the 2017 International Society of Vascularized Composite Allotransplantation meeting in Salzburg, “Before we can correlate causative factors of chronic rejection, we have to define what chronic rejection in VCA is.” Results: The first meeting report was presented at the sixth Biennial meeting of the American Society of Reconstructive Transplantation in November 2018. Based on collaborative efforts and descriptions of clinical cases of chronic rejection in vascularized composite allograft recipients, a working definition of chronic rejection in vascularized composite allografts with respect to overt functional decline, subclinical functional decline, histologic evidence without functional decline, and normal allograft function in the absence of histologic evidence of chronic rejection is proposed. Conclusions: It is the intent of this collaborative working group that these working definitions will help to focus ongoing research to define the incidence, risk factors and treatment regimens that will identify mechanisms of chronic rejection in vascularized composite allografts. As with all good research, our initial efforts have generated more questions than answers. We hope that this is the first of many updates.
Collapse
Affiliation(s)
| | - Jean Kanitakis
- Department of Dermatology, Ed. Herriot Hospital, Lyon, France
| | | | | | | | | | | | | | | | - Sue McDiarmid
- Ronald Reagan UCLA Medical Center, Los Angeles, CA, USA
| | | | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Ed. Herriot Hospital, Lyon, France
| |
Collapse
|
21
|
Putilin DA, Evchenko SY, Fedoniuk LY, Tokarskyy OS, Kamyshny OM, Migenko LM, Andreychyn SM, Hanberher II, Bezruk TO. The Influence of Metformin to the Transcriptional Activity of the mTOR and FOX3 Genes in Parapancreatic Adipose Tissue of Streptozotocin-Induced Diabetic Rats. J Med Life 2020; 13:50-55. [PMID: 32341701 PMCID: PMC7175427 DOI: 10.25122/jml-2020-0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mammalian target of rapamycin is not only a central regulator of lipid metabolism that controls the processes of adipogenesis and lipolysis but also a regulator of the immunometabolism of immune cells that infiltrate adipose tissue. In turn, the level of progression of diabetes is significantly influenced by the Treg subpopulation, the complexity and heterogeneity of which is confirmed by the detection of numerous tissue-specific Tregs, including the so-called VAT Tregs (visceral adipose tissue CD4+Foxp3+ regulatory T cells). Therefore, the purpose of the study was to determine the mRNA expression levels of mTOR, Foxp3, IL1β, and IL17A genes in rat parapancreatic adipose tissue with experimental streptozotocin-induced diabetes mellitus, with or without metformin administration. The experiments were performed on male Wistar rats with induced diabetes as a result of streptozotocin administration. Molecular genetic studies were performed using real-time reverse transcription-polymerase chain reaction. The development of diabetes caused transcriptional activation of the mammalian target of rapamycin protein kinase gene, as well as increased mRNA expression of the pro-inflammatory cytokines IL1β and IL17A, but did not affect Foxp3 mRNA expression. The intervention with metformin in diabetic rats inhibited the mammalian target of rapamycin mRNA expression and caused an increase in the transcriptional activity of the Foxp3 gene in parapancreatic adipose tissue.
Collapse
Affiliation(s)
| | - Sergey Yuryevich Evchenko
- Department of Microbiology, Virology and Immunology, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | | | | | - Oleksandr Mikhailovich Kamyshny
- Department of Microbiology, Virology and Immunology, Molecular Genetics Laboratory, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | | | - Serhiy Mikhailovich Andreychyn
- Department of Propedeutics of Internal Medicine and Phthisiology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Ihorivna Hanberher
- Department of Propedeutics of Internal Medicine and Phthisiology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Tetyana Oleksandrivna Bezruk
- Department of Internal Medicine and Infectious Diseases, Bukovinian State Medical University, Chernivtsi, Ukraine
| |
Collapse
|
22
|
Farag AGA, Maraee AH, Rifaat Al-Sharaky D, Elshaib ME, Kohla MSM, Shehata WA. Tissue expression of IL-17A and FOXP3 in acne vulgaris patients. J Cosmet Dermatol 2020; 20:330-337. [PMID: 32413182 DOI: 10.1111/jocd.13485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/22/2020] [Accepted: 05/08/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND/OBJECTIVES CD4+ T helper (Th) cells through its pro-inflammatory cell type, interleukin-17 (IL-17)-generating cells and its anti-inflammatory category forkhead box P3-positive (FOXP3+ ) regulatory T (Treg) cells, play a vital role in the immune balance in inflammatory disorders. Therefore, assessment of both IL-17 and FOXP3 in acne vulgaris (AV), a chronic inflammatory disease of the pilosebaceous unit, could be of value in understanding AV pathogenesis. This study aimed to investigate the immunohistochemical expression of IL-17A and FOXP3 in acne vulgaris lesions versus normal skin. METHODS Forty-five AV patients and 25 controls were included in this case-control study. Biopsies from participants were analyzed for IL-17A and FOXP3 immunohistochemical profiles using IL-17A and FOXP3 polyclonal antibodies. RESULTS Compared to controls, AV patients exhibited a significant increase of IL-17A percent of expression in epidermis (P ≤ .001), in lymphocytes in papillary dermis (P ≤ .001), and in perifollicular lymphocytic inflammatory infiltrate in AV lesions. Also, there was a significant elevation in FOXP3 percent of expression in epidermis (P = .049) and in lymphocytes in papillary dermis (P ≤ .027) in acne patients than control. A significant positive correlation between IL-17A expression in papillary lymphocytes and in epidermal keratinocyte was observed (r = .537, P = .001). In acne vulgaris patients, the associations between IL-17A and FOXP3 expressions could not reach level of significance. CONCLUSIONS There was an up-regulation of IL-17A and FOXP3 in acne vulgaris development, but with independent roles. Moreover, targeting of IL-17A and FOXP3 may open the door for development of new therapeutic agents in acne vulgaris treatment.
Collapse
Affiliation(s)
- Azza Gaber Antar Farag
- Dermatology, Andrology and STDs Department, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| | - Alaa Hassan Maraee
- Dermatology, Andrology and STDs Department, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| | | | | | | | - Wafaa Ahmed Shehata
- Dermatology, Andrology and STDs Department, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| |
Collapse
|
23
|
Vaeth M, Wang YH, Eckstein M, Yang J, Silverman GJ, Lacruz RS, Kannan K, Feske S. Tissue resident and follicular Treg cell differentiation is regulated by CRAC channels. Nat Commun 2019; 10:1183. [PMID: 30862784 PMCID: PMC6414608 DOI: 10.1038/s41467-019-08959-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/11/2019] [Indexed: 12/30/2022] Open
Abstract
T regulatory (Treg) cells maintain immunological tolerance and organ homeostasis. Activated Treg cells differentiate into effector Treg subsets that acquire tissue-specific functions. Ca2+ influx via Ca2+ release-activated Ca2+ (CRAC) channels formed by STIM and ORAI proteins is required for the thymic development of Treg cells, but its function in mature Treg cells remains unclear. Here we show that deletion of Stim1 and Stim2 genes in mature Treg cells abolishes Ca2+ signaling and prevents their differentiation into follicular Treg and tissue-resident Treg cells. Transcriptional profiling of STIM1/STIM2-deficient Treg cells reveals that Ca2+ signaling regulates transcription factors and signaling pathways that control the identity and effector differentiation of Treg cells. In the absence of STIM1/STIM2 in Treg cells, mice develop a broad spectrum of autoantibodies and fatal multiorgan inflammation. Our findings establish a critical role of CRAC channels in controlling lineage identity and effector functions of Treg cells. Regulatory T (Treg) cells are important for maintaining immune homeostasis. Here the authors show that STIM1 and STIM2, which activate the Ca2+ channel ORAI1, are essential for the differentiation of peripheral Treg cells into tissue-resident and follicular Treg cells and their ability to limit autoimmunity in mice.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.,Institute for Systems Immunology, Julius-Maximilians University of Würzburg, 97078, Würzburg, Germany
| | - Yin-Hu Wang
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Miriam Eckstein
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA.,Institute for Systems Immunology, Julius-Maximilians University of Würzburg, 97078, Würzburg, Germany
| | - Jun Yang
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Gregg J Silverman
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Kasthuri Kannan
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.,Genome Technology Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
24
|
Abstract
There has been a prolific amount of research dedicated to the T-regulatory cells (Tregs) and their role in achieving immune homeostasis. Here, the authors briefly discuss the known biology, utilization, and potential of Tregs, for current trials and future immunotherapy. Most current trials of Treg therapies include either ex vivo expanded Tregs transferred into the peripheral blood of patients with diseases of immunologic origin or interleukin 2 injected to stimulate Tregs directly. Ongoing trials designed to measure the clinical efficacy and safety profile of these novel therapeutic approaches have resulted in largely favorable outcomes in a variety of autoimmune and alloimmune diseases.
Collapse
|
25
|
Barros JF, Waclawiak I, Pecli C, Borges PA, Georgii JL, Ramos-Junior ES, Canetti C, Courau T, Klatzmann D, Kunkel SL, Penido C, Canto FB, Benjamim CF. Role of Chemokine Receptor CCR4 and Regulatory T Cells in Wound Healing of Diabetic Mice. J Invest Dermatol 2018; 139:1161-1170. [PMID: 30465800 DOI: 10.1016/j.jid.2018.10.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/14/2018] [Accepted: 10/02/2018] [Indexed: 01/04/2023]
Abstract
Wound healing is a well-coordinated process that involves inflammatory mediators and cellular responses; however, if any disturbances are present during this process, tissue repair is impaired. Chronic wounds are one of the serious long-term complications associated with diabetes mellitus. The chemokine receptor CCR4 and its respective ligands, CCL17 and CCL22, are involved in regulatory T cell recruitment and activation in inflamed skin; however, the role of regulatory T cells in wounds is still not clear. Our aim was to investigate the role of CCR4 and regulatory T cells in cutaneous wound healing in diabetic mice. Alloxan-induced diabetic wild- type mice (diabetic) developed wounds that were difficult to heal, differently from CCR4-/- diabetic mice (CCR4-/- diabetic), and also from anti-CCL17/22 or anti-CD25-injected diabetic mice that presented with accelerated wound healing and fewer regulatory T cells in the wound bed. Consequently, CCR4-/- diabetic mice also presented with alteration on T cells population in the wound and draining lymph nodes; on day 14, these mice also displayed an increase of collagen fiber deposition. Still, cytokine levels were decreased in the wounds of CCR4-/- diabetic mice on day 2. Our data suggest that the receptor CCR4 and regulatory T cells negatively affect wound healing in diabetic mice.
Collapse
Affiliation(s)
- Janaína F Barros
- Institute of Biomedical Sciences, Pharmacology and Inflammation Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ingrid Waclawiak
- Institute of Biophysics Carlos Chagas Filho, Immunobiology Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cyntia Pecli
- Institute of Biomedical Sciences, Pharmacology and Inflammation Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula A Borges
- Institute of Biomedical Sciences, Pharmacology and Inflammation Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Janaína L Georgii
- Institute of Biomedical Sciences, Pharmacology and Inflammation Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Erivan S Ramos-Junior
- Institute of Biophysics Carlos Chagas Filho, Immunobiology Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Canetti
- Institute of Biophysics Carlos Chagas Filho, Immunobiology Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tristan Courau
- Sorbonne Universités, University of Pierre and Madam Curie, University of Paris, Paris, France
| | - David Klatzmann
- Sorbonne Universités, University of Pierre and Madam Curie, University of Paris, Paris, France; Institut National de la Santé et de la Recherche Médicale les Unités Mixtes de Recherche S959, Paris, France
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Carmen Penido
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Applied Pharmacology, Institute of Drug Technology, Farmanguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio B Canto
- Institute of Microbiology Paulo de Góes, Immunology Department, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil; Department of Immunobiology, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Claudia F Benjamim
- Institute of Biomedical Sciences, Pharmacology and Inflammation Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil; Institute of Biophysics Carlos Chagas Filho, Immunobiology Program, Federal University of Rio de Janeiro, Center for Health Sciences, Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Niedzielska M, Israelsson E, Angermann B, Sidders BS, Clausen M, Catley M, Malhotra R, Dumont C. Differential gene expression in human tissue resident regulatory T cells from lung, colon, and blood. Oncotarget 2018; 9:36166-36184. [PMID: 30546835 PMCID: PMC6281418 DOI: 10.18632/oncotarget.26322] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
As we learn more about how immune responses occur in situ, it is becoming clear that each organ/tissue is characterized with its own anatomy and microenvironment which may affect and even determine the outcome of the immune responses. With emerging data from animal studies showing that regulatory T cells infiltrating non-lymphoid tissues exhibit unique phenotypes and transcriptional signatures and display functions beyond their well-established suppressive roles, there is an urgent need to explore the function of tissue Treg cells in humans. Here we characterized the transcriptome of Treg residing at the human mucosal tissue obtained from the normal area of cancer resections and their peripheral blood counterparts, identifying human lung and colon tissue Treg signature genes and their upstream regulators. Pathway analysis highlighted potential differences in the cross-talk between tissue Treg cells and other non-immune tissue-specific cell types. For example, genes associated with wnt pathway were differentially regulated in lung Treg cells compared to blood or colon indicating a potential role for lung Treg cells in epithelium repair and regeneration. Moreover, we identified several non-coding RNAs specifically expressed by tissue-resident Tregs. These results provide a comprehensive view of lung and colon tissue Treg transcriptional landscape.
Collapse
Affiliation(s)
- Magdalena Niedzielska
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Israelsson
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Bastian Angermann
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Benjamin S Sidders
- Bioscience, Oncology, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Maryam Clausen
- Translational Genomics, Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Matthew Catley
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rajneesh Malhotra
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Céline Dumont
- Bioscience, Respiratory, Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
27
|
Varanasi SK, Rajasagi N, Jaggi U, Rouse B. Role of IL-18 induced Amphiregulin expression on virus induced ocular lesions. Mucosal Immunol 2018; 11:1705-1715. [PMID: 30087443 PMCID: PMC6279570 DOI: 10.1038/s41385-018-0058-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/03/2018] [Accepted: 06/12/2018] [Indexed: 02/04/2023]
Abstract
This report deals with the possible mechanism by which IL-18 can contribute to the control and resolution of inflammatory lesions in the cornea caused by herpes simplex virus infection. Our results demonstrate that the expression of the IL-18R by both regulatory T cells (Treg) and effector T cells was a pivotal event that influenced lesion pathogenesis. The engagement of IL-18R on Treg with its cytokine ligand resulted in Amphiregulin expression a molecule associated with tissue repair. In support of this scheme of events, lesion severity became more severe in animals unable to express the IL-18R because of gene knockout and was reduced in severity when IL-18 was overexpressed in the cornea. These changes in lesion severity correlated with the frequency and number of both Treg and Teff that expressed Amphiregulin. Additional experiments indicated that IL-12 and IL-18 acted synergistically to enhance Amphiregulin expression in Treg, an event partly dependent on P38 MAPK activity. Finally, sub-conjunctival administration of Amphiregulin resulted in resolution of both developing and developed lesions. Thus, overall our results imply that IL-18 may participate in controlling the severity of SK and contribute to tissue repair by converting both Treg and effector T cells into those that produce Amphiregulin.
Collapse
Affiliation(s)
- Siva Karthik Varanasi
- Department of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee
| | - Naveen Rajasagi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Ujjaldeep Jaggi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Barry Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee,Corresponding author. Barry T. Rouse:
| |
Collapse
|
28
|
Sharabi A, Tsokos MG, Ding Y, Malek TR, Klatzmann D, Tsokos GC. Regulatory T cells in the treatment of disease. Nat Rev Drug Discov 2018; 17:823-844. [DOI: 10.1038/nrd.2018.148] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Mizui M, Tsokos GC. Targeting Regulatory T Cells to Treat Patients With Systemic Lupus Erythematosus. Front Immunol 2018; 9:786. [PMID: 29755456 PMCID: PMC5932391 DOI: 10.3389/fimmu.2018.00786] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/29/2018] [Indexed: 12/30/2022] Open
Abstract
Regulatory T cells (Tregs) are central in integration and maintenance of immune homeostasis. Since breakdown of self-tolerance is a major culprit in the pathogenesis of systemic lupus erythematosus (SLE), restoration of the immune tolerance through the manipulation of Tregs can be exploited to treat patients with SLE. New information has revealed that Tregs besides their role in suppressing the immune response are important in tissue protection and regeneration. Expansion of Tregs with low-dose IL-2 represents an approach to control the autoimmune response. Moreover, control of Treg metabolism can be exploited to restore or improve their function. Here, we summarize the function and diversity of Tregs and recent strategies to improve their function in patients with SLE.
Collapse
Affiliation(s)
- Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Giusti D, Gatouillat G, Le Jan S, Plée J, Bernard P, Antonicelli F, Pham BN. Anti-Type VII Collagen Antibodies Are Identified in a Subpopulation of Bullous Pemphigoid Patients With Relapse. Front Immunol 2018; 9:570. [PMID: 29619029 PMCID: PMC5871753 DOI: 10.3389/fimmu.2018.00570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/06/2018] [Indexed: 11/19/2022] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune bullous skin disease characterized by anti-BP180 and anti-BP230 autoantibodies (AAbs). Mucous membrane involvement is an uncommon clinical feature of BP which may evoke epidermolysis bullosa acquisita, another skin autoimmune disease characterized by anti-type VII collagen AAbs. We therefore evaluated the presence of anti-type VII collagen AAbs in the serum of BP patients with and without mucosal lesions at time of diagnosis and under therapy. Anti-BP180, anti-BP230, and anti-type VII collagen AAbs were measured by ELISA in the serum of unselected patients fulfilling clinical and histo/immunopathological BP criteria at baseline (n = 71) and at time of relapse (n = 24). At baseline, anti-type VII collagen AAbs were detected in 2 out of 24 patients with BP presenting with mucosal involvement, but not in patients without mucosal lesions (n = 47). At the time of relapse, 10 out of 24 BP patients either displayed a significant induction or increase of concentrations of anti-type VII collagen AAbs (P < 0.01), independently of mucosal involvement. Those 10 relapsing BP patients were also characterized by a sustained high concentration of anti-BP180 AAb, whereas the serum anti-BP230 AAb concentrations did not vary in BP patients with relapse according to the presence of anti-type VII collagen AAbs. Thus, our study showed that anti-type VII collagen along with anti-BP180 AAbs detection stratified BP patients at time of relapse, illustrating a still dysregulated immune response that could reflect a potential epitope spreading mechanism in those BP patients.
Collapse
Affiliation(s)
- Delphine Giusti
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Grégory Gatouillat
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Sébastien Le Jan
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France
| | - Julie Plée
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Department of Dermatology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Philippe Bernard
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Department of Dermatology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Frank Antonicelli
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Department of Biological Sciences, Immunology, Faculty of Odontology, University of Reims Champagne-Ardenne, Reims, France
| | - Bach-Nga Pham
- Laboratory of Dermatology, Faculty of Medicine, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
31
|
Clapé A, Muller C, Gatouillat G, Le Jan S, Barbe C, Pham BN, Antonicelli F, Bernard P. Mucosal Involvement in Bullous Pemphigoid Is Mostly Associated with Disease Severity and to Absence of Anti-BP230 Autoantibody. Front Immunol 2018; 9:479. [PMID: 29662486 PMCID: PMC5890137 DOI: 10.3389/fimmu.2018.00479] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/22/2018] [Indexed: 01/10/2023] Open
Abstract
Bullous pemphigoid (BP) is the most common autoimmune bullous disease and typically affects the elderly. Binding of specific autoantibodies to BP180/230 hemidesmosomal components induces an inflammatory response leading to skin blister formation. Unusual manifestations of BP include additional mucous membrane involvement, without pathophysiological knowledge associated to the formation of these lesions. We here performed a prospective study on series of consecutive BP patients with (n = 77) and without (n = 18) mucosal involvements at baseline to further investigate why some BP patients display mucosal lesion and other not. Analysis of disease activity showed that BP patients with mucosal involvement displayed a higher total BP Disease Area Index (BPDAI) score (P = 0.008), but also higher skin and blister/erosion BPDAI scores (P = 0.02 and P = 0.001, respectively). By contrast, the erythema/urticaria BPDAI score was identical between the two groups of patients. The erythema/urticaria BPDAI score, but not the blister/erosion BPDAI score, was correlated with the serum concentration of anti-BP180 NC16A autoantibodies in patients with mucosal involvement. In multivariate analysis, the absence of anti-BP230 autoantibody was the only factor independently associated with mucosal involvement (OR 7.8; 95% CI, 3.1–19.6) (P < 0.0001). Analysis of the distribution of BP patients according to BPDAI scores revealed a shift toward higher blister/erosion BPDAI scores for BP patients with mucosal involvement. This study indicates that mucosal lesions are clinically mainly related to disease severity and immunologically to the absence of anti-BP230 antibodies.
Collapse
Affiliation(s)
- Ariane Clapé
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France.,Department of Dermatology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Céline Muller
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France
| | - Grégory Gatouillat
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Sébastien Le Jan
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France
| | - Coralie Barbe
- Clinical Research Unit, Reims University Hospital, Reims, France
| | - Bach-Nga Pham
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France.,Laboratory of Immunology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| | - Frank Antonicelli
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France.,Department of Biological Sciences, Immunology, UFR Odontology, University of Reims Champagne-Ardenne, Reims, France
| | - Philippe Bernard
- Laboratory of Dermatology, Faculty of Medicine, EA7319, University of Reims Champagne-Ardenne, Reims, France.,Department of Dermatology, Reims University Hospital, University of Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
32
|
Kobayashi T, Nagao K. Mapping regulatory circuits in allergic skin inflammation. Sci Immunol 2018; 3:3/21/eaar7655. [PMID: 29500226 DOI: 10.1126/sciimmunol.aar7655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 01/29/2018] [Indexed: 11/02/2022]
Abstract
RORα+ skin-resident regulatory T cells restrain innate lymphoid cell driven allergic skin inflammation.
Collapse
Affiliation(s)
- Tetsuro Kobayashi
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keisuke Nagao
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
33
|
Chen Z, Zhou L, Won T, Gao Z, Wu X, Lu L. Characterization of CD45RO + memory T lymphocytes in keloid disease. Br J Dermatol 2018; 178:940-950. [PMID: 29194570 DOI: 10.1111/bjd.16173] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Memory T cells, a highly effective subset of T lymphocytes, have been reported to be involved in many inflammatory skin disorders. However, the potential role of memory T cells in keloid disease (KD) remains unclear. OBJECTIVES Due to their important role in regulating inflammation, we investigated the characteristics of CD45RO+ memory T cells in KD. METHODS Primary cutaneous cells were isolated from keloid scars and normal skin by enzymic digestion. Peripheral blood mononuclear cells were isolated from a related blood sample, and flow cytometry was applied to identify the phenotypic and functional abnormalities of memory T cells in KD. RESULTS We observed that the majority of T lymphocytes in keloid scars had the memory phenotype, and a greater number of the CD8+ memory T cells in keloid scars produced lower levels of tumour necrosis factor (TNF)-α. This abnormal cytokine production was even more distinct in Forkhead box (FOX)P3- CD8- memory T cells, with lower TNF-α production and enhanced interferon-γ production. Furthermore, FOXP3+ CD8- memory T cells in keloid scars were abnormal, including showing reduced CD25 and cytotoxic T-lymphocyte-associated antigen 4 expression and interleukin-10 production. In addition, a significant decrease in the number of CD4+ CD25high FOXP3+ regulatory T cells was identified in patients with multiple keloid scars. We also found that there was significantly increased infiltration of CD103+ CD8+ memory T cells in keloid scars. CONCLUSIONS Our findings preliminarily elucidate the abnormalities of CD45RO+ memory T cells in keloid scars and provide early evidence that a disrupted T-cell response contributes to the progression of KD.
Collapse
Affiliation(s)
- Z Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - L Zhou
- Research Center, Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, China
| | - T Won
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Z Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - X Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - L Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
34
|
Niccolai E, Ricci F, Russo E, Nannini G, Emmi G, Taddei A, Ringressi MN, Melli F, Miloeva M, Cianchi F, Bechi P, Prisco D, Amedei A. The Different Functional Distribution of "Not Effector" T Cells (Treg/Tnull) in Colorectal Cancer. Front Immunol 2017; 8:1900. [PMID: 29375559 PMCID: PMC5770731 DOI: 10.3389/fimmu.2017.01900] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, ranking as high as the second leading cause of cancer-related deaths in industrialized countries. Consistent with immunosurveillance theory, the immune system is crucial to protect the host from developing tumors, and the major players in tumoral immunity are effector T cells. Anyway, cancer cells develop strategies of immunoevasion influencing the cancer-specific lymphocyte priming, activation, and effector function. Therefore, the T cell subsets that mature during the stages of tumor growth, differently contribute to disease progression and/or regression. In our study, we analyzed the intra-tumoral and peripheral T cell subsets’ distribution in 30 patients with CRC, in order to clarify their functional role toward cancer. We found that percentage of infiltrating effector T cells decreased in cancer tissue than in healthy mucosa and that the tumor microenvironment negatively influences the cytolytic activity of T lymphocytes reactive to cancer cells. Moreover, we found that the tumor tissue was infiltrated by a large amount of “not effector” T (neT) cells with a regulatory or an anergic profile, which are unable to kill cancer cells, may be contributing to the CRC promotion. The presence of neT cells was investigated also in the peripheral blood of CRC patients, demonstrating that the peripheral T regulatory cells can inhibit the proliferation of effector T cells, confirming their immunosuppressive properties. Finally, monitoring the changes in circulating neT cells’ frequencies after the tumor removal, we confirmed the role of cancer in the modulation of immune system, in particular, in supporting a Tregs-mediated immunosuppression.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Federica Ricci
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Giulia Nannini
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Giacomo Emmi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Antonio Taddei
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | | | - Filippo Melli
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Manouela Miloeva
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Fabio Cianchi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Paolo Bechi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Domenico Prisco
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
35
|
Guo F, Hu M, Huang D, Zhao Y, Heng B, Guillemin G, Lim CK, Hawthorne WJ, Yi S. Human regulatory macrophages are potent in suppression of the xenoimmune response via indoleamine-2,3-dioxygenase-involved mechanism(s). Xenotransplantation 2017; 24. [PMID: 28771838 DOI: 10.1111/xen.12326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 05/17/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND For xenotransplantation to truly succeed, we must develop immunomodulatory strategies to suppress the xenoimmune response but by minimizing immunosuppression over the long term. Regulatory macrophages (Mreg) have been shown to suppress polyclonal T-cell proliferation in vitro and prolong allograft survival in vivo. However, the question of whether they are capable of suppressing xenoimmune responses remains unknown. This study assessed the potential of human Mreg to be used as an effective immunomodulatory method in xenotransplantation. METHODS CD14+ monocytes selected from human peripheral blood mononuclear cells (PBMC) were cultured with macrophage colony-stimulating factor (M-CSF) for 7 days with IFN-γ added at day 6 for Mreg induction. Mreg phenotyping was performed by flow cytometric analysis, and the in vitro suppressive function was assessed by mixed lymphocyte reaction (MLR) using irradiated pig PBMC as the xenogeneic stimulator cells, human PBMC as responder cells, and autologous Mreg as suppressor cells. To assess mRNA expression of Mreg functional molecules indoleamine-2,3-dioxygenase (IDO), IL-10, inducible nitric oxide synthase (iNOS) and TGF-β were measured by real-time PCR. Supernatants were collected from the MLR cultures for IDO activity assay by high-performance liquid chromatography (HPLC). The effects of the IDO inhibitor 1-D/L-methyl-tryptophan (1-MT), iNOS inhibitor NG -monomethyl-l-arginine (L-NMMA), and anti-IFN-γ or anti-TGF-β monoclonal antibody (mAb) treatment on Mreg suppressive capacity were tested from the supernatants of the MLR assays. RESULTS We demonstrated that induced Mreg with a phenotype of CD14low CD16-/low CD80low CD83-/low CD86+/hi HLA-DR+/hi were capable of suppressing proliferating human PBMC, CD4+, and CD8+ T cells, even at a higher responder:Mreg ratio of 32:1 in a pig-human xenogeneic MLR. The strong suppressive potency of Mreg was further correlated with their upregulated IDO expression and activity. The IDO upregulation of Mreg was associated with an increased production of IFN-γ, an IDO stimulator, by xenoreactive responder cells in the xenogeneic MLR. While no effect on Mreg suppressive potency was detected by addition of the iNOS inhibitor L-NMMA or anti-TGF-β mAb into the MLR assays, inhibition of IDO activity by neutralizing IFN-γ or by IDO inhibitor 1-MT substantially impaired the capacity of Mreg to suppress the xenogeneic response, indicating the importance of upregulated IDO activity in Mreg-mediated suppression of the xenogeneic response in vitro. CONCLUSION This study demonstrates that human Mreg are capable of suppressing the xenoimmune response in vitro via IDO-involved mechanism(s), suggesting their potential role as an effective immunomodulatory tool in xenotransplantation.
Collapse
Affiliation(s)
- Fei Guo
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia.,Cell Transplantation and Gene Therapy Institute of Central South University at the 3rd Xiangya Hospital, Changsha, Hunan, China
| | - Min Hu
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Dandan Huang
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Yuanfei Zhao
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Benjamin Heng
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Gilles Guillemin
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Chai K Lim
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Wayne J Hawthorne
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Shounan Yi
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
36
|
Abstract
Body weight regain often causes failure of obesity therapies while the underlying mechanism remains largely unknown. In this study, we report that immune cells, especially CD4+ T cells, mediate the ‘memory’ of previous obese status. In a weight gain-loss-regain model, we found that C57BL/6J mice with an obesity history showed a much faster rate of body weight regain. This obesity memory could last for at least 2 months after previously obese mice were kept at the same body weight as non-obese mice. Surprisingly, such obesity memory was abrogated by dexamethasone treatment, whereas immunodeficient Rag1−/− and H2A−/− mice failed to establish such memory. Rag1−/− mice repossessed the obesity memory when immune cells or CD4+ T cells isolated from previously obese mice were transferred. Furthermore, depletion of CD4+ T cells led to obesity memory ablation. Taken together, we conclude that CD4+ T cells mediate obesity memory and promote weight regain.
Collapse
|
37
|
Fang F, Xiao W, Tian Z. NK cell-based immunotherapy for cancer. Semin Immunol 2017; 31:37-54. [DOI: 10.1016/j.smim.2017.07.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
|
38
|
Fu G, Xu Q, Qiu Y, Jin X, Xu T, Dong S, Wang J, Ke Y, Hu H, Cao X, Wang D, Cantor H, Gao X, Lu L. Suppression of Th17 cell differentiation by misshapen/NIK-related kinase MINK1. J Exp Med 2017; 214:1453-1469. [PMID: 28400474 PMCID: PMC5413330 DOI: 10.1084/jem.20161120] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 01/09/2017] [Accepted: 02/22/2017] [Indexed: 12/22/2022] Open
Abstract
T helper type 17 cells (Th17 cells) are major contributors to many autoimmune diseases. In this study, we demonstrate that the germinal center kinase family member MINK1 (misshapen/NIK-related kinase 1) negatively regulates Th17 cell differentiation. The suppressive effect of MINK1 on induction of Th17 cells is mediated by the inhibition of SMAD2 activation through direct phosphorylation of SMAD2 at the T324 residue. The importance of MINK1 to Th17 cell differentiation was strengthened in the animal model of experimental autoimmune encephalomyelitis (EAE). Moreover, we show that the reactive oxygen species (ROS) scavenger N-acetyl cysteine boosts Th17 cell differentiation in a MINK1-dependent manner and exacerbates the severity of EAE. Thus, we have not only established MINK1 as a critical regulator of Th17 cell differentiation, but also clarified that accumulation of ROS may limit the generation of Th17 cells. The contribution of MINK1 to ROS-regulated Th17 cell differentiation may suggest an important mechanism for the development of autoimmune diseases influenced by antioxidant dietary supplements.
Collapse
Affiliation(s)
- Guotong Fu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qin Xu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuanjun Qiu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuexiao Jin
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ting Xu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shunli Dong
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jianli Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuehai Ke
- Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hu Hu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Harvey Cantor
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115.,Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115
| | - Xiang Gao
- Key Laboratory of Model Animals for Disease Study of the Ministry of Education, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Linrong Lu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China .,Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Innovation Center for Cell Signaling Network, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou 310058, China.,Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
39
|
Tian Z, Cao X, Chen Y, Lyu Q. Regional immunity in tissue homeostasis and diseases. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1205-1209. [PMID: 27928702 DOI: 10.1007/s11427-016-0351-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Zhigang Tian
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, 230027, China.
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China.
| | - Yongyan Chen
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, 230027, China
| | - Qunyan Lyu
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China
| |
Collapse
|
40
|
Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines (Basel) 2016; 4:vaccines4030028. [PMID: 27509527 PMCID: PMC5041022 DOI: 10.3390/vaccines4030028] [Citation(s) in RCA: 323] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/27/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
Recent years have seen significant efforts in understanding and modulating the immune response in cancer. In this context, immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), have come under intense investigation for their proposed roles in suppressing tumor-specific immune responses and establishing an immunosuppressive tumor microenvironment, thus enabling tumor immune evasion. Additionally, recent evidence indicates that Tregs comprise diverse and heterogeneous subsets; phenotypically and functionally distinct subsets of tumor-infiltrating Tregs could contribute differently to cancer prognosis and clinical outcomes. Understanding Treg biology in the setting of cancer, and specifically the tumor microenvironment, is important for designing effective cancer therapies. In this review, we critically examine the role of Tregs in the tumor microenvironment and in cancer progression focusing on human studies. We also discuss the impact of current therapeutic modalities on Treg biology and the therapeutic opportunities for targeting Tregs to enhance anti-tumor immune responses and clinical benefits.
Collapse
|
41
|
Marcel N, Sarin A. Notch1 regulated autophagy controls survival and suppressor activity of activated murine T-regulatory cells. eLife 2016; 5. [PMID: 27267497 PMCID: PMC4894756 DOI: 10.7554/elife.14023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Cell survival is one of several processes regulated by the Notch pathway in mammalian cells. Here we report functional outcomes of non-nuclear Notch signaling to activate autophagy, a conserved cellular response to nutrient stress, regulating survival in murine natural T-regulatory cells (Tregs), an immune subset controlling tolerance and inflammation. Induction of autophagy required ligand-dependent, Notch intracellular domain (NIC) activity, which controlled mitochondrial organization and survival of activated Tregs. Consistently, NIC immune-precipitated Beclin and Atg14, constituents of the autophagy initiation complex. Further, ectopic expression of an effector of autophagy (Atg3) or recombinant NIC tagged to a nuclear export signal (NIC-NES), restored autophagy and suppressor function in Notch1(-/-) Tregs. Furthermore, Notch1 deficiency in the Treg lineage resulted in immune hyperactivity, implicating Notch activity in Treg homeostasis. Notch1 integration with autophagy, revealed in these experiments, holds implications for Notch regulated cell-fate decisions governing differentiation.
Collapse
Affiliation(s)
- Nimi Marcel
- National Centre for Biological Sciences, Bengaluru, India.,Department of Biology, Manipal University, Manipal, India
| | - Apurva Sarin
- National Centre for Biological Sciences, Bengaluru, India.,Institute for Stem Cell Biology & Regenerative Medicine, Bengaluru, India
| |
Collapse
|
42
|
Dioszeghy V, Mondoulet L, Puteaux E, Dhelft V, Ligouis M, Plaquet C, Dupont C, Benhamou PH. Differences in phenotype, homing properties and suppressive activities of regulatory T cells induced by epicutaneous, oral or sublingual immunotherapy in mice sensitized to peanut. Cell Mol Immunol 2016; 14:770-782. [PMID: 27063469 PMCID: PMC5596241 DOI: 10.1038/cmi.2016.14] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 12/20/2022] Open
Abstract
Allergen-specific immunotherapy has been proposed as an attractive strategy to actively treat food allergy using the following three different immunotherapy routes: oral (OIT), sublingual (SLIT) and epicutaneous (EPIT) immunotherapy. Regulatory T cells (Tregs) have been shown to have a pivotal role in the mechanisms of immunotherapy. The aim of this study was to compare the phenotype and function of Tregs induced in peanut-sensitized BALB/c mice using these three routes of treatment. We show that although EPIT, OIT and SLIT were all able to effectively desensitize peanut-sensitized mice, they induced different subsets of Tregs. Foxp3+ Tregs were induced by the three treatment routes but with greater numbers induced by EPIT. EPIT and OIT also increased the level of LAP+ Tregs, whereas SLIT induced IL-10+ cells. The suppressive activity of EPIT-induced Tregs did not depend on IL-10 but required CTLA-4, whereas OIT acted through both mechanisms and SLIT was strictly dependent on IL-10. Moreover, the three routes influenced the homing properties of induced Tregs differently, with a larger repertoire of chemokine receptors expressed by EPIT-induced Tregs compared with OIT- and SLIT- induced cells, resulting in different protective consequences against allergen exposure. Furthermore, whereas OIT- or SLIT-induced Tregs lost their suppressive activities after treatment was discontinued, the suppressive activities of EPIT-induced Tregs were still effective 8 weeks after the end of treatment, suggesting the induction of a more long-lasting tolerance. In summary, EPIT, OIT and SLIT mediated desensitization through the induction of different subsets of Tregs, leading to important differences in the subsequent protection against allergen exposure and the possible induction of tolerance.
Collapse
Affiliation(s)
| | | | - Emilie Puteaux
- Research Department, DBV Technologies, Paris, 92220, France
| | | | | | | | - Christophe Dupont
- Pédiatrie-Gastroentérologie, Université Paris Descartes &APHP-Hôpital Necker, Paris, 75743, France
| | | |
Collapse
|
43
|
Perdigoto AL, Chatenoud L, Bluestone JA, Herold KC. Inducing and Administering Tregs to Treat Human Disease. Front Immunol 2016; 6:654. [PMID: 26834735 PMCID: PMC4722090 DOI: 10.3389/fimmu.2015.00654] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
Regulatory T cells (Tregs) control unwanted immune responses, including those that mediate tolerance to self as well as to foreign antigens. Their mechanisms of action include direct and indirect effects on effector T cells and important functions in tissue repair and homeostasis. Tregs express a number of cell surface markers and transcriptional factors that have been instrumental in defining their origins and potentially their function. A number of immune therapies, such as rapamycin, IL-2, and anti-T cell antibodies, are able to induce Tregs and are being tested for their efficacy in diverse clinical settings with exciting preliminary results. However, a balance exists with the use of some, such as IL-2, that may have effects on unwanted populations as well as promoting expansion and survival of Tregs requiring careful selection of dose for clinical use. The use of cell surface markers has enabled investigators to isolate and expand ex vivo Tregs more than 500-fold routinely. Clinical trials have begun, administering these expanded Tregs to patients as a means of suppressing autoimmune and alloimmune responses and potentially inducing immune tolerance. Studies in the future are likely to build on these initial technical achievements and use combinations of agents to improve the survival and functional capacity of Tregs.
Collapse
Affiliation(s)
- Ana Luisa Perdigoto
- Department of Immunobiology, Yale University, New Haven, CT, USA; Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Lucienne Chatenoud
- Université Paris Descartes, Sorbonne Paris Cité, F-75475, Paris, France; INSERM U1151, CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - Jeffrey A Bluestone
- Diabetes Center, University of California San Francisco , San Francisco, CA , USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, USA; Department of Internal Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
44
|
Guo L, Zhang Y, Zhang L, Huang F, Li J, Wang S. MicroRNAs, TGF-β signaling, and the inflammatory microenvironment in cancer. Tumour Biol 2016; 37:115-25. [PMID: 26563372 PMCID: PMC4841843 DOI: 10.1007/s13277-015-4374-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
Inflammatory cells and mediators form a major part of the tumor microenvironment and play important roles in the regulation of cancer initiation, tumor cell proliferation, and metastasis. MicroRNAs (miRNAs) play important roles in several physiological and pathological processes, including the regulation of the inflammatory microenvironment in cancer. Transforming growth factor-β (TGF-β) is an inflammation-related cytokine that functions in both tumor suppression and promotion; however, its underlying molecular mechanisms remain unclear. Recent evidence indicates an association between miRNAs and TGF-β signaling, providing new insight into the nature of the inflammatory microenvironment in cancer. The present review is an overview of the interaction between miRNAs and inflammatory cytokines, with emphasis on the cross talk between TGF-β signaling and miRNAs and their influence on cancer cell behavior. The emerging roles of miRNAs in cancer-related inflammation and the potential to target miRNA signaling pathways for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Lingling Guo
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Yongsheng Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lifeng Zhang
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fengbo Huang
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Jinfan Li
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Shouli Wang
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China.
- Institute of Radiology and Oncology, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
45
|
Hu ZQ, Zhao WH. The IL-33/ST2 axis is specifically required for development of adipose tissue-resident regulatory T cells. Cell Mol Immunol 2015; 12:521-4. [PMID: 26277897 PMCID: PMC4579657 DOI: 10.1038/cmi.2015.49] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 05/11/2015] [Indexed: 12/30/2022] Open
Affiliation(s)
- Zhi-Qing Hu
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Wei-Hua Zhao
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
How regulatory T cells sense and adapt to inflammation. Cell Mol Immunol 2015; 12:519-20. [PMID: 26277895 DOI: 10.1038/cmi.2015.65] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/10/2015] [Accepted: 06/10/2015] [Indexed: 12/29/2022] Open
|