1
|
Pfeffer K, Ho TH, Ruiz Y, Lake DF. A method for screening functional anti-Treg antibodies using a Treg-like cell line. J Leukoc Biol 2025; 117:qiae257. [PMID: 39739859 DOI: 10.1093/jleuko/qiae257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/15/2024] [Accepted: 12/30/2024] [Indexed: 01/02/2025] Open
Abstract
Regulatory T cells can suppress activated T-cell proliferation by direct cell contact, although the exact mechanism is poorly understood. Identification of a Treg-specific cell surface molecule that mediates suppression would offer a unique target for cancer immunotherapy to inhibit Treg immunosuppressive function or deplete Tregs in the tumor microenvironment. In this study, we explored a method of whole-cell immunization using a Treg-like cell line (MoT cells) to generate and screen monoclonal antibodies that bound cell surface proteins in their native conformations and functionally reversed Treg-mediated suppression. From the 105 hybridomas that bound to the MoT cell surface, a functional screen utilizing conventional Treg suppression assays revealed 32 candidate antibodies that exhibited functional activity (reversed or enhanced suppressive activity). As an example, we characterized 1 anti-MoT mAb, 12E7, that exhibited strong binding to MoT cells and conventional Treg cell surfaces. This candidate antibody was subsequently found to bind to a potential suppressive target, CD44, and demonstrated the ability to partially reverse MoT and conventional human Treg-mediated suppression.
Collapse
Affiliation(s)
- Kirsten Pfeffer
- School of Life Sciences, Arizona State University, 6161 E. Mayo Blvd, Phoenix, AZ 85054, United States
| | - Thai H Ho
- Division of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, United States
| | - Yvette Ruiz
- School of Life Sciences, Arizona State University, 6161 E. Mayo Blvd, Phoenix, AZ 85054, United States
| | - Douglas F Lake
- School of Life Sciences, Arizona State University, 6161 E. Mayo Blvd, Phoenix, AZ 85054, United States
| |
Collapse
|
2
|
Abu-Romman A, Scholand KK, Pal-Ghosh S, Yu Z, Kelagere Y, Yazdanpanah G, Kao WWY, Coulson-Thomas VJ, Stepp MA, de Paiva CS. Conditional deletion of CD25 in the corneal epithelium reveals sex differences in barrier disruption. Ocul Surf 2023; 30:57-72. [PMID: 37516317 PMCID: PMC10812880 DOI: 10.1016/j.jtos.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/06/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
PURPOSE IL-2 promotes activation, clonal expansion, and deletion of T cells. IL-2 signals through its heterotrimeric receptor (IL-2R) consisting of the CD25, CD122 and CD132 chains. CD25 knockout (KO) mice develop Sjögren Syndrome-like disease. This study investigates whether corneal CD25/IL-2 signaling is critical for ocular health. METHODS Eyes from C57BL/6 mice were collected and prepared for immunostaining or in-situ hybridization. Bulk RNA sequencing was performed on the corneal epithelium from wild-type and CD25KO mice. We generated a conditional corneal-specific deletion of CD25 in the corneal epithelium (CD25Δ/ΔCEpi). Corneal barrier function was evaluated based on the uptake of a fluorescent dye. Mice were subjected to unilateral corneal debridement, followed by epithelial closure over time. RESULTS In C57BL/6 mice, CD25 mRNA was expressed in ocular tissues. Protein expression of CD25, CD122, and CD132 was confirmed in the corneal epithelium. Delayed corneal re-epithelization was seen in female but not male CD25KO mice. There were 771 differentially expressed genes in the corneal epithelium of CD25KO compared to wild-type mice. While barrier function is disrupted in CD25Δ/ΔCEpi mice, re-epithelialization rates are not delayed. CONCLUSIONS All three chains of the IL-2R are expressed in the corneal epithelium. Our results indicate for the first time, deleting CD25 systemically in all tissues in the mouse and deleting CD25 locally in just the corneal epithelium compromises corneal epithelial barrier function, leading to dry eye disease in female mice. Future studies are needed to delineate the pathways used by IL-2 signaling to influence cornea homeostasis.
Collapse
Affiliation(s)
- Anmar Abu-Romman
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Kaitlin K Scholand
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States; Department of Biosciences, Rice University, Houston, TX, United States.
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.
| | - Zhiyuan Yu
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Yashaswini Kelagere
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Ghasem Yazdanpanah
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States.
| | - Winston W-Y Kao
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, United States.
| | | | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States; Department of Ophthalmology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, United States; Department of Biosciences, Rice University, Houston, TX, United States.
| |
Collapse
|
3
|
Wang X, Lu J, Guo G, Yu J. Immunotherapy for recurrent glioblastoma: practical insights and challenging prospects. Cell Death Dis 2021; 12:299. [PMID: 33741903 PMCID: PMC7979733 DOI: 10.1038/s41419-021-03568-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GB) is the most common high-grade intracranial malignant tumor with highly malignant biological behavior and a high recurrence rate. Although anti-PD-1/PD-L1 antibodies have achieved significant survival benefits in several kinds of solid tumors, the phase III clinical trial Checkmate 143 demonstrated that nivolumab, which targets PD-1, did not achieve survival benefits compared with bevacizumab in recurrent glioblastoma (rGB) patients. Nevertheless, neoadjuvant anti-PD-1 therapy followed by surgery and adjuvant anti-PD-1 therapy could effectively activate local and systemic immune responses and significantly improve the OS of rGB patients. Furthermore, several studies have also confirmed the progress made in applying tumor-specific peptide vaccination or chimeric antigen receptor-T (CAR-T) cell therapy to treat rGB patients, and successes with antibodies targeting other inhibitory checkpoints or costimulatory molecules have also been reported. These successes inspired us to explore candidate combination treatments based on anti-PD-1/PD-L1 antibodies. However, effective predictive biomarkers for clinical efficacy are urgently needed to avoid economic waste and treatment delay. Attempts to prolong the CAR-T cell lifespan and increase T cell infiltration through engineering techniques are addressing the challenge of strengthening T cell function. In this review, we describe the immunosuppressive molecular characteristics of rGB; clinical trials exploring anti-PD-1/PD-L1 therapy, tumor-specific peptide vaccination, and CAR-T cell therapy; candidate combination strategies; and issues related to strengthening T cell function.
Collapse
Affiliation(s)
- Xin Wang
- Departmenlt of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China. .,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, China.
| | - Jie Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, 250117, Shandong Province, China
| | - Gaochao Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital Zhengzhou University, People's Hospital Henan University, Zhengzhou, 450003, Henan, China
| | - Jinming Yu
- Departmenlt of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China. .,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, China.
| |
Collapse
|
4
|
Radichev IA, Yoon J, Scott DW, Griffin K, Savinov AY. Towards antigen-specific Tregs for type 1 diabetes: Construction and functional assessment of pancreatic endocrine marker, HPi2-based chimeric antigen receptor. Cell Immunol 2020; 358:104224. [PMID: 33068914 DOI: 10.1016/j.cellimm.2020.104224] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/05/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease marked by direct elimination of insulin-producing β cells by autoreactive T effectors. Recent T1D clinical trials utilizing autologous Tregs transfers to restore immune balance and improve disease has prompted us to design a novel Tregs-based antigen-specific T1D immunotherapy. We engineered a Chimeric Antigen Receptor (CAR) expressing a single-chain Fv recognizing the human pancreatic endocrine marker, HPi2. Human T cells, transduced with the resultant HPi2-CAR, proliferated and amplified Granzyme B accumulation when co-cultured with human, but not mouse β cells. Furthermore, following exposure of HPi2-CAR transduced cells to islets, CD8+ lymphocytes demonstrated enhanced CD107a (LAMP-1) expression, while CD4+ cells produced increased levels of IL-2. HPi2-CAR Tregs failed to maintain expansion due to a persistent tonic signaling from the CAR engagement to unexpectantly HPi2 antigen present on Tregs. Overall, we show lack of functionality of HPi2-CAR and highlight the importance of careful selection of CAR recognition driver for the sustainable activity and expandability of engineered T cells.
Collapse
Affiliation(s)
- Ilian A Radichev
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA
| | - Jeongheon Yoon
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - David W Scott
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Kurt Griffin
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA
| | - Alexei Y Savinov
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA.
| |
Collapse
|
5
|
Raffin C, Vo LT, Bluestone JA. T reg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020; 20:158-172. [PMID: 31811270 PMCID: PMC7814338 DOI: 10.1038/s41577-019-0232-6] [Citation(s) in RCA: 473] [Impact Index Per Article: 94.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/25/2022]
Abstract
Cellular therapies using regulatory T (Treg) cells are currently undergoing clinical trials for the treatment of autoimmune diseases, transplant rejection and graft-versus-host disease. In this Review, we discuss the biology of Treg cells and describe new efforts in Treg cell engineering to enhance specificity, stability, functional activity and delivery. Finally, we envision that the success of Treg cell therapy in autoimmunity and transplantation will encourage the clinical use of adoptive Treg cell therapy for non-immune diseases, such as neurological disorders and tissue repair.
Collapse
Affiliation(s)
- Caroline Raffin
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Linda T Vo
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Yu SJ, Greten TF. Deciphering and Reversing Immunosuppressive Cells in the Treatment of Hepatocellular Carcinoma. JOURNAL OF LIVER CANCER 2020; 20:1-16. [PMID: 37383056 PMCID: PMC10035699 DOI: 10.17998/jlc.20.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 06/30/2023]
Abstract
Use of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC) has been partially successful. However, most HCC patients do not respond to immunotherapy. HCC has been shown to induce several immune suppressor mechanisms in patients. These suppressor mechanisms include involvement of myeloid-derived suppressor cells, regulatory T-cells, functionally impaired dendritic cells (DCs), neutrophils, monocytes, and tumor associated macrophages. The accumulation of immunosuppressive cells may lead to an immunosuppressive tumor microenvironment as well as the dense fibrotic stroma which may contribute to immune tolerance. Our laboratory has been investigating different cellular mechanisms of immune suppression in HCC patients. In vitro as well as in vivo studies have demonstrated that abrogation of the suppressor cells enhances or unmasks tumor-specific antitumor immune responses. Two or three effective systemic therapies including ICIs and/or molecular targeted therapies and the addition of innovative combination therapies targeting immune suppressor cells may lead to increased immune recognition with a greater tumor response. We reviewed the literature for the latest research on immune suppressor cells in HCC, and here we provide a comprehensive summary of the recent studies in this field.
Collapse
Affiliation(s)
- Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
- NCI CCR Liver Cancer Program, Bethesda, USA
| |
Collapse
|
7
|
Zhao X, Li Y, Wang X, Wu J, Yuan Y, Lv S, Ren J. Synergistic association of FOXP3+ tumor infiltrating lymphocytes with CCL20 expressions with poor prognosis of primary breast cancer: A retrospective cohort study. Medicine (Baltimore) 2019; 98:e18403. [PMID: 31852159 PMCID: PMC6922488 DOI: 10.1097/md.0000000000018403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Studies have shown that forkhead/winged helix transcription factor P3 (FOXP3) tumor infiltrating lymphocytes (TILs) are intimately associated with invasion and survival of many invasive tumors. The inflammatory chemokine ligand 20 (CCL20) and its receptor CCR6 were found to be associated with tumor prognosis in some studies. Although increases in FOXP3 TILs infiltration and CCL20 expression have been revealed in several malignancies, their correlation in human breast tumors is as yet unclear.Surgically resected samples from 156 patients with invasive breast cancer (BC) were assessed for the expression of FOXP3 and CCL20 by immunohistochemistry. Correlation between their expressions and the association with clinicopathological characteristics and patient's prognosis were studied. Forty pairs of fresh BC and their nontumor adjacent tissues (NATs) in BC were carried out by real-time quantitative PCR (qRT-PCR) to evaluate the correlation between FOXP3 and CCL20 mRNA expression.CCL20 and FOXP3 TILs mRNA expression in tumor tissue demonstrated a high correlation (rs = 0.359, P < .001) in this cohort of breast cancer patients. Both elevated CCL20 expression and FOXP3 TILs infiltration were significantly correlated with high histological grade, positive human epidermal growth factor receptor-2 (HER2), high Ki67 index, and axillary lymph node metastases. Tumors with concomitant high expressions of both markers had the worst prognosis. Multivariate analysis showed that these 2 markers were independent predictors of overall survival. The patients with axillary lymph node metastases with the concomitant CCL20 high expression and increased FOXP3 TILs infiltration had the worst overall survival (OS) (P < .001), In lymph node-negative breast cancer patients, the status of CCL20 and FOXP3 was not related to OS (P = .22).The results suggest that CCL20 and FOXP3 TILs may have synergistic effects, and their upregulated expressions may lead to immune evasion in breast cancer. Combinatorial immunotherapeutic approaches aiming at blocking CCL20 and depleting FOXP3 might improve therapeutic efficacy in breast cancer patients.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yanping Li
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Yanhua Yuan
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
| | - Shuzhen Lv
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgical Breast Cancer, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines
- Department of Surgery, Duke University Medical Center, Durham, NC, US
| |
Collapse
|
8
|
Singh A, Ramachandran S, Graham ML, Daneshmandi S, Heller D, Suarez-Pinzon WL, Balamurugan AN, Ansite JD, Wilhelm JJ, Yang A, Zhang Y, Palani NP, Abrahante JE, Burlak C, Miller SD, Luo X, Hering BJ. Long-term tolerance of islet allografts in nonhuman primates induced by apoptotic donor leukocytes. Nat Commun 2019; 10:3495. [PMID: 31375697 PMCID: PMC6677762 DOI: 10.1038/s41467-019-11338-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Immune tolerance to allografts has been pursued for decades as an important goal in transplantation. Administration of apoptotic donor splenocytes effectively induces antigen-specific tolerance to allografts in murine studies. Here we show that two peritransplant infusions of apoptotic donor leukocytes under short-term immunotherapy with antagonistic anti-CD40 antibody 2C10R4, rapamycin, soluble tumor necrosis factor receptor and anti-interleukin 6 receptor antibody induce long-term (≥1 year) tolerance to islet allografts in 5 of 5 nonsensitized, MHC class I-disparate, and one MHC class II DRB allele-matched rhesus macaques. Tolerance in our preclinical model is associated with a regulatory network, involving antigen-specific Tr1 cells exhibiting a distinct transcriptome and indirect specificity for matched MHC class II and mismatched class I peptides. Apoptotic donor leukocyte infusions warrant continued investigation as a cellular, nonchimeric and translatable method for inducing antigen-specific tolerance in transplantation. Injection of donor apoptotic cells induces graft tolerance in mice. Here the authors combine this approach with short immunosuppressive therapy to achieve long-term tolerance to allogeneic islets and restoration of normoglycemia in diabetic nonhuman primates, and delineate cellular and molecular correlates of tolerance induction.
Collapse
Affiliation(s)
- Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sabarinathan Ramachandran
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Melanie L Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Saeed Daneshmandi
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Heller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wilma Lucia Suarez-Pinzon
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Appakalai N Balamurugan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA.,Center for Cellular Transplantation, Cardiovascular Innovation Institute, Department of Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Jeffrey D Ansite
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joshua J Wilhelm
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Amy Yang
- Biostatistics Collaboration Center, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nagendra P Palani
- University of Minnesota Genomics Center, Minneapolis, MN, 55455, USA
| | - Juan E Abrahante
- University of Minnesota Informatics Institute, Minneapolis, MN, 55455, USA
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunology Center, Northwestern University, Chicago, IL, 60611, USA.
| | - Xunrong Luo
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Biostatistics Collaboration Center, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Duke Transplant Center, Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
9
|
Zhang J, Liu W, Xie T, Huang L, Hu Y, Wen B, Tang P, Guo F, Jin K, Zhang P, Niu L, Liu Z, Qu X. Elevated LAG-3 on CD4 + T cells negatively correlates with neutralizing antibody response during HCV infection. Immunol Lett 2019; 212:46-52. [PMID: 31216427 DOI: 10.1016/j.imlet.2019.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/27/2019] [Accepted: 06/14/2019] [Indexed: 01/18/2023]
Abstract
Lymphocyte activation gene-3 (LAG-3), an inhibitory molecule, which has been shown co-expressed with multiple inhibitory receptors on CD8+ T and natural killer (NK) cells and negatively regulates T and NK cell responses during hepatitis C virus (HCV) infection. However, whether LAG-3 is involved in the regulation of the antibody response remains unclear. This study aims to investigate the relationship of LAG-3 with neutralizing antibody (nAb) response during HCV infection. A total of 66 HCV-infected individuals and 36 sex- and age-matched healthy controls from a population of intravenous drug users were recruited. Circulating follicular helper T (cTfh) cells and LAG-3-expressing CD4+ T cells, type 1 regulatory T (Tr1) cells, and regulatory T (Treg) cells were characterized by flow cytometry. Serum nAb response of HCV-infected individuals was determined using pseudoparticle neutralization assays. We found that HCV infection enhanced LAG-3 expression on CD4+ T cells and exhibited regulatory T cell-like phenotype and inversely associated with the HCV nAb response. Further analysis showed that frequency of CXCR3+ cTfh cells positively correlated with nAb response, however LAG-3+ CD4+ T cells inversely associated with CXCR3+ cTfh cells. This study observed that LAG-3+ CD4+ T cells exhibit a regulatory cell phenotype and negatively associate with the HCV nAb response, implying that LAG-3 may be involved in the negative regulation of humoral immunity during HCV infection.
Collapse
Affiliation(s)
- Jian Zhang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Wenpei Liu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China; Affiliated The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, Hunan, 423000, China
| | - Ting Xie
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Liyan Huang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Yabin Hu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Bo Wen
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Ping Tang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Fengfan Guo
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Kun Jin
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Ping Zhang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Ling Niu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Ziyan Liu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China
| | - Xiaowang Qu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, 423000, China; Affiliated The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, Hunan, 423000, China.
| |
Collapse
|
10
|
Tumangelova-Yuzeir K, Naydenov E, Ivanova-Todorova E, Krasimirova E, Vasilev G, Nachev S, Kyurkchiev D. Mesenchymal Stem Cells Derived and Cultured from Glioblastoma Multiforme Increase Tregs, Downregulate Th17, and Induce the Tolerogenic Phenotype of Monocyte-Derived Cells. Stem Cells Int 2019; 2019:6904638. [PMID: 31191680 PMCID: PMC6525812 DOI: 10.1155/2019/6904638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/08/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess immunosuppressive properties and have been described in the tumor microenvironment of glioblastoma multiforme (GBM). This manuscript has two major topics-first, to describe isolated and cultured MSCs derived from GBM (GB-MSCs) and second, to examine their in vitro immunosuppressive capacity. Our results display cells with morphology and phenotype, clonogenic ability, and osteogenic potential, typical for MSCs. Furthermore, the cultured cells show intracellular expression of the neural markers Nestin and GFAP. They express PD-L1 and secrete TGFβ, CCL-2, PGE2, IL-6, and sVEGF. Coculturing of GB-MSCs with PBMCs isolated from healthy donors results in a decreased percentage of Th17 lymphocytes and an increased percentage of Tregs. Regarding the impact of GB-MSCs on monocytes, we establish an augmented expression of CD14 and CD86 along with diminished expression of HLA-DR and CD80, which is associated with tolerogenic phenotype monocyte-derived cells. In conclusion, our results describe in detail GBM-derived and cultured cells that meet the criteria for MSCs but at the same time express Nestin and GFAP. GB-MSCs express and secrete suppressive molecules, influencing in vitro T cells and monocytes, and are probably another factor involved in the immune suppression exerted by GBM.
Collapse
Affiliation(s)
- Kalina Tumangelova-Yuzeir
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Emanuil Naydenov
- Clinic of Neurosurgery, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Ekaterina Krasimirova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Georgi Vasilev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Sevdalin Nachev
- Laboratory of Clinical Pathology, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| |
Collapse
|
11
|
Chrobák P. Control of T Cell Responses, Tolerance and Autoimmunity by Regulatory T Cells: Current Concepts. ACTA MEDICA (HRADEC KRÁLOVÉ) 2019. [DOI: 10.14712/18059694.2019.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Regulatory T cells have emerged as an important mechanism of regulating tolerance and T cell responses. CD4+ regulatory T cells can be divided into two main groups, natural regulatory T cells, which express high levels of CD25 on their cell surface and phenotypically diverse adaptive (antigen induced) regulatory T cells. Natural regulatory T cells are made in the thymus, and require strong costimulatory signals for induction and maintenance, express a transcription factor called Foxp3, and function by a largely unknown mechanism. Adaptive (antigen induced) regulatory T cells are made by sub-optimal antigenic signals in the periphery, in the presence of immunosuppressive cytokines, often in special circumstances, such as chronic viral infections or after mucosal administration of antigen, and rely on cytokines such as IL-10 and TGF-β for suppression. Regulatory T cells offer a great potential for the treatment of autoimmune diseases and during transplantation.
Collapse
|
12
|
Herzog RW, Kuteyeva V, Saboungi R, Terhorst C, Biswas M. Reprogrammed CD4 + T Cells That Express FoxP3 + Control Inhibitory Antibody Formation in Hemophilia A Mice. Front Immunol 2019; 10:274. [PMID: 30842776 PMCID: PMC6391332 DOI: 10.3389/fimmu.2019.00274] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/31/2019] [Indexed: 01/16/2023] Open
Abstract
Coagulation Factor VIII (FVIII) replacement therapy in hemophilia A patients is complicated by the development of inhibitory antibodies, which often render the treatment ineffective. Previous studies demonstrated a strong correlation between induction of regulatory T cells (Treg) and tolerance to the therapeutic protein. We, therefore, set out to evaluate whether the adoptive transfer of FVIII-specific CD4+ Treg cells prevents inhibitor response to FVIII protein therapy. To this end, we first retrovirally transduced FoxP3+ into FVIII-specific CD4+ cells, which resulted in cells that stably express FoxP3, are phenotypically similar to peripherally induced Tregs and are antigen specific suppressors, as judged by in vitro assays. Upon transfer of the FVIII-specific CD4+ FoxP3+ cells into hemophilia A mice, development of inhibitory antibodies in response to administering FVIII protein was completely suppressed. Suppression was extended for 2 months, even after transferred cells were no longer detectable in the secondary lymphoid organs of recipient animals. Upon co-transfer of FoxP3+-transduced cells with the B cell depleting anti-CD20 into mice with pre-existing inhibitory antibodies to FVIII, the escalation of inhibitory antibody titers in response to subsequent FVIII protein therapy was dramatically reduced. We conclude that reprogramed FoxP3 expressing cells are capable of inducing the in vivo conversion of endogenous FVIII peripheral Tregs, which results in sustained suppression of FVIII inhibitors caused by replacement therapy in recipient hemophilia A animals.
Collapse
Affiliation(s)
- Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Veronica Kuteyeva
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Rania Saboungi
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States,*Correspondence: Moanaro Biswas
| |
Collapse
|
13
|
Wang X, Guo G, Guan H, Yu Y, Lu J, Yu J. Challenges and potential of PD-1/PD-L1 checkpoint blockade immunotherapy for glioblastoma. J Exp Clin Cancer Res 2019; 38:87. [PMID: 30777100 PMCID: PMC6380009 DOI: 10.1186/s13046-019-1085-3] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/06/2019] [Indexed: 01/23/2023] Open
Abstract
PD-1/PD-L1 checkpoint blockades have achieved significant progress in several kinds of tumours. Pembrolizumab, which targets PD-1, has been approved as a first-line treatment for advanced non-small cell lung cancer (NSCLC) patients with positive PD-L1 expression. However, PD-1/PD-L1 checkpoint blockades have not achieved breakthroughs in treating glioblastoma because glioblastoma has a low immunogenic response and an immunosuppressive microenvironment caused by the precise crosstalk between cytokines and immune cells. A phase III clinical trial, Checkmate 143, reported that nivolumab, which targets PD-1, did not demonstrate survival benefits compared with bavacizumab in recurrent glioblastoma patients. Thus, the combination of a PD-1/PD-L1 checkpoint blockade with RT, TMZ, antibodies targeting other inhibitory or stimulatory molecules, targeted therapy, and vaccines may be an appealing solution aimed at achieving optimal clinical benefit. There are many ongoing clinical trials exploring the efficacy of various approaches based on PD-1/PD-L1 checkpoint blockades in primary or recurrent glioblastoma patients. Many challenges need to be overcome, including the identification of discrepancies between different genomic subtypes in their response to PD-1/PD-L1 checkpoint blockades, the selection of PD-1/PD-L1 checkpoint blockades for primary versus recurrent glioblastoma, and the identification of the optimal combination and sequence of combination therapy. In this review, we describe the immunosuppressive molecular characteristics of the tumour microenvironment (TME), candidate biomarkers of PD-1/PD-L1 checkpoint blockades, ongoing clinical trials and challenges of PD-1/PD-L1 checkpoint blockades in glioblastoma.
Collapse
Affiliation(s)
- Xin Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province China
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, 250117 Shandong Province China
| | - Gaochao Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Hui Guan
- Department of Radiation Oncology, The Fourth People’s Hospital of Jinan, Jinan, Shandong Province China
| | - Yang Yu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, 250117 Shandong Province China
| | - Jie Lu
- Department of Neurosurgery, Shandong Province Qianfoshan Hospital of Shandong University, Shandong Province, Jinan, 250014 China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, 250117 Shandong Province China
| |
Collapse
|
14
|
Kitz A, Singer E, Hafler D. Regulatory T Cells: From Discovery to Autoimmunity. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029041. [PMID: 29311129 DOI: 10.1101/cshperspect.a029041] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a genetically mediated autoimmune disease of the central nervous system. Allelic variants lead to lower thresholds of T-cell activation resulting in activation of autoreactive T cells. Environmental factors, including, among others, diet, vitamin D, and smoking, in combination with genetic predispositions, play a substantial role in disease development and activation of autoreactive T cells. FoxP3+ regulatory T cells (Tregs) have emerged as central in the control of autoreactive T cells. A consistent finding in patients with MS is defects in Treg cell function with reduced suppression of effector T cells and production of proinflammatory cytokines. Emerging data suggests that functional Tregs become effector-like T cells with loss of function associated with T-bet expression and interferon γ (IFN-γ) secretion.
Collapse
Affiliation(s)
- Alexandra Kitz
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - Emily Singer
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - David Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
15
|
Woods DM, Ramakrishnan R, Laino AS, Berglund A, Walton K, Betts BC, Weber JS. Decreased Suppression and Increased Phosphorylated STAT3 in Regulatory T Cells are Associated with Benefit from Adjuvant PD-1 Blockade in Resected Metastatic Melanoma. Clin Cancer Res 2018; 24:6236-6247. [PMID: 30131384 DOI: 10.1158/1078-0432.ccr-18-1100] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/10/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE PD-1 blockade induces durable responses in patients with metastatic melanoma and prolongs relapse-free survival in patients with resected melanoma; however, current biomarkers do not consistently associate with patient responses. In this study, we investigated the impact of nivolumab therapy on peripheral blood regulatory T cells (Treg) and its relation to patient outcomes. EXPERIMENTAL DESIGN Peripheral blood Tregs and conventional CD4+ T cells from patients with resected high-risk melanoma treated with adjuvant nivolumab were assessed for gene expression changes by RNA-seq. Percentages of circulating Tregs and phosphorylated-STAT3 (pSTAT3) expression levels were assessed by flow cytometry and validated in an independent cohort of active disease patients. Suppressive function of Tregs was assessed in allogeneic mixed lymphocyte reactions. RESULTS Tregs from non-relapse patients had increased expression of proliferation associated genes. An increase in the proportion of circulating Tregs and pSTAT3 expression and a reduction in Treg-suppressive capacity were observed in non-relapsing, but not relapsing patient samples 13 weeks after starting treatment. In vitro blockade of PD-1 increased Treg percentages and pSTAT3 expression, and reduced Treg-suppressive function. PD-1 blockade also led to IL10 production by T cells, resulting in higher Treg proliferation. The addition of a STAT3 inhibitor ameliorated the increase in Tregs, enhanced suppressive function, and decreased T-cell IL10 production in vitro. CONCLUSIONS These results demonstrate that induction of pSTAT3, reduced suppressive function, and a paradoxical increase in Treg proliferation are novel correlates of patient benefit from PD-1 blockade.
Collapse
Affiliation(s)
- David M Woods
- New York University Langone Medical Center, New York, New York.
| | | | | | - Anders Berglund
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kelly Walton
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Brian C Betts
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jeffrey S Weber
- New York University Langone Medical Center, New York, New York
| |
Collapse
|
16
|
Ou Y, Cannon MJ, Nakagawa M. Regulatory T Cells in Gynecologic Cancer. MOJ IMMUNOLOGY 2018; 6:34-42. [PMID: 30637330 PMCID: PMC6329475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Increasing evidence supports that regulatory T cells (Tregs) within the tumor, tumor draining lymph nodes, ascites and peripheral blood of patients with cancer are associated with poor prognosis. Tregs are important mediators of active immune evasion in cancer. In this review, the potential mechanisms of Treg actions and the roles of Tregs specifically in the tumor microenvironment derived from three types of gynecological cancers, cervical, vulvar and ovarian, are described. The correlations between Tregs and clinical immunotherapeutic study outcomes are discussed. Successful modulation of Tregs would likely have significant impact on the effectiveness of immunotherapeutic treatments in cancer patients.
Collapse
Affiliation(s)
- Yang Ou
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Martin J. Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Mayumi Nakagawa
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
17
|
Generation and Characterization of Alloantigen-Specific Regulatory T Cells For Clinical Transplant Tolerance. Sci Rep 2018; 8:1136. [PMID: 29348660 PMCID: PMC5773708 DOI: 10.1038/s41598-018-19621-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/05/2018] [Indexed: 01/28/2023] Open
Abstract
Donor-specific CD4+CD127−CD25+FOXP3+ regulatory T cells (AgTregs) have the potential to induce clinical transplant tolerance; however, their expansion ex vivo remains challenging. We optimized a novel expansion protocol to stimulate donor-specific Tregs using soluble 4-trimer CD40 ligand (CD40L)-activated donor B cells that expressed mature antigen-presenting cell markers. This avoided the use of CD40L-expressing stimulator cells that might otherwise result in potential cellular contamination. Purified allogeneic “recipient” CD4+CD25+ Tregs were stimulated on days 0 and 7 with expanded “donor” B cells in the presence of IL-2, TGFβ and sirolimus (SRL). Tregs were further amplified by polyclonal stimulation with anti-CD3/CD28 beads on day 14 without SRL, and harvested on day 21, with extrapolated fold expansion into the thousands. The expanded AgTregs maintained expression of classical Treg markers including demethylation of the Treg-specific demethylated region (CNS2) and also displayed constricted TcR repertoire. We observed AgTregs more potently inhibited MLR than polyclonally expanded Tregs and generated new Tregs in autologous responder cells (a measure of infectious tolerance). Thus, an optimized and more clinically applicable protocol for the expansion of donor-specific Tregs has been developed.
Collapse
|
18
|
|
19
|
Yang H, Cong Y, Wu T, Tang H, Ma M, Zeng J, Zhang W, Lian Z, Yang X. Clinical efficacy of Yingliu mixture combined with metimazole for treating diffuse goitre with hyperthyroidism and its impact on related cytokines. PHARMACEUTICAL BIOLOGY 2017; 55:258-263. [PMID: 27927064 PMCID: PMC6130729 DOI: 10.1080/13880209.2016.1260595] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 09/08/2016] [Accepted: 11/09/2016] [Indexed: 06/06/2023]
Abstract
CONTEXT Yingliu mixture was developed in 1990s by Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, for treating diffuse goitre with hyperthyroidism (Graves' disease, GD). Former studies have shown Yingliu mixture combined with methimazole (Y-M) can effectively improve thyroid function and decrease thyrotropin-receptor antibody level. Furthermore, we researched its impact on related cytokines to prove that Y-M improve patients' immunity status. OBJECTIVE To observe the clinical efficacy of Y-M for treating GD. METHODS A total of 120 GD patients were randomly divided into two groups, the treatment and the control groups (n = 60). The treatment group's patients were treated with Y-M. The control group's patients were treated with methimazole alone. Yingliu mixture was orally administered, 25 mL three times daily. Methimazole was administered at 5-25 mg/day. After 12 weeks of the treatment, the cytokines, antibodies related to thyroid function, and Chinese medical syndromes were evaluated. RESULTS After the treatment, the free triiodothyronine and thyroxine levels in both groups decreased. The thyroid-stimulating hormone level increased in the treatment group. The thyrotropin-receptor antibody levels and TNF-α levels decreased in both groups. In the control group, IL-6 and IFN-γ levels were lower than that before the treatment. In the treatment group, CD4+ and CD25+ levels were higher than pretreatment levels, but IL-10 levels were reduced. CLINICAL SYMPTOMS the total CMS scores for both groups decreased. CONCLUSIONS The Y-M combination can improve thyroid function, and decrease autoantibodies, cytokines, and clinical symptoms, so its efficacy may surpass that of methimazole alone.
Collapse
Affiliation(s)
- Hua Yang
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yilei Cong
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tengfei Wu
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Tang
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Muhao Ma
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Juanhua Zeng
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenya Zhang
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen Lian
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyu Yang
- Department of Endocrinology, Affiliated Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Li GG, Cao YH, Run Y, Xu RX, Zheng ZD. Inhibition of CD8 + T cells and elimination of myeloid cells by CD4 + Foxp3 - T regulatory type 1 cells in acute respiratory distress syndrome. Clin Exp Pharmacol Physiol 2017; 43:1191-1198. [PMID: 27558304 DOI: 10.1111/1440-1681.12656] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 11/28/2022]
Abstract
Acute lung injury and acute respiratory distress syndrome (ARDS) are caused by rapid-onset bilateral pulmonary inflammation. We therefore investigated the potential role of interleukin (IL)-10+ CD4+ Tr1 cells, a regulatory T cell subset with previously identified immunosuppressive functions, in ARDS patients. We first showed that circulating Tr1 cells were upregulated in active and resolved ARDS patients compared to healthy controls and pneumonia patient controls. A significant fraction of these Tr1 cells expressed granzyme B and perforin, while most Tr1 cells did not express interferon gamma (IFN-γ), IL-4, IL-17 or FOXP3, suggesting that the effector functions of these Tr1 cells were primarily mediated by IL-10, granzyme B, and perforin. Indeed, Tr1 cells effectively suppressed CD8+ T cell IFN-γ production and induced lysis of monocytes and dendritic cells in vitro. The elimination of myeloid antigen-presenting cells depended on granzyme B production. We also discovered that Tr1 cells could be identified in the bronchoalveolar lavage fluid collected from ARDS patients. All these results suggested that Tr1 cells possessed the capacity to downregulate inflammation in ARDS. In support of this, we found that ARDS patients who resolved the inflammation and survived the syndrome contained significantly higher levels of Tr1 cells than ARDS patients who succumbed to the syndrome. Overall, this report added a novel piece of evidence that ARDS could be intervened by regulatory T cell-mediated suppressive mechanisms.
Collapse
Affiliation(s)
- Guang-Gang Li
- Affiliated Bayi Brain Hospital, PLA Army General Hospital, Beijing, China
| | - Ying-Hua Cao
- Affiliated Bayi Brain Hospital, PLA Army General Hospital, Beijing, China
| | - Yue Run
- Department of Oncology, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, PLA Army General Hospital, Beijing, China
| | - Zhen-Dong Zheng
- Department of Oncology, General Hospital of Shenyang Military Area Command, Shenyang, China
| |
Collapse
|
21
|
Horton C, Shanmugarajah K, Fairchild PJ. Harnessing the properties of dendritic cells in the pursuit of immunological tolerance. Biomed J 2017; 40:80-93. [PMID: 28521905 PMCID: PMC6138597 DOI: 10.1016/j.bj.2017.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
The acquisition of self-perpetuating, immunological tolerance specific for graft alloantigens has long been described as the "holy grail" of clinical transplantation. By removing the need for life-long immunosuppression following engraftment, the adverse consequences of immunosuppressive regimens, including chronic infections and malignancy, may be avoided. Furthermore, autoimmune diseases and allergy are, by definition, driven by aberrant immunological responses to ordinarily innocuous antigens. The re-establishment of permanent tolerance towards instigating antigens may, therefore, provide a cure to these common diseases. Whilst various cell types exhibiting a tolerogenic phenotype have been proposed for such a task, tolerogenic dendritic cells (tol-DCs) are exquisitely adapted for antigen presentation and interact with many facets of the immune system: as such, they are attractive candidates for use in strategies for immune intervention. We review here our current understanding of tol-DC mediated induction and maintenance of immunological tolerance. Additionally, we discuss recent in vitro findings from animal models and clinical trials of tol-DC immunotherapy in the setting of transplantation, autoimmunity and allergy which highlight their promising therapeutic potential, and speculate how tol-DC therapy may be developed in the future.
Collapse
|
22
|
Therapeutic Treatment of Arthritic Mice with 15-Deoxy Δ 12,14-Prostaglandin J 2 (15d-PGJ 2) Ameliorates Disease through the Suppression of Th17 Cells and the Induction of CD4 +CD25 -FOXP3 + Cells. Mediators Inflamm 2016; 2016:9626427. [PMID: 27872515 PMCID: PMC5107840 DOI: 10.1155/2016/9626427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/20/2016] [Accepted: 08/04/2016] [Indexed: 12/20/2022] Open
Abstract
The prostaglandin, 15-deoxy Δ12,14-prostaglandin J2 (15d-PGJ2), is a lipid mediator that plays an important role in the control of chronic inflammatory disease. However, the role of prostanoid in rheumatoid arthritis (RA) is not well determined. We demonstrated the therapeutic effect of 15d-PGJ2 in an experimental model of arthritis. Daily administration of 15d-PGJ2 attenuated the severity of CIA, reducing the clinical score, pain, and edema. 15d-PGJ2 treatment was associated with a marked reduction in joint levels of proinflammatory cytokines. Although the mRNA expression of ROR-γt was profoundly reduced, FOXP3 was enhanced in draining lymph node cells from 15d-PGJ2-treated arthritic mice. The specific and polyclonal CD4+ Th17 cell responses were limited during the addition of prostaglandin to cell culture. Moreover, in vitro 15d-PGJ2 increased the expression of FOXP3, GITR, and CTLA-4 in the CD4+CD25− population, suggesting the induction of Tregs on conventional T cells. Prostanoid addition to CD4+CD25− cells selectively suppressed Th17 differentiation and promoted the enhancement of FOXP3 under polarization conditions. Thus, 15d-PGJ2 ameliorated symptoms of collagen-induced arthritis by regulating Th17 differentiation, concomitant with the induction of Tregs, and, consequently, protected mice from diseases aggravation. Altogether, these results indicate that 15d-PGJ2 may represent a potential therapeutic strategy in RA.
Collapse
|
23
|
Tu JF, Ding YH, Ying XH, Wu FZ, Zhou XM, Zhang DK, Zou H, Ji JS. Regulatory T cells, especially ICOS + FOXP3 + regulatory T cells, are increased in the hepatocellular carcinoma microenvironment and predict reduced survival. Sci Rep 2016; 6:35056. [PMID: 27725696 PMCID: PMC5057140 DOI: 10.1038/srep35056] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 09/23/2016] [Indexed: 01/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumour, especially in Asia. Its prognosis is poor, and there are limited methods for predicting patient survival. This study was carried out to analyse the prognostic value of tumour-infiltrating lymphocytes (TILs), especially regulatory T cells (Tregs), in HCC patients. TILs were analysed in 57 randomly selected HCC patients. The prognostic effects of groups with high and low numbers were evaluated by the Kaplan-Meier and Cox model analyses. Although higher densities of CD3+, CD4+, and CD8+ cytotoxic lymphocytes (CTLs) as well as CD56+ NK cells and CD68+ macrophages were observed in peritumoural tissue, increased numbers of forkhead/winged helix transcription factor P3+ (FOXP3+) Tregs were found in intratumoural tissue. Additionally, regarding ICOS+ FOXP3+ Tregs, an increased prevalence in carcinoma was not only associated with the absolute number but also with the percentage of FOXP3+ cells. Higher Treg levels in tumour tissues indicated a worse prognosis, and the FOXP3+ Tregs/CD4+ T cells ratio was an independent prognostic factor for OS. Therefore, FOXP3+ Tregs, especially ICOS+ FOXP3+ Tregs, contribute to the immunosuppressive HCC microenvironment. High tumour-infiltrating Tregs are thought to be an unfavourable prognostic indicator of HCC.
Collapse
Affiliation(s)
- Jian-Fei Tu
- Department of Radiology and Interventional Radiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Xi-Hui Ying
- Department of Radiology and Interventional Radiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Fa-Zong Wu
- Department of Radiology and Interventional Radiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Xin-Mu Zhou
- Department of Pathology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Deng-Ke Zhang
- Department of Radiology and Interventional Radiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Jian-Song Ji
- Department of Radiology and Interventional Radiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
24
|
de Oliveira Bravo M, Carvalho JL, Saldanha-Araujo F. Adenosine production: a common path for mesenchymal stem-cell and regulatory T-cell-mediated immunosuppression. Purinergic Signal 2016; 12:595-609. [PMID: 27557887 DOI: 10.1007/s11302-016-9529-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022] Open
Abstract
Adenosine is an important molecule that exerts control on the immune system, by signaling through receptors lying on the surface of immune cells. This nucleotide is produced, in part, by the action of the ectoenzymes CD39 and CD73. Interestingly, these proteins are expressed on the cell surface of regulatory T-cells (Tregs) and mesenchymal stromal cells (MSCs)-two cell populations that have emerged as potential therapeutic tools in the field of cell therapy. In fact, the production of adenosine constitutes a mechanism used by both cell types to control the immune response. Recently, great scientific progress was obtained regarding the role of adenosine in the inflammatory environment. In this context, the present review focuses on the advances related to the impact of adenosine production over the immune modulatory activity of Tregs and MSCs, and how this nucleotide controls the biological functions of these cells. Finally, we mention the main challenges and hurdles to bring such molecule to clinical settings.
Collapse
Affiliation(s)
| | - Juliana Lott Carvalho
- Genomic Sciences and Biotechnology Center, Catholic University of Brasilia, Brasilia, Brazil
| | | |
Collapse
|
25
|
Gao X, Arpin C, Marvel J, Prokopiou SA, Gandrillon O, Crauste F. IL-2 sensitivity and exogenous IL-2 concentration gradient tune the productive contact duration of CD8(+) T cell-APC: a multiscale modeling study. BMC SYSTEMS BIOLOGY 2016; 10:77. [PMID: 27535120 PMCID: PMC4989479 DOI: 10.1186/s12918-016-0323-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/21/2016] [Indexed: 01/17/2023]
Abstract
Background The CD8+ T cell immune response fights acute infections by intracellular pathogens and, by generating an immune memory, enables immune responses against secondary infections. Activation of the CD8+ T cell immune response involves a succession of molecular events leading to modifications of CD8+ T cell population. To understand the endogenous and exogenous mechanisms controlling the activation of CD8+ T cells and to investigate the influence of early molecular events on the long-term cell population behavior, we developed a multiscale computational model. It integrates three levels of description: a Cellular Potts model describing the individual behavior of CD8+ T cells, a system of ordinary differential equations describing a decision-making molecular regulatory network at the intracellular level, and a partial differential equation describing the diffusion of IL-2 in the extracellular environment. Results We first calibrated the model parameters based on in vivo data and showed the model’s ability to reproduce early dynamics of CD8+ T cells in murine lymph nodes after influenza infection, both at the cell population and intracellular levels. We then showed the model’s ability to reproduce the proliferative responses of CD5hi and CD5lo CD8+ T cells to exogenous IL-2 under a weak TCR stimulation. This stressed the role of short-lasting molecular events and the relevance of explicitly describing both intracellular and cellular scale dynamics. Our results suggest that the productive contact duration of CD8+ T cell-APC is influenced by the sensitivity of individual CD8+ T cells to the activation signal and by the IL-2 concentration in the extracellular environment. Conclusions The multiscale nature of our model allows the reproduction and explanation of some acquired characteristics and functions of CD8+ T cells, and of their responses to multiple stimulation conditions, that would not be accessible in a classical description of cell population dynamics that would not consider intracellular dynamics. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0323-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuefeng Gao
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France
| | - Christophe Arpin
- Inserm, U1111, Lyon, F-69007, France.,CNRS, UMR5308, Lyon, F-69007, France.,Centre International de Recherche en Infectiologie, Université Lyon 1, Lyon, F-69007, France.,Ecole Normale Supérieure de Lyon, Lyon, F-69007, France
| | - Jacqueline Marvel
- Inserm, U1111, Lyon, F-69007, France.,CNRS, UMR5308, Lyon, F-69007, France.,Centre International de Recherche en Infectiologie, Université Lyon 1, Lyon, F-69007, France.,Ecole Normale Supérieure de Lyon, Lyon, F-69007, France
| | - Sotiris A Prokopiou
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France
| | - Olivier Gandrillon
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France. .,Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d'Italie Site Jacques Monod, F-69007, Lyon, France.
| | - Fabien Crauste
- Inria team Dracula, Inria Antenne Lyon la Doua, Bâtiment CEI-2, 56 Boulevard Niels Bohr, 69603, Villeurbanne cedex, France. .,Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5208, Institut Camille Jordan, 43 blvd. du 11 novembre 1918, F-69622, Villeurbanne cedex, France.
| |
Collapse
|
26
|
Arefanian H, Tredget EB, Mok DCM, Ramji Q, Rafati S, Rodriguez-Barbosa J, Korbutt GS, Rajotte RV, Gill RG, Rayat GR. Porcine Islet-Specific Tolerance Induced by the Combination of Anti-LFA-1 and Anti-CD154 mAbs is Dependent on PD-1. Cell Transplant 2016; 25:327-42. [DOI: 10.3727/096368915x688506] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We previously demonstrated that short-term administration of a combination of anti-LFA-1 and anti-CD154 monoclonal antibodies (mAbs) induces tolerance to neonatal porcine islet (NPI) xenografts that is mediated by regulatory T cells (Tregs) in B6 mice. In this study, we examined whether the coinhibitory molecule PD-1 is required for the induction and maintenance of tolerance to NPI xenografts. We also determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or xenogeneic rat islet grafts since we previously demonstrated that tolerance to NPI xenografts could be extended to second-party NPI xenografts. Finally, we determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or second-party porcine skin grafts. Diabetic B6 mice were transplanted with 2,000 NPIs under the kidney capsule and treated with short-term administration of a combination of anti-LFA-1 and anti-CD154 mAbs. Some of these mice were also treated simultaneously with anti-PD-1 mAb at >150 days posttransplantation. Spleen cells from some of the tolerant B6 mice were used for proliferation assays or were injected into B6 rag-/- mice with established islet grafts from allogeneic or xenogeneic donors. All B6 mice treated with anti-LFA-1 and anti-CD154 mAbs achieved and maintained normoglycemia until the end of the study; however, some mice that were treated with anti-PD-1 mAb became diabetic. All B6 rag-/- mouse recipients of first- and second-party NPIs maintained normoglycemia after reconstitution with spleen cells from tolerant B6 mice, while all B6 rag-/- mouse recipients of allogeneic mouse or xenogeneic rat islets rejected their grafts after cell reconstitution. Tolerant B6 mice rejected their allogeneic mouse or xenogeneic second-party porcine skin grafts while remaining normoglycemic until the end of the study. These results show that porcine islet-specific tolerance is dependent on PD-1, which could not be extended to skin grafts.
Collapse
Affiliation(s)
- Hossein Arefanian
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
- Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Pancreatic Islet Biology and Transplantation Unit, Dasman Diabetes Institute, Kuwait, Dasman, Kuwait
| | - Eric B. Tredget
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Dereck C. M. Mok
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Qahir Ramji
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Shahin Rafati
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Jose Rodriguez-Barbosa
- Institute of Biomedicine (Immunobiology), University of Leon, Campus de Vegazana s/n, Leon, Spain
| | - Gregory S. Korbutt
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ray V. Rajotte
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ron G. Gill
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Gina R. Rayat
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Hahn SA, Bellinghausen I, Trinschek B, Becker C. Translating Treg Therapy in Humanized Mice. Front Immunol 2015; 6:623. [PMID: 26697017 PMCID: PMC4677486 DOI: 10.3389/fimmu.2015.00623] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/30/2022] Open
Abstract
Regulatory T cells (Treg) control immune cell function as well as non-immunological processes. Their far-reaching regulatory activities suggest their functional manipulation as a means to sustainably and causally intervene with the course of diseases. Preclinical tools and strategies are however needed to further test and develop interventional strategies outside the human body. “Humanized” mouse models consisting of mice engrafted with human immune cells and tissues provide new tools to analyze human Treg ontogeny, immunobiology, and therapy. Here, we summarize the current state of humanized mouse models as a means to study human Treg function at the molecular level and to design strategies to harness these cells for therapeutic purposes.
Collapse
Affiliation(s)
- Susanne A Hahn
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Iris Bellinghausen
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Bettina Trinschek
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| |
Collapse
|
28
|
Shi C, Zhang Y, Yang H, Dong T, Chen Y, Xu Y, Yang X. Combined effect of ultrasound/SonoVue microbubble on CD4(+)CD25(+) regulatory T cells viability and optimized parameters for its transfection. ULTRASONICS 2015; 62:97-102. [PMID: 26048174 DOI: 10.1016/j.ultras.2015.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/15/2015] [Accepted: 05/14/2015] [Indexed: 06/04/2023]
Abstract
The purpose of this study was to investigate the combined effect of ultrasound and SonoVue microbubble on CD4(+)CD25(+) regulatory T cells (Tregs) viability and to explore the appropriate parameters for Tregs transfection. Tregs were separated from peripheral venous blood of patients with hepatocellular carcinoma and seeded in 96-well plates. The optimal ultrasound exposure time and optimal SonoVue microbubble concentration for Tregs were measured by mechanical index (MI) of 1.2 or 1.4, exposure time of 0, 30, 60, 90, 120, 150, 180s, and 0, 10, 20, 30, 40, 50μL/100μL microbubble per well, respectively. In addition, the combined effect of ultrasound and microbubble on Tregs viability was evaluated according to the following parameters: MI 1.2/1.4+exposure time of 120, 150, 180s+0, 10, 20, 30, 40, 50μL/100μL microbubble per well. Tregs viability investigations were performed in order to explore the optimal transfection condition. The efficiency of plasmid transfer was determined by detection of luciferase activity on the microscopic examinations. The proliferation of Tregs could be promoted by ultrasound exposures, while being decreased with the increasing concentration of microbubbles. Under the current experimental conditions, the optimal ultrasound parameters were MI=1.4 and exposure time=150/180s. The optimal microbubble concentration was 10μL/100μL. Compared with treatment with ultrasound or microbubbles alone, the transfection efficiency of Tregs improved 50% by combining ultrasound and microbubble. The results indicate that both ultrasound and microbubble could affect the Tregs proliferation and the optimal Treg transfection rate was obtained by treating with 10% microbubbles and ultrasound exposure for 150/180s under ultrasound MI of 1.4.
Collapse
Affiliation(s)
- Chunying Shi
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Yu Zhang
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Haichao Yang
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Tianxiu Dong
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Yaodong Chen
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Yutong Xu
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China
| | - Xiuhua Yang
- Department of Abdomind Ultrasound, The First Clinic Hospital Harbin Medical University Heilongjiang, Harbin 150001, China.
| |
Collapse
|
29
|
Liu K, Yang K, Wu B, Chen H, Chen X, Chen X, Jiang L, Ye F, He D, Lu Z, Xue L, Zhang W, Li Q, Zhou Z, Mo X, Hu J. Tumor-Infiltrating Immune Cells Are Associated With Prognosis of Gastric Cancer. Medicine (Baltimore) 2015; 94:e1631. [PMID: 26426650 PMCID: PMC4616881 DOI: 10.1097/md.0000000000001631] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023] Open
Abstract
Immune cells contribute to determining the prognosis of gastric cancer. However, their exact role is less clear. We determined the prognostic significance of different immune cells in intratumoral tissue (T), stromal tissue (S), and adjacent normal tissue (N) of 166 gastric cancer cases and their interactions, including CD3+, CD4+, CD8, CD57+, CD68+, CD66b+, and Foxp3+ cells, and established an effective prognostic nomogram based on the immune reactions. We found high densities of TCD3+, TCD4+, TCD8+, SCD3+, SCD4+, SCD57+, SCD66b+, and NFoxp3+ cells, as well as high TCD8+/SCD8+ ratio, TCD68+/SCD68+ ratio, TCD3+/TFoxp3+ ratio, TCD4+/TFoxp3+ ratio, TCD8+/TFoxp3+ ratio, SCD3+/SFoxp3+ ratio, and SCD4+/SCD8+ ratio were associated with better survival, whereas high densities of TCD66b+, TFoxp3+, SFoxp3+ and NCD66b+ cells as well as high TCD57+/SCD57+ ratio, TCD66b+/SCD66b+ ratio, SCD8+/SFoxp3+ ratio, and TFoxp3+/NFoxp3+ ratio were associated with significantly worse outcome. Multivariate analysis indicated that tumor size, longitudinal tumor location, N stage, TCD68+/SCD68+ ratio, TCD8+/TFoxp3+ ratio, density of TFoxp3+ cells, and TCD66b+/SCD66b+ ratio were independent prognostic factors, which were all selected into the nomogram. The calibration curve for likelihood of survival demonstrated favorable consistency between predictive value of the nomogram and actual observation. The C-index (0.83, 95% CI: 0.78 to 0.87) of our nomogram for predicting prognosis was significantly higher than that of TNM staging system (0.70). Collectively, high TCD68+/SCD68+ ratio and TCD8+/TFoxp3+ ratio were associated with improved overall survival, whereas high density of TFoxp3+ cells and TCD66b+/SCD66b+ ratio demonstrated poor overall survival, which are promising independent predictors for overall survival in gastric cancer.
Collapse
Affiliation(s)
- Kai Liu
- From the Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China (KL,KY, BW,HC, XC, XC, LX, WZ, ZZ, JH); Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China (KL,KY, BW,HC, XC, XC, LX, WZ, JH); Department of Pathology, West China Hospital, Sichuan University, China (LJ, DH); Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (FY); Department of Oncology, West China Hospital, Sichuan University, China (QL); and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China (XM)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Carregaro V, Ribeiro JM, Valenzuela JG, Souza-Júnior DL, Costa DL, Oliveira CJF, Sacramento LA, Nascimento MSL, Milanezi CM, Cunha FQ, Silva JS. Nucleosides present on phlebotomine saliva induce immunossuppression and promote the infection establishment. PLoS Negl Trop Dis 2015; 9:e0003600. [PMID: 25849562 PMCID: PMC4388340 DOI: 10.1371/journal.pntd.0003600] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/05/2015] [Indexed: 01/30/2023] Open
Abstract
Background Sand fly saliva plays a crucial role in establishing Leishmania infection. We identified adenosine (ADO) and adenosine monophosphate (AMP) as active pharmacologic compounds present in Phlebotomus papatasi saliva that inhibit dendritic cell (DC) functions through a PGE2/IL 10-dependent mechanism. Methodology/Principal Findings Herein, we prepared a mixture of ADO and AMP in equimolar amounts similar to those present in the salivary-gland extract (SGE) form one pair of salivary glands of P. papatasi and co-injected it with Leishmania amazonensis or L. major into mouse ears. ADO+AMP mimicked exacerbative effects of P. papatasi saliva in leishmaniasis, increasing parasite burden and cutaneous lesions. Enzymatic catabolism of salivary nucleosides reversed the SGE-induced immunosuppressive effect associated with IL-10 enhancement. Immunosuppressive factors COX2 and IL-10 were upregulated and failed to enhance ear lesion and parasite burden in IL 10-/- infected mice. Furthermore, nucleosides increased regulatory T cell (Treg) marker expression on CD4+CD25- cells, suggesting induction of Tregs on effector T cells (T eff). Treg induction (iTreg) was associated with nucleoside-induced tolerogenic dendritic cells (tDCs) expressing higher levels of COX2 and IL-10. In vitro generation of Tregs was more efficient in DCs treated with nucleosides. Suppressive effects of nucleosides during cutaneous leishmaniasis were mediated through an A2AR-dependent mechanism. Using BALB/c mice deficient in A2A ADO receptor (A2AR–/–), we showed that co-inoculated mice controlled infection, displaying lower parasite numbers at infection sites and reduced iTreg generation. Conclusion/Significance We have demonstrated that ADO and AMP in P. papatasi saliva mediate exacerbative effects of Leishmania infection by acting preferentially on DCs promoting a tolerogenic profile in DCs and by generating iTregs in inflammatory foci through an A2AR mechanism. Leishmania parasites are transmitted to their vertebrate hosts by infected Phlebotomine sand flies during the blood meal of the flies. During the Leishmania transmission, the saliva is inoculated together with parasites and exhibit several pharmacological compounds that facilitate blood feeding, interfering on homeostasis and avoiding inflammation. Thus, these compounds allow the establishment of pathogen infection. We recently identified adenosine (ADO) and adenosine monophosphate (AMP) as major immunomodulatory compounds present within the Old World sand fly species Phlebotomus papatasii, which protected mice from extreme inflammatory insults. ADO limits the magnitude of immune response by displaying a potent anti-inflammatory activity. Here, we demonstrated that ADO and AMP present in Phlebotomus papatasi saliva are involved in the establishment of parasite infection. Such nucleosides act through adenosine A2A receptor (A2AR), inducing a tolerogenic profile on dendritic cells (tDC) that may generate regulatory T cells differentiation, thus leading to suppression of the immune response and parasite survival. The identification of the active salivary constituents could serve as a strategy for the development of new vaccines to control pathogen transmission.
Collapse
Affiliation(s)
- Vanessa Carregaro
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - José M. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jesus G. Valenzuela
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Djalma L. Souza-Júnior
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlo J. F. Oliveira
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Laís A. Sacramento
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Manuela S. L. Nascimento
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristiane M. Milanezi
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q. Cunha
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - João S. Silva
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- * E-mail:
| |
Collapse
|
31
|
Lim KP, Chun NAL, Ismail SM, Abraham MT, Yusoff MN, Zain RB, Ngeow WC, Ponniah S, Cheong SC. CD4+CD25hiCD127low regulatory T cells are increased in oral squamous cell carcinoma patients. PLoS One 2014; 9:e103975. [PMID: 25153698 PMCID: PMC4143252 DOI: 10.1371/journal.pone.0103975] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022] Open
Abstract
Regulatory T cells (Tregs), a subset of CD4+ T cells plays a pivotal role in regulating the immune system. An increase in Treg numbers enables cancer progression by dampening the immune system and allowing tumor cells to evade immune detection and destruction. An increase in Treg numbers and expression of inhibitory cytokines including TGF-β and IL-10 are mechanisms by which Tregs exert their immune suppressive function. However, the presence of Tregs and inhibitory cytokines in oral cancer patients is still unclear. In this study, the presence of circulating Tregs in 39 oral cancer patients and 24 healthy donors was examined by studying the presence of the CD4+CD25hiCD127low cell population in their peripheral blood mononuclear cells using flow cytometry. Serum levels of TGF-β and IL-10 were measured by ELISA. T cell subsets of OSCC patients were found to differ significantly from healthy donors where a decrease in CD8+ cytotoxic T cells and an increase in Tregs (CD4+CD25hiCD127low) were observed. Further, the ratio of CD8+ T cells/Tregs was also decreased in patients compared to healthy donors. The presence of Tregs was accompanied by a decrease in IL-10 but not TGF-β secretion in OSCC patients when compared to donors; in addition, the analysis also revealed that an increased presence of Tregs was accompanied by better patient survival. Amongst OSCC patients, smokers had significantly higher levels of TGF-β. It is apparent that the immune system is compromised in OSCC patients and the characterization of the Treg subpopulation could form a basis for improving our understanding of the perturbations in the immune system that occur during OSCC tumorigenesis.
Collapse
Affiliation(s)
- Kue Peng Lim
- Oral Cancer Research Team, Cancer Research Initiatives Foundation (CARIF), 2 Floor Outpatient Centre, Subang Jaya, Selangor, Malaysia
| | - Nicole Ai Leng Chun
- Oral Cancer Research Team, Cancer Research Initiatives Foundation (CARIF), 2 Floor Outpatient Centre, Subang Jaya, Selangor, Malaysia
- Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Siti Mazlipah Ismail
- Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Mannil Thomas Abraham
- Department of Oral & Maxillofacial Surgery, Tengku Ampuan Rahimah Hospital, Klang, Malaysia
| | - Mohd Nury Yusoff
- Department of Oral & Maxillofacial Surgery, Tengku Ampuan Rahimah Hospital, Klang, Malaysia
| | - Rosnah Binti Zain
- Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
- Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Wei Cheong Ngeow
- Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Sathibalan Ponniah
- Cancer Vaccine Development Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Sok Ching Cheong
- Oral Cancer Research Team, Cancer Research Initiatives Foundation (CARIF), 2 Floor Outpatient Centre, Subang Jaya, Selangor, Malaysia
- Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Gu J, Lu L, Chen M, Xu L, Lan Q, Li Q, Liu Z, Chen G, Wang P, Wang X, Brand D, Olsen N, Zheng SG. TGF-β-induced CD4+Foxp3+ T cells attenuate acute graft-versus-host disease by suppressing expansion and killing of effector CD8+ cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:3388-97. [PMID: 25156367 DOI: 10.4049/jimmunol.1400207] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The use of TGF-β-induced CD4(+)Foxp3(+) T cells (induced regulatory T cells [iTregs]) is an important prevention and treatment strategy in autoimmune diseases and other disorders. However, the potential use of iTregs as a treatment modality for acute graft-versus-host disease (aGVHD) has not been realized because they may be unstable and less suppressive in this disease. We restudied the ability of iTregs to prevent and treat aGVHD in two mouse models. Our results showed that, as long as an appropriate iTreg-generation protocol is used, these iTregs consistently displayed a potent ability to control aGVHD development and reduce mortality in the aGVHD animal models. iTreg infusion markedly suppressed the engraftment of donor CD8(+) cells and CD4(+) cells, the expression of granzyme A and B, the cytotoxic effect of donor CD8(+) cells, and the production of T cell cytokines in aGVHD. Therefore, we conclude that as long as the correct methods for generating iTregs are used, they can prevent and even treat aGVHD.
Collapse
Affiliation(s)
- Jian Gu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033; Clinical Immunology Section, Third Affiliated Hospital at Sun Yat-Sen University, Guangzhou 510630, China
| | - Ling Lu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Maogen Chen
- Organ Transplant Center, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Lili Xu
- Department of Molecular Biosciences, Stockholm University, SE-10691 Stockholm, Sweden
| | - Qin Lan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; and
| | - Qiang Li
- Organ Transplant Center, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; and
| | - Guihua Chen
- Clinical Immunology Section, Third Affiliated Hospital at Sun Yat-Sen University, Guangzhou 510630, China
| | - Ping Wang
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China;
| | - Xuehao Wang
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - David Brand
- Research Service, Veterans Affairs Medical Center, Memphis, TN 38104
| | - Nancy Olsen
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Song Guo Zheng
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033; Clinical Immunology Section, Third Affiliated Hospital at Sun Yat-Sen University, Guangzhou 510630, China;
| |
Collapse
|
33
|
Rabe H, Nordström I, Andersson K, Lundell AC, Rudin A. Staphylococcus aureus convert neonatal conventional CD4(+) T cells into FOXP3(+) CD25(+) CD127(low) T cells via the PD-1/PD-L1 axis. Immunology 2014; 141:467-81. [PMID: 24708420 PMCID: PMC3930383 DOI: 10.1111/imm.12209] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/17/2013] [Accepted: 10/30/2013] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota provides an important stimulus for the induction of regulatory T (Treg) cells in mice, whether this applies to newborn children is unknown. In Swedish children, Staphylococcus aureus has become a common early colonizer of the gut. Here, we sought to study the effects of bacterial stimulation on neonatal CD4+ T cells for the induction of CD25+ CD127low Treg cells in vitro. The proportion of circulating CD25+ CD127low Treg cells and their expression of FOXP3, Helios and CTLA-4 was examined in newborns and adults. To evaluate if commensal gut bacteria could induce Treg cells, CellTrace violet-stained non-Treg cells from cord or peripheral blood from adults were co-cultured with autologous CD25+ CD127low Treg cells and remaining mononuclear cells and stimulated with S. aureus. Newborns had a significantly lower proportion of CD25+ CD127low Treg cells than adults, but these cells were Helios+ and CTLA-4+ to a higher extent than in adults. FOXP3+ CD25+ CD127low T cells were induced mainly in neonatal CellTrace-stained non-Treg cells after stimulation with S. aureus. In cell cultures from adults, S. aureus induced CD25+ CD127low T cells only if sorted naive CD45RA+ non-Treg cells were used, but these cells expressed less FOXP3 than those induced from newborns. Sorted neonatal CD25+ CD127low T cells from S. aureus-stimulated cultures were still suppressive. Finally, blocking PD-L1 during stimulation reduced the induction of FOXP3+ CD25+ CD127low T cells. These results suggest that newborns have a higher proportion of circulating thymically derived Helios+ Treg cells than adults and that S. aureus possess an ability to convert neonatal conventional CD4+ T cells into FOXP3+ CD25+ CD127low Treg cells via the PD-1/PD-L1 axis.
Collapse
Affiliation(s)
- Hardis Rabe
- Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
34
|
Abstract
Regulatory T cells are the central element for the maintenance of peripheral tolerance. Several subtypes of regulatory T (Treg) cells have been described, and most of them belong to the CD4(+) T-helper (Th) cell lineage. These specific subtypes can be discriminated according to phenotype and function. Forkhead box protein 3 (FoxP3)-expressing natural Treg cells (Tregs) and IL-10-producing, T-regulatory type 1 cells (Tr1) are the best-studied types of CD4(+) regulatory T cells in humans and experimental animal models. It was shown that they play a crucial role during autoimmune neuroinflammation. Both cells types seem to be particularly important for multiple sclerosis (MS). Here, we discuss the role of CD4(+) regulatory T cells in autoimmune neuroinflammation with an emphasis on Tregs and Tr1 cells in MS.
Collapse
Affiliation(s)
- Markus Kleinewietfeld
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Faculty of Medicine, Dresden University of Technology (TUD), Dresden, Germany
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
35
|
Recombinant IgG2a Fc (M045) multimers effectively suppress experimental autoimmune myasthenia gravis. J Autoimmun 2014; 52:64-73. [PMID: 24388113 DOI: 10.1016/j.jaut.2013.12.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/15/2013] [Indexed: 11/20/2022]
Abstract
Myasthenia gravis (MG) is an autoimmune disorder caused by target-specific pathogenic antibodies directed toward postsynaptic neuromuscular junction (NMJ) proteins, most commonly the skeletal muscle nicotinic acetylcholine receptor (AChR). In MG, high-affinity anti-AChR Abs binding to the NMJ lead to loss of functional AChRs, culminating in neuromuscular transmission failure and myasthenic symptoms. Intravenous immune globulin (IVIg) has broad therapeutic application in the treatment of a range of autoimmune diseases, including MG, although its mechanism of action is not clear. Recently, the anti-inflammatory and anti-autoimmune activities of IVIg have been attributed to the IgG Fc domains. Soluble immune aggregates bearing intact Fc fragments have been shown to be effective treatment for a number of autoimmune disorders in mice, and fully recombinant multimeric Fc molecules have been shown to be effective in treating collagen-induced arthritis, murine immune thrombocytopenic purpura, and experimental inflammatory neuritis. In this study, a murine model of MG (EAMG) was used to study the effectiveness of this novel recombinant polyvalent IgG2a Fc (M045) in treating established myasthenia, with a direct comparison to treatment with IVIg. M045 treatment had profound effects on the clinical course of EAMG, accompanied by down-modulation of pathogenic antibody responses. These effects were associated with reduced B cell activation and T cell proliferative responses to AChR, an expansion in the population of FoxP3(+) regulatory T cells, and enhanced production of suppressive cytokines, such as IL-10. Treatment was at least as effective as IVIg in suppressing EAMG, even at doses 25-30 fold lower. Multimeric Fc molecules offer the advantages of being recombinant, homogenous, available in unlimited quantity, free of risk from infection and effective at significantly reduced protein loads, and may represent a viable therapeutic alternative to polyclonal IVIg.
Collapse
|
36
|
Tang Q, Bluestone JA. Regulatory T-cell therapy in transplantation: moving to the clinic. Cold Spring Harb Perspect Med 2013; 3:3/11/a015552. [PMID: 24186492 DOI: 10.1101/cshperspect.a015552] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Regulatory T cells (Tregs) are essential to transplantation tolerance and their therapeutic efficacy is well documented in animal models. Moreover, human Tregs can be identified, isolated, and expanded in short-term ex vivo cultures so that a therapeutic product can be manufactured at relevant doses. Treg therapy is being planned at multiple transplant centers around the world. In this article, we review topics critical to effective implementation of Treg therapy in transplantation. We will address issues such as Treg dose, antigen specificity, and adjunct therapies required for transplant tolerance induction. We will summarize technical advances in Treg manufacturing and provide guidelines for identity and purity assurance of Treg products. Clinical trial designs and Treg manufacturing plans that incorporate the most up-to-date scientific understanding in Treg biology will be essential for harnessing the tolerogenic potential of Treg therapy in transplantation.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143
| | | |
Collapse
|
37
|
Natural regulatory T cells in isolated early responders compared with dual responders with allergic asthma. J Allergy Clin Immunol 2013; 133:696-703. [PMID: 24139499 DOI: 10.1016/j.jaci.2013.08.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/06/2013] [Accepted: 08/13/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Natural regulatory T (Treg) cells are implicated in the regulation of the inflammatory response in patients with allergic asthma. OBJECTIVES We sought to determine changes in Treg cell numbers in the airways and peripheral blood of isolated early responder (IER) versus dual responder (DR) subjects with mild allergic asthma before and after allergen challenge. METHODS Induced sputum was collected from 22 subjects with allergic asthma (10 IERs and 12 DRs) and peripheral blood collected from 8 DRs with allergic asthma at 0, 7, and 24 hours after allergen challenge. Treg cells were identified by using fluorescently labeled antibodies to CD4 and forkhead box protein 3 and enumerated by using flow cytometry. RESULTS There was a significant increase in the percentage of sputum CD4(+) cells 24 hours after allergen challenge in both IERs and DRs. The percentage of sputum Treg cells significantly decreased 24 hours after challenge in DRs but not IERs. This change was significantly correlated with the magnitude of the late asthmatic response. There was also a significant increase in the absolute number of sputum CD4(+) cells and Treg cells at 24 hours in DRs only. The ratio of the number of Treg cells to CD4(+) cells at 24 hours was significantly smaller in DRs compared with that in IERs. None of the above changes were observed in peripheral blood. CONCLUSIONS DRs exhibit a diminished percentage of airway Treg cells after allergen challenge that is not observed in IERs and a significantly lower ratio of Treg cells to CD4(+) cells, which might contribute to the development of the late asthmatic response.
Collapse
|
38
|
Haseda F, Imagawa A, Murase-Mishiba Y, Terasaki J, Hanafusa T. CD4⁺ CD45RA⁻ FoxP3high activated regulatory T cells are functionally impaired and related to residual insulin-secreting capacity in patients with type 1 diabetes. Clin Exp Immunol 2013; 173:207-16. [PMID: 23607886 DOI: 10.1111/cei.12116] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2013] [Indexed: 12/16/2022] Open
Abstract
Accumulating lines of evidence have suggested that regulatory T cells (T(regs)) play a central role in T cell-mediated immune response and the development of type 1A and fulminant type 1 diabetes. CD4(+) forkhead box protein 3 (FoxP3)(+) T cells are composed of three phenotypically and functionally distinct subpopulations; CD45RA(+) FoxP3(low) resting T(regs) (r-T(regs)), CD45RA(-) FoxP3(high) activated T(regs) (a-T(regs)) and CD45RA(-) FoxP3(low) non-suppressive T cells (non-T(regs)). We aimed to clarify the frequency of these three subpopulations in CD4(+) FoxP3(+) T cells and the function of a-T(regs) with reference to subtypes of type 1 diabetes. We examined 20 patients with type 1A diabetes, 15 patients with fulminant type 1 diabetes, 20 patients with type 2 diabetes and 30 healthy control subjects. A flow cytometric analysis in the peripheral blood was performed for the frequency analysis. The suppressive function of a-T(regs) was assessed by their ability to suppress the proliferation of responder cells in a 1/2:1 co-culture. A flow cytometric analysis in the peripheral blood demonstrated that the frequency of a-T(regs) was significantly higher in type 1A diabetes, but not in fulminant type 1 diabetes, than the controls. Further, the proportion of a-T(regs) among CD4(+) FoxP3(+) T cells was significantly higher in patients with type 1A diabetes with detectable C-peptide but not in patients with type 1A diabetes without it and with fulminant type 1 diabetes. A proliferation suppression assay showed that a-T(regs) were functionally impaired both in fulminant type 1 diabetes and in type 1A diabetes. In conclusion, a-T(regs) were functionally impaired, related to residual insulin-secreting capacity and may be associated with the development of type 1 diabetes.
Collapse
Affiliation(s)
- F Haseda
- Department of Internal Medicine (I), Osaka Medical College, Takatsuki, Osaka, Japan
| | | | | | | | | |
Collapse
|
39
|
Vudattu NK, Strle K, Steere AC, Drouin EE. Dysregulation of CD4+CD25(high) T cells in the synovial fluid of patients with antibiotic-refractory Lyme arthritis. ACTA ACUST UNITED AC 2013; 65:1643-53. [PMID: 23450683 DOI: 10.1002/art.37910] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/14/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the role of immune dysregulation in antibiotic-refractory Lyme arthritis by comparing the phenotype, frequency, and function of CD4+ Teff cells and Treg cells in patients with antibiotic- responsive arthritis and patients with antibiotic-refractory arthritis. METHODS Matched peripheral blood and synovial fluid samples from 15 patients with antibiotic-responsive arthritis were compared with those from 16 patients with antibiotic-refractory arthritis, using flow cytometry, suppression assays, and cytokine assays. RESULTS Critical differences between the 2 patient groups were observed in the synovial fluid CD4+CD25(high) population, a cell subset usually composed of FoxP3-positive Treg cells. In patients with antibiotic-refractory arthritis, this cell population often had fewer FoxP3-positive cells and a greater frequency of FoxP3-negative (Teff) cells compared with patients with antibiotic-responsive arthritis. Moreover, the expression of glucocorticoid-induced tumor necrosis factor receptor and OX40 on CD4+CD25(high) cells was significantly higher in the antibody-refractory group. Suppression assays showed that CD4+CD25(high) cells in patients with antibiotic-refractory arthritis did not effectively suppress proliferation of CD4+CD25- cells or secretion of interferon-γ and tumor necrosis factor α, whereas those cells in patients with antibiotic-responsive arthritis did suppress proliferation of CD4+CD25- cells and secretion of interferon-γ and tumor necrosis factor α. Finally, in the antibiotic-refractory group, higher ratios of CD25(high) FoxP3-negative cells to CD25(high) FoxP3-positive cells correlated directly with a longer duration of arthritis after antibiotic treatment. CONCLUSION Patients with antibiotic-refractory Lyme arthritis often have lower frequencies of Treg cells, higher expression of activation coreceptors, and less effective inhibition of proinflammatory cytokines. This suggests that immune responses in these patients are excessively amplified, leading to immune dysregulation and antibiotic-refractory arthritis.
Collapse
Affiliation(s)
- Nalini K Vudattu
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
40
|
Gagliani N, Jofra T, Valle A, Stabilini A, Morsiani C, Gregori S, Deng S, Rothstein DM, Atkinson M, Kamanaka M, Flavell RA, Roncarolo MG, Battaglia M. Transplant tolerance to pancreatic islets is initiated in the graft and sustained in the spleen. Am J Transplant 2013; 13:1963-75. [PMID: 23834659 PMCID: PMC3869180 DOI: 10.1111/ajt.12333] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 01/25/2023]
Abstract
The immune system is comprised of several CD4(+) T regulatory (Treg) cell types, of which two, the Foxp3(+) Treg and T regulatory type 1 (Tr1) cells, have frequently been associated with transplant tolerance. However, whether and how these two Treg-cell types synergize to promote allograft tolerance remains unknown. We previously developed a mouse model of allogeneic transplantation in which a specific immunomodulatory treatment leads to transplant tolerance through both Foxp3(+) Treg and Tr1 cells. Here, we show that Foxp3(+) Treg cells exert their regulatory function within the allograft and initiate engraftment locally and in a non-antigen (Ag) specific manner. Whereas CD4(+) CD25(-) T cells, which contain Tr1 cells, act from the spleen and are key to the maintenance of long-term tolerance. Importantly, the role of Foxp3(+) Treg and Tr1 cells is not redundant once they are simultaneously expanded/induced in the same host. Moreover, our data show that long-term tolerance induced by Foxp3(+) Treg-cell transfer is sustained by splenic Tr1 cells and functionally moves from the allograft to the spleen.
Collapse
Affiliation(s)
- N. Gagliani
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy,San Raffaele Telethon Institute for Gene Therapy, Milan, Italy,Vita-Salute San Raffaele University, Milan, Italy
| | - T. Jofra
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy
| | - A. Valle
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy
| | - A. Stabilini
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy
| | - C. Morsiani
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy
| | - S. Gregori
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | - S. Deng
- Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - D. M. Rothstein
- Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - M. Atkinson
- Department of Pathology, The University of Florida, Gainesville, FL
| | - M. Kamanaka
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - R. A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Howard Hughes Medical Institute, New Haven, CT
| | - M. G. Roncarolo
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy,Vita-Salute San Raffaele University, Milan, Italy
| | - M. Battaglia
- San Raffaele Scientific Institute, Diabetes Research Institute, Milan, Italy,Corresponding author: Manuela Battaglia,
| |
Collapse
|
41
|
Helicobacter pylori Infection: Regulatory T Cells and Their Participation in the Immune Response. Jundishapur J Microbiol 2013. [DOI: 10.5812/jjm.5183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
42
|
Wang LK, Kuang M, Hua YP, He Q, Chen B, Wang Y, Peng BG. Profile of regulatory T cells and interferon γ secretion in the tumor-draining lymph node from mouse Hepa1-6 cells. J Surg Res 2013; 183:900-6. [PMID: 23481561 DOI: 10.1016/j.jss.2013.02.001] [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: 11/09/2012] [Revised: 01/29/2013] [Accepted: 02/01/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND The tumor-draining lymph node (TDLN) is the critical and initial site of the immune decision made between activation and tolerance to tumor antigens. Tumor-reactive lymphadenopathy in TDLN has been observed for decades, but the profiles of immune regulation in these nodes remain unclear. MATERIALS AND METHODS Both regulatory T cells (Tregs) and effector T cells were examined using 6 × 10(5) Hepa1-6 hepatocellular carcinoma cells implanted in footpads of syngeneic C57BL/6J mice, which formed TDLN. FOXP3(+) Tregs and CD8(+) T cells in TDLN were detected by immunohistochemical staining. The frequency of CD4(+)FOXP3(+) T cells and FOXP3 mRNA expression were determined by flow cytometry and real-time quantitative polymerase chain reaction. The interferon γ secretion ability of CD8(+) T cells in TDLN was measured by enzyme-linked immunospot technique. RESULTS There was significant expansion of Tregs and CD8(+) T cells in the tumor-draining popliteal lymph node compared with nondraining popliteal lymph node and spleen in the same mouse. Tregs were diffusely distributed in the CD8(+) T cell compartment. The CD8(+) T cells primed in TDLN showed a strong ability of interferon γ secretion via in vitro stimulation. CONCLUSIONS These findings support the notion that Tregs suppress CD8(+) T cells by secreting cytokines such as transforming growth factor β and interleukin 10, but do not make CD8(+) T cells lose function in the TDLN. Deletion of Tregs at the secondary lymphoid organs could be crucial for the establishment of a tumor-specific immunotherapy.
Collapse
Affiliation(s)
- Li-Kun Wang
- Department of General Surgery, the Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Maazi H, Shirinbak S, Willart M, Hammad HM, Cabanski M, Boon L, Ganesh V, Baru AM, Hansen G, Lambrecht BN, Sparwasser T, Nawijn MC, van Oosterhout AJM. Contribution of regulatory T cells to alleviation of experimental allergic asthma after specific immunotherapy. Clin Exp Allergy 2013; 42:1519-28. [PMID: 22994349 DOI: 10.1111/j.1365-2222.2012.04064.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (SIT) has been used since 1911, yet its mechanism of action remains to be elucidated. There is evidence indicating that CD4(+)FOXP3(+) regulatory T cells (Treg cells) are induced during SIT in allergic patients. However, the contribution of these cells to SIT has not been evaluated in vivo. OBJECTIVE To evaluate the in vivo contribution of (i) CD4(+) CD25(+) T cells during SIT and of (ii) SIT-generated inducible FOXP3(+) Treg cells during allergen exposure to SIT-mediated suppression of asthmatic manifestations. METHODS We used a mouse model of SIT based on the classical OVA-driven experimental asthma. Treg cells were quantified by flow cytometry 24 and 96 h post SIT treatment. We depleted CD4(+) CD25(+) T cells prior to SIT, and CD4(+)FOXP3(+) T cells prior to allergen challenges to study their contribution to the suppression of allergic manifestations by SIT treatment. RESULTS Our data show that depletion of CD4(+)CD25(+) T cells at the time of SIT treatment reverses the suppression of airway hyperresponsiveness (AHR), but not of airway eosinophilia and specific IgE levels in serum. Interestingly, the number of CD4(+)CD25(+)FOXP3(+) T cells is transiently increased after SIT in the spleen and blood, suggesting the generation of inducible and presumably allergen-specific Treg cells during treatment. Depletion of CD4(+)FOXP3(+) Treg cells after SIT treatment partially reverses the SIT-induced suppression of airway eosinophilia, but not of AHR and serum levels of specific IgE. CONCLUSION AND CLINICAL RELEVANCE We conclude that SIT-mediated tolerance induction towards AHR requires CD4(+)CD25(+) T cells at the time of allergen injections. In addition, SIT generates CD4(+)CD25(+)FOXP3(+) T cells that contribute to the suppression of airway eosinophilia upon allergen challenges. Therefore, enhancing Treg cell number or their activity during and after SIT could be of clinical relevance to improve the therapeutic effects of SIT.
Collapse
Affiliation(s)
- H Maazi
- Laboratory of Allergology & Pulmonary Diseases, Department of Pathology & Medical Biology, GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Rabinowitz KM, Wang Y, Chen EY, Hovhannisyan Z, Chiang D, Berin MC, Dahan S, Chaussabel D, Ma'ayan A, Mayer L. Transforming growth factor β signaling controls activities of human intestinal CD8(+)T suppressor cells. Gastroenterology 2013; 144:601-612.e1. [PMID: 23232296 PMCID: PMC3967796 DOI: 10.1053/j.gastro.2012.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 11/08/2012] [Accepted: 12/05/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS In healthy individuals, interactions between intestinal epithelial cells and lamina propria lymphocytes give rise to a population of CD8(+) T cells with suppressor functions (Ts cells). Disruption of Ts cell activities can lead to mucosal inflammation. We investigated what factors were required for expansion of the Ts cell population or loss of their activity in patients with Crohn's disease (CD). METHODS We developed a method to generate Ts cell lines from freshly isolated lamina propria lymphocytes from patients with ulcerative colitis (UC), patients with CD, or healthy individuals (controls). Cells were stimulated with a monoclonal antibody against CD3, interleukin (IL)-7, and IL-15. After 14 days in culture, CD8(+)T cells were purified and cultured with IL-7 and IL-15. The resulting Ts cells were analyzed for suppressor activity, expression of surface markers, and cytokine secretion profiles. RNA was isolated from the 3 groups of Ts cells and used in microarray analyses. RESULTS Ts cells from patients with UC and controls suppressed proliferation of CD4(+) T cells; the suppression required cell contact. In contrast, Ts cells from patients with CD had a reduced capacity to suppress CD4(+) T-cell proliferation. The difference in suppressive ability was not associated with surface or intracytoplasmic markers or secretion of cytokines. Microarray analysis identified changes in expression of genes regulated by transforming growth factor (TGF)-β that were associated with the suppressive abilities of Ts cells. We found that TGF-β or supernatants from Ts cells of patients with CD reduced the suppressor activity of control Ts cells. CONCLUSIONS Ts cells isolated from patients with CD have a reduced ability to suppress proliferation of CD4(+)T cells compared with control Ts cells. TGF-β signaling reduces the suppressor activity of Ts cells.
Collapse
Affiliation(s)
- Keren M Rabinowitz
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dunn GP, Fecci PE, Curry WT. Cancer immunoediting in malignant glioma. Neurosurgery 2013; 71:201-22; discussion 222-3. [PMID: 22353795 DOI: 10.1227/neu.0b013e31824f840d] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Significant work from many laboratories over the last decade in the study of cancer immunology has resulted in the development of the cancer immunoediting hypothesis. This contemporary framework of the naturally arising immune system-tumor interaction is thought to comprise 3 phases: elimination, wherein immunity subserves an extrinsic tumor suppressor function and destroys nascent tumor cells; equilibrium, wherein tumor cells are constrained in a period of latency under immune control; and escape, wherein tumor cells outpace immunity and progress clinically. In this review, we address in detail the relevance of the cancer immunoediting concept to neurosurgeons and neuro-oncologists treating and studying malignant glioma by exploring the de novo immune response to these tumors, how these tumors may persist in vivo, the mechanisms by which these cells may escape/attenuate immunity, and ultimately how this concept may influence our immunotherapeutic approaches.
Collapse
Affiliation(s)
- Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
46
|
Ratio of T Helper to Regulatory T Cells in Synovial Fluid and Postoperative Joint Laxity After Allograft Anterior Cruciate Ligament Reconstruction. Transplantation 2012; 94:1160-6. [DOI: 10.1097/tp.0b013e31826dddeb] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Conditioned medium from adipose tissue-derived mesenchymal stem cells induces CD4+FOXP3+ cells and increases IL-10 secretion. J Biomed Biotechnol 2012; 2012:295167. [PMID: 23251077 PMCID: PMC3521492 DOI: 10.1155/2012/295167] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 10/05/2012] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a new and promising tool for therapy of autoimmune disorders. In recent years their possibility to take part in the modulation of the immune response is discussed. The exact mechanisms for immunoregulation realized by MSCs are not clear yet, but interactions with other immunoregulatory cells may be involved in this process. The investigation of the influence of MSCs on the expression of FoxP3 and cytokine secretion by T helper cells was the aim of this study. T helper cells were isolated from PBMCs by magnetic separation and MSCs were isolated from human adipose tissue, and CD4+ T cells were cultured with conditional medium of MSCs. The methods which were used include flow cytometry, ELISA, and Human Proteome profiler kits. The results demonstrated that secretory factors in MSCs conditional medium lead to increased expression of FoxP3 and increased secretion of IL-10 by T helpers. The obtained results give us opportunity to discuss the interaction between two kinds of immunoregulatory cells: MSCs and FoxP3+ T helpers. We suppose that this interaction leads to increased number of immunosuppressive helpers which secrete IL-10. MSCs provide some of their immunosuppressive functions acting on T regulatory cells, and we believe that IL-6 secreted by MSCs is involved in this process.
Collapse
|
48
|
Abstract
Hepatocellular carcinoma (HCC) has been shown to induce several immune suppressor mechanisms in patients. Our laboratory has been investigating different cellular mechanisms of immune suppression in patients with HCC. These suppressor mechanisms range from CD4(+) regulatory T cells, functionally impaired dendritic cells, neutrophils, and monocytes to myeloid-derived suppressor cells. In vitro as well as in vivo studies have demonstrated that abrogation of the suppressor cells enhances or unmasks tumor-specific anti-tumor immune responses. We performed a literature search for immune suppressor cells in HCC, and here we provide a comprehensive summary of the latest studies in this field.
Collapse
Affiliation(s)
- Fei Zhao
- Gastrointestinal Malignancy Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | |
Collapse
|
49
|
The role of different subsets of regulatory T cells in immunopathogenesis of rheumatoid arthritis. ARTHRITIS 2012; 2012:805875. [PMID: 23133752 PMCID: PMC3486158 DOI: 10.1155/2012/805875] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 09/11/2012] [Accepted: 09/20/2012] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease and a systemic inflammatory disease which is characterized by chronic joint inflammation and variable degrees of bone and cartilage erosion and hyperplasia of synovial tissues. Considering the role of autoreactive T cells (particularly Th1 and Th17 cells) in pathophysiology of RA, it might be assumed that the regulatory T cells (Tregs) will be able to control the initiation and progression of disease. The frequency, function, and properties of various subsets of Tregs including natural Tregs (nTregs), IL-10-producing type 1 Tregs (Tr1 cells), TGF-β-producing Th3 cells, CD8+ Tregs, and NKT regulatory cells have been investigated in various studies associated with RA and collagen-induced arthritis (CIA) as experimental model of this disease. In this paper, we intend to submit the comprehensive information about the immunobiology of various subsets of Tregs and their roles and function in immunopathophysiology of RA and its animal model, CIA.
Collapse
|
50
|
Höfer T, Krichevsky O, Altan-Bonnet G. Competition for IL-2 between Regulatory and Effector T Cells to Chisel Immune Responses. Front Immunol 2012; 3:268. [PMID: 22973270 PMCID: PMC3433682 DOI: 10.3389/fimmu.2012.00268] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/05/2012] [Indexed: 01/24/2023] Open
Abstract
In this review we discuss how the competition for cytokines between different cells of the immune system can shape the system wide immune response. We focus on interleukin-2 (IL-2) secretion by activated effector T cells (T(eff)) and on the competition for IL-2 consumption between T(eff) and regulatory T cells (T(reg)). We discuss the evidence for the mechanism in which the depletion of IL-2 by T(reg) cells would be sufficient to suppress an autoimmune response, yet not strong enough to prevent an immune response. We present quantitative estimations and summarize our modeling effort to show that the tug-of-war between T(reg) and T(eff) cells for IL-2 molecules can be won by T(reg) cells in the case of weak activation of T(eff) leading to the suppression of the immune response. Or, for strongly activated T(eff) cells, it can be won by T(eff) cells bringing about the activation of the whole adaptive immune system. Finally, we discuss some recent applications attempting to achieve clinical effects through the modulation of IL-2 consumption by T(reg) compartment.
Collapse
Affiliation(s)
- Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center Heidelberg, Germany
| | | | | |
Collapse
|