1
|
Liu XF, Song B, Sun CB, Zhu Q, Yue JH, Liang YJ, He J, Zeng XL, Qin YC, Chen QY, Mai HQ, Zhang X, Li J. Tumor-infiltrated double-negative regulatory T cells predict outcome of T cell-based immunotherapy in nasopharyngeal carcinoma. Cell Rep Med 2025; 6:102096. [PMID: 40315843 DOI: 10.1016/j.xcrm.2025.102096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/24/2025] [Accepted: 04/04/2025] [Indexed: 05/04/2025]
Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) has demonstrated clinical success in solid tumors. We analyze 47 TIL infusion products and 62 pretreatment tumor microenvironments (TMEs) from a randomized phase 2 clinical study of concurrent chemoradiotherapy plus TIL-ACT (NCT02421640). Using single-cell and bulk RNA sequencing along with flow cytometry, we identify 14 CD3+ T cell clusters within 26 TIL infusion products: 11 CD3+CD8+ TILs, 2 CD3+CD4+ TILs, and 1 CD3+CD8-CD4- double-negative (DN) TIL. (DN) TILs, significantly associated with poor TIL-ACT outcomes, exhibit an activated regulatory T cell-like phenotype and include two CD56+ and four CD56- subsets. Among them, CD56-KZF2+ (DN) TILs are predominantly suppressive. (DN) TILs inhibit CD8+ TIL expansion via Fas-FasL, transforming growth factor β (TGF-β), and interleukin (IL)-10 signaling. Distinct CD8+ T subsets differentially impact on TIL-ACT outcomes, while 9 baseline TME gene signatures and 14 intracellular T cell genes hold prognostic value. Our findings identify predictive TIL subsets and biomarkers for TIL-ACT outcomes.
Collapse
Affiliation(s)
- Xiu-Feng Liu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Bin Song
- BGI, Shenzhen 518083, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Chang-Bin Sun
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, P.R. China
| | - Qian Zhu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Yu-Jing Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jia He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xi-Liang Zeng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Xi Zhang
- BGI, Shenzhen 518083, P.R. China.
| | - Jiang Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
2
|
Yu J, Yang Y, Gu Z, Shi M, La Cava A, Liu A. CAR immunotherapy in autoimmune diseases: promises and challenges. Front Immunol 2024; 15:1461102. [PMID: 39411714 PMCID: PMC11473342 DOI: 10.3389/fimmu.2024.1461102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
In recent years, the use of chimeric antigen receptor (CAR)-T cells has emerged as a promising immunotherapy in multiple diseases. CAR-T cells are T cells genetically modified to express a surface receptor, known as CAR, for the targeting of cognate antigens on specific cells. The effectiveness of CAR-T cell therapy in hematologic malignancies including leukemia, myeloma, and non-Hodgkin's lymphoma has led to consider its use as a potential avenue of treatment for autoimmune diseases. However, broadening the use of CAR-T cell therapy to a large spectrum of autoimmune conditions is challenging particularly because of the possible development of side effects including cytokine release syndrome and neurotoxicity. The design of CAR therapy that include additional immune cells such as double-negative T cells, γδ T cells, T regulatory cells and natural killer cells has shown promising results in preclinical studies and clinical trials in oncology, suggesting a similar potential utility in the treatment of autoimmune diseases. This review examines the mechanisms, efficacy, and safety of CAR approaches with a focus on their use in autoimmune diseases including systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, multiple sclerosis, myasthenia gravis, lupus nephritis and other autoimmune diseases. Advantages and disadvantages as compared to CAR-T cell therapy will also be discussed.
Collapse
Affiliation(s)
- Jingjing Yu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiming Yang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhanjing Gu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Shi
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Antonio La Cava
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Department of Medicina Molecolare e Biotecnologie Mediche, Federico II University, Naples, Italy
| | - Aijing Liu
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Chen X, Hu G, Ning D, Wang D. Exploring gut microbiota's role in rheumatic valve disease: insights from a Mendelian randomization study and mediation analysis. Front Immunol 2024; 15:1362753. [PMID: 38895120 PMCID: PMC11183100 DOI: 10.3389/fimmu.2024.1362753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Background Investigating the relationship between gut microbiota and Rheumatic Valve Disease (RVD) is crucial for understanding the disease's etiology and developing effective interventions. Our study adopts a novel approach to examine the potential causal connections between these factors. Methods Utilizing a two-sample Mendelian Randomization (MR) framework, we incorporated a multi-variable MR (MVMR) strategy to assess the mediatory mechanisms involved. This approach involved analyzing data from the MiBioGen consortium for gut microbiota and the FinnGen for RVD, among other sources. Instrumental variables (IVs) were carefully selected based on rigorous MR principles, and statistical analysis was conducted using bidirectional two-sample MR, such as inverse variance-weighted (IVW), weighted median, MR-Egger regression and MR Steiger Test methods. The MR-PRESSO strategy was employed for outlier detection, and MVMR was used to untangle the complex relationships between multiple microbiota and RVD. Results Our analysis highlighted several gut microbiota classes and families with potential protective effects against RVD, including Lentisphaerae, Alphaproteobacteria, and Streptococcaceae. In contrast, certain genera, such as Eubacterium eligens and Odoribacter, were identified as potential risk factors. The MVMR analysis revealed significant mediation effects of various immune cell traits and biomarkers, such as CD4-CD8- T cells, CD3 on Terminally Differentiated CD8+ T cell and Pentraxin-related protein PTX, elucidating the complex pathways linking gut microbiota to RVD. Conclusion This study underscores the intricate and potentially causal relationship between gut microbiota and RVD, mediated through a range of immune and hormonal factors. The use of MVMR in our methodological approach provides a more comprehensive understanding of these interactions, highlighting the gut microbiota's potential as therapeutic targets in RVD management. Our findings pave the way for further research to explore these complex relationships and develop targeted interventions for RVD.
Collapse
Affiliation(s)
- Xiwei Chen
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Guangwen Hu
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Dong Ning
- Department of Physiology, Human Biology Building, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | - Daxin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| |
Collapse
|
4
|
Xiao X, Liu H, Qiu X, Chen P, Li X, Wang D, Song G, Cheng Y, Yang L, Qian W. CD19-CAR-DNT cells (RJMty19) in patients with relapsed or refractory large B-cell lymphoma: a phase 1, first-in-human study. EClinicalMedicine 2024; 70:102516. [PMID: 38444429 PMCID: PMC10912040 DOI: 10.1016/j.eclinm.2024.102516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/04/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Background Current approved chimeric antigen receptor (CAR) T-cell products are autologous cell therapies that are costly and poorly accessible to patients. We aimed to evaluate the safety and antitumor activity of a novel off-the-shelf anti-CD19 CAR-engineered allogeneic double-negative T cells (RJMty19) in patients with relapsed/refractory large B-cell lymphoma. We report the results from a first-in-human, open-label, single-dose, phase 1 study of allogeneic CD19-specific CAR double-negative T (CAR-DNT) cells. Methods Eligibility criteria included the presence of measurable lesions, at least 2 lines of prior immunochemotherapy, and an ECOG score of 0-1. We evaluated four dose levels (DL) of RJMty19 in a 3 + 3 dose-escalation scheme: 1 × 106, 3 × 106, 9 × 106 and 2 × 107 CAR-DNT cells per kilogram of body weight. All patients received lymphodepleting chemotherapy with fludarabine and cyclophosphamide. The primary endpoints were dose-limiting toxicities (DLTs), incidence of adverse events (AEs), and clinically significant laboratory abnormalities. Secondary endpoints included evaluation of standard cellular pharmacokinetic parameters, immunogenicity, objective response rates (ORR), and disease control rate (DCR) per Lugano 2014 criteria. Findings A total of 12 patients were enrolled between 22 July 2022 and 27 July 2023. Among these patients, 66% were classified as stage IV, 75% had an IPI score of 3 or higher, representing an intermediate risk or worse. The maximum tolerated dose was not reached because no DLT was observed. Four patient experienced grade 1 or 2 cytokine release syndrome and dizziness. The most common AEs were hematologic toxicities, including neutropenia (N = 12, 100%), leukopenia (N = 12, 100%), lymphopenia (N = 10, 83%), thrombocytopenia (N = 6, 50%), febrile neutropenia (N = 3, 25%), and anemia (N = 3, 25%). Seven subjects died till the cut-off date, five of them died of disease progression and two of them died of COVID 19. In all patients (N = 12), the ORR was 25% and CRR was 8.3%. DL1 and DL2 patients benefited less from the therapy (ORR: 17%, N = 1; DCR: 33%, N = 2). However, all DL3 patients achieved disease control (N = 3, 100%), and all DL4 patients achieved objective response (N = 3, 100%). Interpretation Our results demonstrate that CD19-CAR-DNT cells appear to be well tolerated with promising antitumor activity in LBCL patients. Further study of this product with a larger sample size is warranted. This phase 1 study is registered on clinicaltrials.gov (NCT05453669). Funding Wyze Biotech. Co., Ltd.
Collapse
Affiliation(s)
- Xibin Xiao
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Liu
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xi Qiu
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Panpan Chen
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xian Li
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dan Wang
- Wyze Biotech Co., Ltd, Zhongshan, Guangdong, China
| | | | - Yu Cheng
- Wyze Biotech Co., Ltd, Zhongshan, Guangdong, China
| | - Liming Yang
- Wyze Biotech Co., Ltd, Zhongshan, Guangdong, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Briceño O, Peralta-Prado A, Garrido-Rodríguez D, Romero-Mora K, Chávez-Torres M, Pinto Cardoso S, Alvarado de la Barrera C, Reyes-Terán G, Ávila-Ríos S. Double-Negative T Cell Number and Phenotype Alterations Before and After Effective Antiretroviral Treatment in Persons Living with HIV. AIDS Res Hum Retroviruses 2023; 39:104-113. [PMID: 36511386 DOI: 10.1089/aid.2022.0047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Double-negative (DN) T cells represent a small and phenotypically heterogeneous population that display regulatory functions. In HIV infection, DN T cells are decreased in peripheral blood and have been negatively associated with T cell activation. This study was aimed at describing the dynamics and phenotypic characteristics of DN T cells in peripheral blood of people living with HIV (PLHIV) before and after antiretroviral therapy (ART) initiation. We included 41 newly diagnosed, ART-naive individuals with advanced HIV infection, who were followed up for 6 months after ART initiation. The control group included 34 people without HIV (PWHIV), on preexposure prophylaxis for HIV infection. DN T cells in peripheral blood were characterized by flow cytometry. The absolute counts of DN T cells were lower in PLHIV than in PWHIV (p = 0.0223), and were particularly low in individuals with advanced HIV disease (p = 0.0311). Activation of DN T cells before ART initiation was directly associated with viral load (VL) (p = 0.0081, r = 0.4083) and inversely associated with CD4+ T cell counts (p = 0.0004, r = -0.4041). Compared with PWHIV, DN T cells of PLHIV expressed higher levels of CD57 (p = 0.0019), Ki67 (p = 0.0065), PD-1 (p = 0.0187), and CD38/HLA-DR (p < 0.0001). After 6 months on ART, expression of Ki67, PD-1, and CD38/HLA-DR on DN T cells returned to similar levels to those observed in PWHIV (p > 0.05 in all cases). However, expression of CD57 decreased only in individuals that start ART with high VL (p = 0.0127). DN T cell counts are decreased in HIV infection. Low DN T cell counts remained despite ART-induced immune reconstitution and viremia control. DN T cell phenotype is altered during chronic untreated infection with a high proportion of proliferating, activated, exhausted, and senescent cells. Most markers return to levels similar to those observed in PWHIV after ART. The impact of altered phenotype of DN T and their regulatory functions warrants further exploration.
Collapse
Affiliation(s)
- Olivia Briceño
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Amy Peralta-Prado
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Daniela Garrido-Rodríguez
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Karla Romero-Mora
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Monserrat Chávez-Torres
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Sandra Pinto Cardoso
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Claudia Alvarado de la Barrera
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Gustavo Reyes-Terán
- Institutos Nacionales de Salud y Hospitales de Alta Especialidad, Secretaría de Salud de México, Ciudad de Mexico, Mexico
| | - Santiago Ávila-Ríos
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| |
Collapse
|
6
|
Bafor EE, Valencia JC, Young HA. Double Negative T Regulatory Cells: An Emerging Paradigm Shift in Reproductive Immune Tolerance? Front Immunol 2022; 13:886645. [PMID: 35844500 PMCID: PMC9283768 DOI: 10.3389/fimmu.2022.886645] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Immune regulation of female reproductive function plays a crucial role in fertility, as alterations in the relationship between immune and reproductive processes result in autoimmune subfertility or infertility. The breakdown of immune tolerance leads to ovulation dysfunction, implantation failure, and pregnancy loss. In this regard, immune cells with regulatory activities are essential to restore self-tolerance. Apart from regulatory T cells, double negative T regulatory cells (DNTregs) characterized by TCRαβ+/γδ+CD3+CD4–CD8– (and negative for natural killer cell markers) are emerging as effector cells capable of mediating immune tolerance in the female reproductive system. DNTregs are present in the female reproductive tract of humans and murine models. However, their full potential as immune regulators is evolving, and studies so far indicate that DNTregs exhibit features that can also maintain tolerance in the female reproductive microenvironment. This review describes recent progress on the presence, role and mechanisms of DNTregs in the female reproductive system immune regulation and tolerance. In addition, we address how DNTregs can potentially provide a paradigm shift from the known roles of conventional regulatory T cells and immune tolerance by maintaining and restoring balance in the reproductive microenvironment of female fertility.
Collapse
Affiliation(s)
- Enitome E Bafor
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Julio C Valencia
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Howard A Young
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
7
|
Newman-Rivera AM, Kurzhagen JT, Rabb H. TCRαβ+ CD4-/CD8- "double negative" T cells in health and disease-implications for the kidney. Kidney Int 2022; 102:25-37. [PMID: 35413379 PMCID: PMC9233047 DOI: 10.1016/j.kint.2022.02.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 12/22/2022]
Abstract
Double negative (DN) T cells, one of the least studied T lymphocyte subgroups, express T cell receptor αβ but lack CD4 and CD8 coreceptors. DN T cells are found in multiple organs including kidney, lung, heart, gastrointestinal tract, liver, genital tract, and central nervous system. DN T cells suppress inflammatory responses in different disease models including experimental acute kidney injury, and significant evidence supports an important role in the pathogenesis of systemic lupus erythematosus. However, little is known about these cells in other kidney diseases. Therefore, it is important to better understand different functions of DN T cells and their signaling pathways as promising therapeutic targets, particularly with the increasing application of T cell-directed therapy in humans. In this review, we aim to summarize studies performed on DN T cells in normal and diseased organs in the setting of different disease models with a focus on kidney.
Collapse
Affiliation(s)
| | | | - Hamid Rabb
- Nephrology Division, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
8
|
Vasic D, Lee JB, Leung Y, Khatri I, Na Y, Abate-Daga D, Zhang L. Allogeneic double-negative CAR-T cells inhibit tumor growth without off-tumor toxicities. Sci Immunol 2022; 7:eabl3642. [PMID: 35452255 DOI: 10.1126/sciimmunol.abl3642] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of autologous chimeric antigen receptor T (CAR-T) cell therapies has revolutionized cancer treatment. Nevertheless, the delivery of CAR-T cell therapy faces challenges, including high costs, lengthy production times, and manufacturing failures. To overcome this, attempts have been made to develop allogeneic CAR-T cells using donor-derived conventional CD4+ or CD8+ T cells (Tconvs), but severe graft-versus-host disease (GvHD) and host immune rejection have made this challenging. CD3+CD4-CD8- double-negative T cells (DNTs) are a rare subset of mature T cells shown to fulfill the requirements of an off-the-shelf cellular therapy, including scalability, cryopreservability, donor-independent anticancer function, resistance to rejection, and no observed off-tumor toxicity including GvHD. To overcome the challenges faced with CAR-Tconvs, we evaluated the feasibility, safety, and efficacy of using healthy donor-derived allogeneic DNTs as a CAR-T cell therapy platform. We successfully transduced DNTs with a second-generation anti-CD19-CAR (CAR19) without hampering their endogenous characteristics or off-the-shelf properties. CAR19-DNTs induced antigen-specific cytotoxicity against B cell acute lymphoblastic leukemia (B-ALL). In addition, CAR19-DNTs showed effective infiltration and tumor control against lung cancer genetically modified to express CD19 in xenograft models. CAR19-DNT efficacy was comparable with that of CAR19-Tconvs. However, unlike CAR19-Tconvs, CAR19-DNTs did not cause alloreactivity or xenogeneic GvHD-related mortality in xenograft models. These studies demonstrate the potential of using allogeneic DNTs as a platform for CAR technology to provide a safe, effective, and patient-accessible CAR-T cell treatment option.
Collapse
Affiliation(s)
- Daniel Vasic
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jong Bok Lee
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yuki Leung
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ismat Khatri
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yoosu Na
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Daniel Abate-Daga
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Wu Z, Zheng Y, Sheng J, Han Y, Yang Y, Pan H, Yao J. CD3 +CD4 -CD8 - (Double-Negative) T Cells in Inflammation, Immune Disorders and Cancer. Front Immunol 2022; 13:816005. [PMID: 35222392 PMCID: PMC8866817 DOI: 10.3389/fimmu.2022.816005] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/21/2022] [Indexed: 12/28/2022] Open
Abstract
The crucial role of CD4+ and CD8+ T cells in shaping and controlling immune responses during immune disease and cancer development has been well established and used to achieve marked clinical benefits. CD3+CD4-CD8- double-negative (DN) T cells, although constituting a rare subset of peripheral T cells, are gaining interest for their roles in inflammation, immune disease and cancer. Herein, we comprehensively review the origin, distribution and functions of this unique T cell subgroup. First, we focused on characterizing multifunctional DN T cells in various immune responses. DN regulatory T cells have the capacity to prevent graft-versus-host disease and have therapeutic value for autoimmune disease. T helper-like DN T cells protect against or promote inflammation and virus infection depending on the specific settings and promote certain autoimmune disease. Notably, we clarified the role of DN tumor-infiltrating lymphocytes and outlined the potential for malignant proliferation of DN T cells. Finally, we reviewed the recent advances in the applications of DN T cell-based therapy for cancer. In conclusion, a better understanding of the heterogeneity and functions of DN T cells may help to develop DN T cells as a potential therapeutic tool for inflammation, immune disorders and cancer.
Collapse
Affiliation(s)
- Zhiheng Wu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jin Sheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yicheng Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Yang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Junlin Yao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Nakayama M, Hori A, Toyoura S, Yamaguchi SI. Shaping of T Cell Functions by Trogocytosis. Cells 2021; 10:cells10051155. [PMID: 34068819 PMCID: PMC8151334 DOI: 10.3390/cells10051155] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Trogocytosis is an active process whereby plasma membrane proteins are transferred from one cell to the other cell in a cell-cell contact-dependent manner. Since the discovery of the intercellular transfer of major histocompatibility complex (MHC) molecules in the 1970s, trogocytosis of MHC molecules between various immune cells has been frequently observed. For instance, antigen-presenting cells (APCs) acquire MHC class I (MHCI) from allografts, tumors, and virally infected cells, and these APCs are subsequently able to prime CD8+ T cells without antigen processing via the preformed antigen-MHCI complexes, in a process called cross-dressing. T cells also acquire MHC molecules from APCs or other target cells via the immunological synapse formed at the cell-cell contact area, and this phenomenon impacts T cell activation. Compared with naïve and effector T cells, T regulatory cells have increased trogocytosis activity in order to remove MHC class II and costimulatory molecules from APCs, resulting in the induction of tolerance. Accumulating evidence suggests that trogocytosis shapes T cell functions in cancer, transplantation, and during microbial infections. In this review, we focus on T cell trogocytosis and the related inflammatory diseases.
Collapse
|
11
|
Shen CF, Ho TS, Wang SM, Liao YT, Hu YS, Tsai HP, Chen SH. The cellular immunophenotype expression of influenza A virus and influenza B virus infection in children. Clin Immunol 2020; 219:108548. [PMID: 32735869 DOI: 10.1016/j.clim.2020.108548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/13/2020] [Accepted: 07/24/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The innate immune response is the primary defense against influenza virus infection. METHODS This is a prospective study carried out in children <18 years of age who were diagnosed with influenza A or influenza B infection. Demographic and clinical data, laboratory findings and cell immunophenotypes on first presentation were compared. RESULTS With respect to immunophenotype, influenza A infection resulted in a higher fraction of CD14+ and CD4+IL-17A+cells compared to children infected with influenza B. By contrast, influenza B infection resulted in a comparatively higher percentage of double-negative CD4-CD8- lymphocyte subsets. Influenza A infection was associated with comparatively higher percentages of CD4+CD25highFoxp3+ and CD4+CD25lowFoxp3+ cells. By contrast, the percentage of CD8+CD25high and CD8+CD25low cells was similar among patients with influenza A infection and influenza B infection. CONCLUSIONS An improved understanding of the fraction of regulatory T cells with influenza virus infections may provide further understandings on immune responses.
Collapse
Affiliation(s)
- Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan
| | - Shih-Min Wang
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan.
| | - Yu-Ting Liao
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan
| | - Yu-Shiang Hu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Shun-Hua Chen
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan; Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|
12
|
Liu L, Takeda K, Akkoyunlu M. Disease Stage-Specific Pathogenicity of CD138 (Syndecan 1)-Expressing T Cells in Systemic Lupus Erythematosus. Front Immunol 2020; 11:1569. [PMID: 32849532 PMCID: PMC7401833 DOI: 10.3389/fimmu.2020.01569] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
CD138 (syndecan 1), a member of the heparan-sulfate proteoglycan family, regulates diverse biological responses by interacting with chemokines, cytokines, growth factors, and adhesion molecules. Expression of CD138 has been detected on T cells from both healthy and sick mice mimicking systemic lupus erythematosus (SLE) disease. However, the characteristics and the role of CD138+ T cells in SLE pathogenesis remain largely unknown. We analyzed the lupus-prone MRL/Lpr mice and the control MRL/MpJ strain as well as the common laboratory strains Balb/c, and C57BL/6 for CD138-expression and found that only the MRL/Lpr strain harbored TCRβ+CD138+ cells in various organs. The frequency of TCRβ+CD138+ cells progressively expanded in MRL/Lpr mice with age and correlated with disease severity. Majority of the TCRβ+CD138+ cells were CD4 and CD8 double-negative and 20% were CD4. At least a portion of TCRβ+CD138+ cells originated from CD4+ cells because substantial number of CD4+TCRβ+CD138- cells expressed CD138 after in vitro cultivation. Compared to TCRβ+CD138- cells, TCRβ+CD138+ cells exhibited central memory (Tcm) phenotype with reduced ability to proliferate and produce the cytokines IFNγ and IL-17. When co-cultured with B cells, the ability of TCRβ+CD138+ cells to promote plasma cell formation and autoreactive antibody production was dependent on the presence of autoantigen, CD4 co-receptor expression and cell-to-cell contact. Surprisingly, adoptively transferred TCRβ+CD138+ T cells slowed down disease progression in young recipient MRL/Lpr mice but had the opposite effect when DNA was co-administered with TCRβ+CD138+ T cells or when TCRβ+CD138+ cells were transferred to older MRL/Lpr mice with established disease. Thus, CD138-expressing T cells with Tcm phenotype enhance disease progression in SLE by rapidly activating autoreactive B cells when self-antigens are exposed to the immune system.
Collapse
Affiliation(s)
- Lunhua Liu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
13
|
Partridge BR, O'Brien TJ, Lorenzo MF, Coutermarsh-Ott SL, Barry SL, Stadler K, Muro N, Meyerhoeffer M, Allen IC, Davalos RV, Dervisis NG. High-Frequency Irreversible Electroporation for Treatment of Primary Liver Cancer: A Proof-of-Principle Study in Canine Hepatocellular Carcinoma. J Vasc Interv Radiol 2020; 31:482-491.e4. [PMID: 31956003 PMCID: PMC7418213 DOI: 10.1016/j.jvir.2019.10.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To determine the safety and feasibility of percutaneous high-frequency irreversible electroporation (HFIRE) for primary liver cancer and evaluate the HFIRE-induced local immune response. MATERIALS AND METHODS HFIRE therapy was delivered percutaneously in 3 canine patients with resectable hepatocellular carcinoma (HCC) in the absence of intraoperative paralytic agents or cardiac synchronization. Pre- and post-HFIRE biopsy samples were processed with histopathology and immunohistochemistry for CD3, CD4, CD8, and CD79a. Blood was collected on days 0, 2, and 4 for complete blood count and chemistry. Numeric models were developed to determine the treatment-specific lethal thresholds for malignant canine liver tissue and healthy porcine liver tissue. RESULTS HFIRE resulted in predictable ablation volumes as assessed by posttreatment CT. No detectable cardiac interference and minimal muscle contraction occurred during HFIRE. No clinically significant adverse events occurred secondary to HFIRE. Microscopically, a well-defined ablation zone surrounded by a reactive zone was evident in the majority of samples. This zone was composed primarily of maturing collagen interspersed with CD3+/CD4-/CD8- lymphocytes in a proinflammatory microenvironment. The average ablation volumes for the canine HCC patients and the healthy porcine tissue were 3.89 cm3 ± 0.74 and 1.56 cm3 ± 0.16, respectively (P = .03), and the respective average lethal thresholds were 710 V/cm ± 28.2 and 957 V/cm ± 24.4 V/cm (P = .0004). CONCLUSIONS HFIRE can safely and effectively be delivered percutaneously, results in a predictable ablation volume, and is associated with lymphocytic tumor infiltration. This is the first step toward the use of HFIRE for treatment of unresectable liver tumors.
Collapse
Affiliation(s)
- Brittanie R Partridge
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Timothy J O'Brien
- Department of Biomedical Engineering and Mechanics, Virginia Tech University, Blacksburg, Virginia
| | - Melvin F Lorenzo
- Department of Biomedical Engineering and Mechanics, Virginia Tech University, Blacksburg, Virginia
| | - Sheryl L Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Sabrina L Barry
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Krystina Stadler
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Noelle Muro
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Mitchell Meyerhoeffer
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Irving C Allen
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech University, Blacksburg, Virginia
| | - Nikolaos G Dervisis
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, DSACS, Phase II, 205 Duck Pond Drive (0442), Blacksburg, VA 24061.
| |
Collapse
|
14
|
Maina E, Devriendt B, Cox E. Food allergen-specific sublingual immunotherapy modulates peripheral T cell responses of dogs with adverse food reactions. Vet Immunol Immunopathol 2019; 212:38-42. [PMID: 31213250 DOI: 10.1016/j.vetimm.2019.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 11/28/2022]
Abstract
Food allergen-specific sublingual immunotherapy (FA-SLIT) is a novel, safe and effective approach in dogs with adverse food reactions (AFR) to reduce their clinical symptoms. However, little is known about the specific immune components which mediate this reduction in clinical symptoms. In humans, regulatory T cells seem to play an important role in this desensitisation process. Here, we investigated changes in peripheral T cell responses of dogs with AFR upon FA-SLIT. Five dogs received a dose escalation of FA-SLIT over a six-month period. An oral food challenge was performed at the beginning and end of the study to assess the efficacy of the FA-SLIT. Using in vitro allergen-recall assays, we assessed the proliferation of T cell subsets before and at the end of the treatment. FA-SLIT significantly increased the percentage of proliferating CD4-CD8- double-negative (DN) T cells, while the percentage of allergen-specific CD4-CD8+ and CD4+CD8+ double-positive (DP) T cells decreased upon treatment. These findings indicate that sublingual immunotherapy in dogs activates DN T cells, which might be important for the desensitisation of dogs with adverse food reactions. However, further research is needed to corroborate these findings and to further elucidate the mechanism of action of FA-SLIT in dogs with AFR.
Collapse
Affiliation(s)
- Elisa Maina
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium.
| |
Collapse
|
15
|
Haug T, Aigner M, Peuser MM, Strobl CD, Hildner K, Mougiakakos D, Bruns H, Mackensen A, Völkl S. Human Double-Negative Regulatory T-Cells Induce a Metabolic and Functional Switch in Effector T-Cells by Suppressing mTOR Activity. Front Immunol 2019; 10:883. [PMID: 31105702 PMCID: PMC6498403 DOI: 10.3389/fimmu.2019.00883] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/05/2019] [Indexed: 01/07/2023] Open
Abstract
The recently discovered population of TCRαβ+ CD4–/CD8– (double-negative, DN) T-cells are highly potent suppressor cells in mice and humans. In preclinical transplantation models, adoptive transfer of DN T-cells specifically inhibits alloreactive T-cells and prevents transplant rejection or graft-vs.-host disease (GvHD). Interestingly, clinical studies in patients who underwent allogeneic stem cell transplantation reveal an inverse correlation between the frequency of circulating DN T-cells and the severity of GvHD, suggesting a therapeutic potential of human DN T-cells. However, their exact mode of action has not been elucidated yet. Investigating the impact of DN T-cells on conventional T-cells, we found that human DN T-cells selectively inhibit mTOR signaling in CD4 T-cells. Given that mTOR is a critical regulator of cellular metabolism, we further determined the impact of DN T-cells on the metabolic framework of T-cells. Intriguingly, DN T-cells diminished expression of glucose transporters and glucose uptake, whereas fatty acid uptake was not modified, indicating that DN T-cells prevent metabolic adaptation of CD4 T-cells upon activation (i.e., glycolytic switch) thereby contributing to their suppression. Further analyses demonstrated that CD4 T-cells also do not upregulate homing receptors associated with inflammatory processes. In contrast, expression of central memory-cell associated cell surface markers and transcription factors were increased by DN T-cells. Moreover, CD4 T-cells failed to produce inflammatory cytokines after co-culture with DN T-cells, whereas IL-2 secretion was enhanced. Taken together DN T-cells impair metabolic reprogramming of conventional CD4 T-cells by abrogating mTOR signaling, thereby modulating CD4 T-cell functionality. These results uncover a new mechanism of DN T-cell-mediated suppression, pointing out that DN T-cells could serve as cell-based therapy to limit alloreactive immune response.
Collapse
Affiliation(s)
- Tabea Haug
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Moritz M Peuser
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Carolin D Strobl
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Kai Hildner
- Department of Internal Medicine 1, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
16
|
Abstract
In this chapter, we describe the history of transplantation, the multiple cell types, and mechanisms that are involved in rejection and tolerance of a transplanted organ, as well as summarize the common and promising new therapeutics used in transplant patients.
Collapse
Affiliation(s)
- Jessica Stolp
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Masaaki Zaitsu
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
Wang L, Ni M, Hückelhoven-Krauss A, Sellner L, Hoffmann JM, Neuber B, Luft T, Hegenbart U, Schönland S, Kleist C, Sill M, Chen BA, Wuchter P, Eckstein V, Krüger W, Hilgendorf I, Yerushalmi R, Nagler A, Müller-Tidow C, Ho AD, Dreger P, Schmitt M, Schmitt A. Modulation of B Cells and Homing Marker on NK Cells Through Extracorporeal Photopheresis in Patients With Steroid-Refractory/Resistant Graft-Vs.-Host Disease Without Hampering Anti-viral/Anti-leukemic Effects. Front Immunol 2018; 9:2207. [PMID: 30349527 PMCID: PMC6186805 DOI: 10.3389/fimmu.2018.02207] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Graft-vs.-host disease (GvHD), a severe complication of allogeneic hematopoietic stem cell transplantation, significantly affects the post-transplant morbidity and mortality. Systemic steroids remain the gold standard for the initial management of GvHD. However, up to 60% of patients will not sufficiently respond to steroids. Extracorporeal photopheresis (ECP), a cell-based immunotherapy, has shown good clinical results in such steroid-refractory/resistant GvHD patients. Given its immunomodulatory, but not global immunosuppressive and steroid-sparing capacity, ECP constitutes an attractive option. In the case of GvHD, the balance of immune cells is destroyed: effector cells are not any longer efficiently controlled by regulatory cells. ECP therapy may restore this balance. However, the precise mechanism and the impact of ECP on anti-viral/anti-leukemic function remain unclear. In this study, 839 ECP treatments were performed on patients with acute GvHD (aGvHD) and chronic GvHD (cGvHD). A comprehensive analysis of effector and regulatory cells in patients under ECP therapy included multi-parametric flow cytometry and tetramer staining, LuminexTM-based cytokine, interferon-γ enzyme-linked immunospot, and chromium-51 release assays. Gene profiling of myeloid-derived suppressor cells (MDSCs) was performed by microarray analysis. Immunologically, modulations of effector and regulatory cells as well as proinflammatory cytokines were observed under ECP treatment: (1) GvHD-relevant cell subsets like CD62L+ NK cells and newly defined CD19hiCD20hi B cells were modulated, but (2) quantity and quality of anti-viral/anti-leukemic effector cells were preserved. (3) The development of MDSCs was promoted and switched from an inactivated subset (CD33-CD11b+) to an activated subset (CD33+CD11b+). (4) The frequency of Foxp3+CD4+ regulatory T cells (Tregs) and CD24+CD38hi regulatory B cells was considerably increased in aGvHD patients, and Foxp3+CD8+ Tregs in cGvHD patients. (5) Proinflammatory cytokines like IL-1β, IL-6, IL-8, and TNF-α were significantly reduced. In summary, ECP constitutes an effective immunomodulatory therapy for patients with steroid-refractory/resistant GvHD without impairment of anti-viral/leukemia effects.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,Department of Hematology, the Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, University Clinic Heidelberg, Heidelberg, Germany
| | - Martin Sill
- Division Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Bao-An Chen
- Department of Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Patrick Wuchter
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,German Red Cross Blood Service, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology Mannheim, Mannheim, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - William Krüger
- Department of Internal Medicine C, Haematology, Oncology, Stem Cell Transplantation, Palliative Care, University Clinic Greifswald, Greifswald, Germany
| | - Inken Hilgendorf
- Department of Internal Medicine II, University Clinic Jena, Jena, Germany
| | - Ronit Yerushalmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
18
|
Achita P, Dervovic D, Ly D, Lee JB, Haug T, Joe B, Hirano N, Zhang L. Infusion of ex-vivo expanded human TCR-αβ + double-negative regulatory T cells delays onset of xenogeneic graft-versus-host disease. Clin Exp Immunol 2018; 193:386-399. [PMID: 30066399 DOI: 10.1111/cei.13145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2018] [Indexed: 12/25/2022] Open
Abstract
Despite the demonstration of potent immunosuppressive function of T cell receptor (TCR)-αβ+ double-negative regulatory T cells (DN Tregs ), scarce numbers and lack of effective expansion method limit their clinical applications. Here we describe an approach that allows for ∼3500-fold ex-vivo expansion of human DN Tregs within 3 weeks with > 97% purity. Ex-vivo-expanded DN Tregs suppress proliferation of polyclonally stimulated autologous T and B cells in vitro through direct cell-to-cell contact. In vivo, we demonstrate for the first time that infusion of human DN Tregs delayed an onset of xenogeneic graft-versus-host disease (GVHD) significantly in a humanized mouse model. Furthermore, preincubation of ex-vivo-expanded DN Tregs with a mechanistic target of rapamycin (mTOR) inhibitor rapamycin enhanced their immune regulatory function further. Taken together, this study demonstrates that human DN Tregs can be expanded ex vivo to therapeutic numbers. The expanded DN Tregs can suppress proliferation of T and B cells and attenuate GVHD, highlighting the potential clinical use of DN Tregs to mitigate GVHD.
Collapse
Affiliation(s)
- P Achita
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, Toronto, ON, Canada
| | - D Dervovic
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - D Ly
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - J B Lee
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - T Haug
- Department of Internal Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - B Joe
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - N Hirano
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - L Zhang
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Saudemont A, Jespers L, Clay T. Current Status of Gene Engineering Cell Therapeutics. Front Immunol 2018; 9:153. [PMID: 29459866 PMCID: PMC5807372 DOI: 10.3389/fimmu.2018.00153] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/17/2018] [Indexed: 12/27/2022] Open
Abstract
Ex vivo manipulations of autologous patient’s cells or gene-engineered cell therapeutics have allowed the development of cell and gene therapy approaches to treat otherwise incurable diseases. These modalities of personalized medicine have already shown great promises including product commercialization for some rare diseases. The transfer of a chimeric antigen receptor or T cell receptor genes into autologous T cells has led to very promising outcomes for some cancers, and particularly for hematological malignancies. In addition, gene-engineered cell therapeutics are also being explored to induce tolerance and regulate inflammation. Here, we review the latest gene-engineered cell therapeutic approaches being currently explored to induce an efficient immune response against cancer cells or viruses by engineering T cells, natural killer cells, gamma delta T cells, or cytokine-induced killer cells and to modulate inflammation using regulatory T cells.
Collapse
Affiliation(s)
| | | | - Timothy Clay
- GlaxoSmithKline, Collegeville, PA, United States
| |
Collapse
|
20
|
Fortner KA, Bond JP, Austin JW, Boss JM, Budd RC. The molecular signature of murine T cell homeostatic proliferation reveals both inflammatory and immune inhibition patterns. J Autoimmun 2017; 82:47-61. [PMID: 28551033 PMCID: PMC5902411 DOI: 10.1016/j.jaut.2017.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/04/2017] [Accepted: 05/09/2017] [Indexed: 01/21/2023]
Abstract
T lymphocyte homeostatic proliferation, driven by the engagement of T cell antigen receptor with self-peptide/major histocompatibility complexes, and signaling through the common γ-chain-containing cytokine receptors, is critical for the maintenance of the T cell compartment and is regulated by the Fas death receptor (Fas, CD95). In the absence of Fas, Fas-deficient lymphoproliferation spontaneous mutation (lpr) mice accumulate homeostatically expanded T cells. The functional consequences of sequential rounds of homeostatic expansion are not well defined. We thus examined the gene expression profiles of murine wild-type and Fas-deficient lpr CD8+ T cell subsets that have undergone different amounts of homeostatic proliferation as defined by their level of CD44 expression, and the CD4-CD8-TCRαβ+ T cell subset that results from extensive homeostatic expansion of CD8+ T cells. Our studies show that recurrent T cell homeostatic proliferation results in global gene expression changes, including the progressive upregulation of both cytolytic proteins such as Fas-Ligand and granzyme B as well as inhibitory proteins such as programmed cell death protein 1 (PD-1) and lymphocyte activating 3 (Lag3). These findings provide an explanation for how augmented T cell homeostatic expansion could lead to the frequently observed clinical paradox of simultaneous autoinflammatory and immunodeficiency syndromes and provide further insight into the regulatory programs that control chronically stimulated T cells.
Collapse
Affiliation(s)
- Karen A Fortner
- Department of Medicine, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA.
| | - Jeffrey P Bond
- Department of Microbiology and Molecular Genetics, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA
| | - James W Austin
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ralph C Budd
- Department of Medicine, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA
| |
Collapse
|
21
|
Lu X, Su B, Xia H, Zhang X, Liu Z, Ji Y, Yang Z, Dai L, Mayr LM, Moog C, Wu H, Huang X, Zhang T. Low Double-Negative CD3 +CD4 -CD8 - T Cells Are Associated with Incomplete Restoration of CD4 + T Cells and Higher Immune Activation in HIV-1 Immunological Non-Responders. Front Immunol 2016; 7:579. [PMID: 28018346 PMCID: PMC5145861 DOI: 10.3389/fimmu.2016.00579] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022] Open
Abstract
Failure of immune reconstitution increases the risk of AIDS or non-AIDS related morbidity and mortality in HIV-1-infected patients. CD3+CD4−CD8− T cells, which are usually described as double-negative (DN) T cells, display CD4-like helper and immunoregulatory functions. Here, we have measured the percentage of DN T cells in the immune reconstituted vs. non-immune reconstituted HIV-1-infected individuals. We observed that immunological non-responders (INRs) had a low number of DN T cells after long-term antiretroviral therapy (ART), and the number of these cells positively correlated with the CD4+ T cell count. The ART did not result in complete suppression of immune activation recorded by the percentage of CD38+HLA-DR+CD8+ T cells in INRs, and a strong inverse correlation was observed between DN T cells and immune activation. A low proportion of TGF-β1+DN T cells was found in INRs. Further mechanism study demonstrated that the level of TGF-β1-producing DN T cells and immune activation had a negative correlation after ART. Taken together, our study suggests that DN T cells control the immunological response in HIV-1-infected patients. These findings expand our understanding of the mechanism of immune reconstitution and could develop specific treatments to return the immune system to homeostasis following initiation of HIV-1 therapy.
Collapse
Affiliation(s)
- Xiaofan Lu
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Bin Su
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Huan Xia
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Xin Zhang
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Zhiying Liu
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Yunxia Ji
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Zixuan Yang
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Lili Dai
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Luzia M Mayr
- INSERM UMR S_1109, Faculté de Médecine, FMTS, Centre de Recherche en Immunologie et Hématologie, Université de Strasbourg , Strasbourg , France
| | - Christiane Moog
- INSERM UMR S_1109, Faculté de Médecine, FMTS, Centre de Recherche en Immunologie et Hématologie, Université de Strasbourg , Strasbourg , France
| | - Hao Wu
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Xiaojie Huang
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University , Beijing , China
| | - Tong Zhang
- STD/HIV Research Laboratory, Beijing You'an Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory for HIV/AIDS Research, Beijing, China; Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Cong M, Liu T, Tian D, Guo H, Wang P, Liu K, Lin J, Tian Y, Shi W, You H, Jia J, Zhang D. Interleukin-2 Enhances the Regulatory Functions of CD4+T Cell-Derived CD4−CD8− Double Negative T Cells. J Interferon Cytokine Res 2016; 36:499-505. [PMID: 27135902 DOI: 10.1089/jir.2015.0093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Min Cong
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Tianhui Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Hongbo Guo
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Ping Wang
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Jun Lin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Yue Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Hong You
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Jidong Jia
- National Clinical Research Center of Digestive Diseases, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| |
Collapse
|
23
|
Stallone G, Infante B, Di Lorenzo A, Rascio F, Zaza G, Grandaliano G. mTOR inhibitors effects on regulatory T cells and on dendritic cells. J Transl Med 2016; 14:152. [PMID: 27245075 PMCID: PMC4886438 DOI: 10.1186/s12967-016-0916-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/18/2016] [Indexed: 01/03/2023] Open
Abstract
The mammalian target of rapamycin (mTOR), a cytoplasmic serine/threonine kinase, represents a key biologic "switch" modulating cell metabolisms in response to environmental signals and is now recognized as a central regulator of the immune system. There is an increasing body of evidence supporting the hypothesis that mTOR inhibitors exhibit several biological properties in addition to immunosuppression, including anti-neoplastic effects, cardio-protective activities, and an array of immunomodulatory actions facilitating the development of an operational graft tolerance. The biological mechanisms explaining how mTOR inhibition can enable a tolerogenic state are still largely unclear. The induction of transplant tolerance might at the same time decrease rejection rate and minimize immunosuppression-related side effects, leading to an improvement in long-term graft outcome. In this scenario, T cell immunoregulation has been defined as the hallmark of peripheral tolerance. Two main immunologic cell populations have been reported to play a central role in this setting: regulatory T cells (Tregs) and dendritic cells (DCs). In this review we focus on mTOR inhibitors effects on Treg and DCs differentiation, activation, and function in the transplantation setting.
Collapse
Affiliation(s)
- Giovanni Stallone
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Barbara Infante
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Adelaide Di Lorenzo
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Federica Rascio
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy.
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy.
| |
Collapse
|
24
|
Hyperactive mTOR pathway promotes lymphoproliferation and abnormal differentiation in autoimmune lymphoproliferative syndrome. Blood 2016; 128:227-38. [PMID: 27099149 DOI: 10.1182/blood-2015-11-685024] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/14/2016] [Indexed: 12/23/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a human disorder characterized by defective Fas signaling, resulting in chronic benign lymphoproliferation and accumulation of TCRαβ(+) CD4(-) CD8(-) double-negative T (DNT) cells. Although their phenotype resembles that of terminally differentiated or exhausted T cells, lack of KLRG1, high eomesodermin, and marginal T-bet expression point instead to a long-lived memory state with potent proliferative capacity. Here we show that despite their terminally differentiated phenotype, human ALPS DNT cells exhibit substantial mitotic activity in vivo. Notably, hyperproliferation of ALPS DNT cells is associated with increased basal and activation-induced phosphorylation of serine-threonine kinases Akt and mechanistic target of rapamycin (mTOR). The mTOR inhibitor rapamycin abrogated survival and proliferation of ALPS DNT cells, but not of CD4(+) or CD8(+) T cells in vitro. In vivo, mTOR inhibition reduced proliferation and abnormal differentiation by DNT cells. Importantly, increased mitotic activity and hyperactive mTOR signaling was also observed in recently defined CD4(+) or CD8(+) precursor DNT cells, and mTOR inhibition specifically reduced these cells in vivo, indicating abnormal programming of Fas-deficient T cells before the DNT stage. Thus, our results identify the mTOR pathway as a major regulator of lymphoproliferation and aberrant differentiation in ALPS.
Collapse
|
25
|
A novel differentiation pathway from CD4⁺ T cells to CD4⁻ T cells for maintaining immune system homeostasis. Cell Death Dis 2016; 7:e2193. [PMID: 27077809 PMCID: PMC4855662 DOI: 10.1038/cddis.2016.83] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/07/2016] [Accepted: 03/09/2016] [Indexed: 12/26/2022]
Abstract
CD4+ T lymphocytes are key players in the adaptive immune system and can differentiate into a variety of effector and regulatory T cells. Here, we provide evidence that a novel differentiation pathway of CD4+ T cells shifts the balance from a destructive T-cell response to one that favors regulation in an immune-mediated liver injury model. Peripheral CD4−CD8−NK1.1− double-negative T cells (DNT) was increased following Concanavalin A administration in mice. Adoptive transfer of DNT led to significant protection from hepatocyte necrosis by direct inhibition on the activation of lymphocytes, a process that occurred primarily through the perforin-granzyme B route. These DNT converted from CD4+ rather than CD8+ T cells, a process primarily regulated by OX40. DNT migrated to the liver through the CXCR3-CXCL9/CXCL10 interaction. In conclusion, we elucidated a novel differentiation pathway from activated CD4+ T cells to regulatory DNT cells for maintaining homeostasis of the immune system in vivo, and provided key evidence that utilizing this novel differentiation pathway has potential application in the prevention and treatment of autoimmune diseases.
Collapse
|
26
|
Wu D, Wang H, Yan W, Chen T, Wang M, Han M, Wu Z, Wang X, Ai G, Xi D, Shen G, Luo X, Ning Q. A disparate subset of double-negative T cells contributes to the outcome of murine fulminant viral hepatitis via effector molecule fibrinogen-like protein 2. Immunol Res 2016; 64:518-530. [PMID: 26482053 DOI: 10.1007/s12026-015-8727-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The underlying immune-mediated mechanisms involved in virus-induced severe hepatitis have not been well elucidated. In this study, we investigated the role of CD3(+)CD4(-)CD8(-) double-negative T (DN T) cells in the pathogenesis of fulminant viral hepatitis (FVH) induced by murine hepatitis virus strain 3 (MHV-3). After MHV-3 infection, the proportions of DN T cells increased significantly in BALB/cJ mice, and splenic DN T cells expressing high levels of CD69 were recruited by MHV-3-infected hepatocytes to the liver. Serum levels of alanine aminotransferase, aspartate aminotransferase and total bilirubin increased, accompanied by massive hepatocyte necrosis. These DN T cells were predominantly consisted of a TCRαβ(+) subset expressing high levels of CD44 and did not produce cytokine except IL-2. Adoptive transfer of this subset of DN T cells to the MHV-3-infected mice resulted in an increase in murine fibrinogen-like protein 2 (mfgl2) expressions in association with massive fibrin deposition in the liver. Following MHV-3 infection, membrane mfgl2 expression and functional procoagulant activity increased remarkably in the DN T cells. Introduction of a recombinant adenovirus which encoded a microRNA specifically targeting mfgl2 gene (Ad-mfgl2-miRNA) in vivo significantly inhibited the hepatic expression of mfgl2 and improved survival in mice. However, under this condition, adoptive transfer of the DN T cells accelerated the disease progression and reversed the benefit from mfgl2 gene silence, leading to a 100 % death rate. Our results demonstrate that DN T cells contribute to the outcome of MHV-3-induced FVH via an important effector molecule mfgl2.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Fibrinogen/genetics
- Fibrinogen/metabolism
- Gene Silencing
- Hepatitis, Viral, Animal/immunology
- Hepatitis, Viral, Animal/metabolism
- Hepatitis, Viral, Animal/mortality
- Hepatitis, Viral, Animal/therapy
- Immunophenotyping
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Lymphocyte Activation
- Mice
- Murine hepatitis virus/immunology
- Phenotype
- RNA, Small Interfering/genetics
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Di Wu
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongwu Wang
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiming Yan
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tao Chen
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Wang
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meifang Han
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zeguang Wu
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaojing Wang
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guo Ai
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong Xi
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guanxin Shen
- Institute of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Ning
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
27
|
Lech M, Lorenz G, Kulkarni OP, Grosser MOO, Stigrot N, Darisipudi MN, Günthner R, Wintergerst MWM, Anz D, Susanti HE, Anders HJ. NLRP3 and ASC suppress lupus-like autoimmunity by driving the immunosuppressive effects of TGF-β receptor signalling. Ann Rheum Dis 2015; 74:2224-35. [PMID: 25135254 DOI: 10.1136/annrheumdis-2014-205496] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/30/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVES The NLRP3/ASC inflammasome drives host defence and autoinflammatory disorders by activating caspase-1 to trigger the secretion of mature interleukin (IL)-1β/IL-18, but its potential role in autoimmunity is speculative. METHODS We generated and phenotyped Nlrp3-deficient, Asc-deficient, Il-1r-deficient and Il-18-deficient C57BL/6-lpr/lpr mice, the latter being a mild model of spontaneous lupus-like autoimmunity. RESULTS While lack of IL-1R or IL-18 did not affect the C57BL/6-lpr/lpr phenotype, lack of NLRP3 or ASC triggered massive lymphoproliferation, lung T cell infiltrates and severe proliferative lupus nephritis within 6 months, which were all absent in age-matched C57BL/6-lpr/lpr controls. Lack of NLRP3 or ASC increased dendritic cell and macrophage activation, the expression of numerous proinflammatory mediators, lymphocyte necrosis and the expansion of most T cell and B cell subsets. In contrast, plasma cells and autoantibody production were hardly affected. This unexpected immunosuppressive effect of NLRP3 and ASC may relate to their known role in SMAD2/3 phosphorylation during tumour growth factor (TGF)-β receptor signalling, for example, Nlrp3-deficiency and Asc-deficiency significantly suppressed the expression of numerous TGF-β target genes in C57BL/6-lpr/lpr mice and partially recapitulated the known autoimmune phenotype of Tgf-β1-deficient mice. CONCLUSIONS These data identify a novel non-canonical immunoregulatory function of NLRP3 and ASC in autoimmunity.
Collapse
Affiliation(s)
- Maciej Lech
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Georg Lorenz
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Onkar P Kulkarni
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Marian O O Grosser
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Nora Stigrot
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Murthy N Darisipudi
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Roman Günthner
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Maximilian W M Wintergerst
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - David Anz
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Heni Eka Susanti
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilians Universität, München-Innenstadt, Munich, Germany
| |
Collapse
|
28
|
Advances on Non-CD4 + Foxp3+ T Regulatory Cells: CD8+, Type 1, and Double Negative T Regulatory Cells in Organ Transplantation. Transplantation 2015; 99:1553-9. [PMID: 26193065 DOI: 10.1097/tp.0000000000000813] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The overwhelming body of research on T regulatory cells (Treg) has focused on CD4 + CD25 + Foxp3+ T cells. However, recent years have witnessed a resurgence in interest in CD4 - CD8+, CD4 - CD8- (double negative [DN]), and CD4 + Foxp3- type 1 Treg (Tr1) Treg and their role in controlling autoimmune diseases and in promoting the survival of organ allografts and xenografts. CD8+ and DN Treg can arise spontaneously (natural Treg) or can be induced in situ. Both CD8+ and DN Treg have been shown to enhance the survival of organ allografts and xenografts. Additionally, both can suppress alloimmune responses by contact-dependent mechanisms by either inducing apoptosis or mediating direct cytolysis of effector T cells. CD8+, DN, and Tr1 Treg can also act in a contact-independent manner by elaborating soluble immunosuppressive factors, such as TGF-β and IL-10. Applying CD8+, DN, and Tr1 Treg for enhancing the survival of organ allografts and xenografts is still in its infancy but holds significant potential. Furthermore, there is a need for a more comprehensive understanding of how current immunosuppressive therapies applied to organ transplantations affect the wide array of Treg populations.
Collapse
|
29
|
Double negative (DN) αβ T cells: misperception and overdue recognition. Immunol Cell Biol 2014; 93:305-10. [PMID: 25420721 DOI: 10.1038/icb.2014.99] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/24/2022]
Abstract
CD4(-)CD8(-)double negative (DN) αβ T cells are legitimate components of the normal immune system. However, they are poorly understood and largely ignored by immunologists because of their historical association with the lymphoproliferation that occurs in mice (lpr and gld) and humans (autoimmune lymphoproliferative syndromes patients) with impaired Fas-mediated apoptosis where they are considered abnormal T cells. We believe that the traditional view that DN T cells that cause lymphoproliferation (hereafter referred to as lpr DN T cells) are CD4 and CD8 T cells that lost their coreceptor, conceived more than two decades ago, is flawed and that conflating lpr DN T cells with DN T cells found in normal immune system (hereafter referred to as nDN T cells) is unnecessarily dampening interest of this potentially important cell type. To begin rectifying these misperceptions, we will revisit the traditional view of lpr DN T cells and show that it does not hold true in light of recent immunological advances. In lieu of it, we offer a new model proposing that Fas-mediated apoptosis actively removes normally existing DN T cells from the periphery and that impaired Fas-mediated apoptosis leads to accumulation of these cells rather than de novo generation of DN T cells from activated CD4 or CD8 T cells. By doing so, we hope to provoke a new discussion that may lead to a consensus about the origin of lpr DN T cells and regulation of their homeostasis by the Fas pathway and reignite wider interest in nDN T cells.
Collapse
|
30
|
Jabbour MN, Issa G, Charafeddine K, Simaan Y, Karam M, Khalifeh H, Habib R, Khalifeh I. The immune microenvironment in cutaneous leishmaniasis. J Eur Acad Dermatol Venereol 2014; 29:1170-9. [PMID: 25351105 DOI: 10.1111/jdv.12781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/19/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND Cutaneous leishmaniasis is an infection that has spread to non-endemic regions, stimulating recent interest for the enhanced understanding of this disease. Downregulation of the CD1a receptor on Langerhans cells has been described in various cutaneous infections. OBJECTIVE In this study, the immune response across different Ridley patterns and parasitic indices is outlined in a case series of cutaneous leishmaniasis. METHODS Skin punch biopsies from the interface of normal and lesional cutaneous leishmaniasis were collected from 33 patients with molecularly confirmed Leishmania tropica or L. major infection. Ridley patterns (2-5) were assessed for various clinicopathological features including age, gender, disease duration, parasitic index and constituents of the inflammatory infiltrate. CD1a, CD68, CD3, CD4, CD8, CD20 and CD138 stains were performed on normal skin tissue, cutaneous leishmaniasis biopsies and cytospin/cell block cytology preparations of cultured leishmania promastigotes. CD1a was quantified per mm2 in the epidermis and dermis. The remaining stains were graded according to a 4-tiered grading system [0 (0-4%); 1 (5-24%); 2 (25-49%); 3 (50-74%) and 4 (75-100%). RESULTS Total CD1a expression significantly decreased (14-fold) from parasitic indices (0-2) to (5-6); (ρ < 0.001). CD1a expression in the epidermis was at least 5-fold lower than normal skin (58 vs. 400 cells/mm2), inversely correlating with the parasitic index. There was an increase in dermal CD1a Langerhans cells (33 vs. 0 cells/mm² in the dermis). CD1a and CD68 staining of amastigotes was strong and diffuse, whereas promastigotes were negative. The major inflammatory infiltrate, in all Ridley patterns, consisted of macrophages and double-negative CD3(+) CD4(-) CD8(-) T lymphocytes. The double-negative CD3 T cells formed a ring around the parasitic laden macrophages. Apart from CD1a, there was no significant difference in inflammatory markers between the various Ridley patterns and parasitic indices. Disease duration did not correlate with Ridley pattern. CONCLUSION The significant decrease in CD1a expression is postulated by two mechanisms; either via direct CD1a receptor uptake by leishmania amastigotes and/or negative feedback inhibition of CD1a Langerhans cells by double-negative CD3 T-regulatory cells. Modulation of the immune microenvironment in cutaneous leishmaniasis represents a potential therapeutic and prophylactic target.
Collapse
Affiliation(s)
- M N Jabbour
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - G Issa
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - K Charafeddine
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Y Simaan
- Department of Biology, University of Balamand, Tripoli, Lebanon
| | - M Karam
- Department of Biology, University of Balamand, Tripoli, Lebanon
| | - H Khalifeh
- Children's Cancer Center Lebanon, American University of Beirut Medical Center, Beirut, Lebanon
| | - R Habib
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - I Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
31
|
Collin R, Dugas V, Pelletier AN, Chabot-Roy G, Lesage S. The mouse idd2 locus is linked to the proportion of immunoregulatory double-negative T cells, a trait associated with autoimmune diabetes resistance. THE JOURNAL OF IMMUNOLOGY 2014; 193:3503-12. [PMID: 25165153 DOI: 10.4049/jimmunol.1400189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Autoimmune diseases result from a break in immune tolerance. Various mechanisms of peripheral tolerance can protect against autoimmunity, including immunoregulatory CD4(-)CD8(-) double-negative (DN) T cells. Indeed, we have previously shown that diabetes-prone mouse strains exhibit a low proportion of DN T cells relative to that of diabetes-resistant mice, and that a single autologous transfer of DN T cells can impede autoimmune diabetes development, at least in the 3A9 TCR transgenic setting. In this study, we aim to understand the genetic basis for the difference in DN T cell proportion between diabetes-resistant and diabetes-prone mice. We thus perform an unbiased linkage analysis in 3A9 TCR F2 (NOD.H2(k) × B10.BR) mice and reveal that a locus on chromosome 9, which coincides with Idd2, is linked to the proportion of DN T cells in the lymph nodes. We generate two NOD.H2(k).B10-Chr9 congenic mouse strains and validate the role of this genetic interval in defining the proportion of DN T cells. Moreover, we find that the increased proportion of DN T cells in lymphoid organs is associated with a decrease in both diabetes incidence and serum IgG Ab levels. Together, the data suggest that Idd2 is linked to DN T cell proportion and that a physiological increase in DN T cell number may be sufficient to confer resistance to autoimmune diabetes. Altogether, these findings could help identify new candidate genes for the development of therapeutic avenues aimed at modulating DN T cell number for the prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Roxanne Collin
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Véronique Dugas
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Adam-Nicolas Pelletier
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Geneviève Chabot-Roy
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada
| | - Sylvie Lesage
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 0G4, Canada
| |
Collapse
|
32
|
Martina MN, Bandapalle S, Rabb H, Hamad AR. Isolation of double negative αβ T cells from the kidney. J Vis Exp 2014. [PMID: 24893925 DOI: 10.3791/51192] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
There is currently no standard protocol for the isolation of DN T cells from the non-lymphoid tissues despite their increasingly reported involvement in various immune responses. DN T cells are a unique immune cell type that has been implicated in regulating immune and autoimmune responses and tolerance to allotransplants(1-6). DN T cells are, however, rare in peripheral blood and secondary lymphoid organs (spleen and lymph nodes), but are major residents of the normal kidney. Very little is known about their pathophysiologic function(7) due to their paucity in the periphery. We recently described a comprehensive phenotypic and functional analysis of this population in the kidney(8) in steady state and during ischemia reperfusion injury. Analysis of DN T cell function will be greatly enhanced by developing a protocol for their isolation from the kidney. Here, we describe a novel protocol that allows isolation of highly pure ab CD4+ CD8+ T cells and DN T cells from the murine kidney. Briefly, we digest kidney tissue using collagenase and isolate kidney mononuclear cells (KMNC) by density gradient. This is followed by two steps to enrich hematopoietic T cells from 3% to 70% from KMNC. The first step consists of a positive selection of hematopoietic cells using a CD45+ isolation kit. In the second step, DN T cells are negatively isolated by removal of non-desired cells using CD4, CD8, and MHC class II monoclonal antibodies and CD1d α-galcer tetramer. This strategy leads to a population of more than 90% pure DN T cells. Surface staining with the above mentioned antibodies followed by FACs analysis is used to confirm purity.
Collapse
Affiliation(s)
- Maria N Martina
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | - Hamid Rabb
- Department of Medicine, Johns Hopkins University School of Medicine
| | - Abdel R Hamad
- Department of Pathology, Johns Hopkins University School of Medicine;
| |
Collapse
|
33
|
Li W, Tian Y, Li Z, Gao J, Shi W, Zhu J, Zhang D. Ex vivo converted double negative T cells suppress activated B cells. Int Immunopharmacol 2014; 20:164-9. [PMID: 24613134 DOI: 10.1016/j.intimp.2014.02.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/21/2014] [Accepted: 02/24/2014] [Indexed: 02/06/2023]
Abstract
Although the ability of endogenous CD4(-)CD8(-) double negative (DN) T cells to suppress B cells has been documented, the extent to which ex vivo converted DN T cells suppress B cells activity is still being explored. The aim of this study was to determine whether and what extent ex vivo converted CD4(-)CD8(-) DN T cells suppress B cell activation and antibody production. We found that ex vivo converted DN T cells suppressed proliferation of activated B cells in a perforin and cell-cell contact dependent manner. In addition, ex vivo converted DN T cells significantly inhibited the production of IgG by stimulated B cells. This study provides evidence that ex vivo converted CD4(-)CD8(-) double negative T cells can down-regulate immune responses by suppressing B cell proliferation and IgG production, and supports efforts to develop ex vivo DN T cell therapies.
Collapse
Affiliation(s)
- WenXia Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yue Tian
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Wen Shi
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China
| | - JiYe Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Dong Zhang
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China.
| |
Collapse
|
34
|
Su Y, Jevnikar AM, Huang X, Lian D, Zhang ZX. Spi6 protects alloreactive CD4(+) but not CD8 (+) memory T cell from granzyme B attack by double-negative T regulatory cell. Am J Transplant 2014; 14:580-593. [PMID: 24730048 DOI: 10.1111/ajt.12614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Memory T (Tm) cells pose a major barrier to long-term transplant survival. Whether regulatory T cells (Tregs)can control them remains poorly defined. Previously,we established that double-negative (DN) Tregs suppress effector T (Teff) cells. Here, we demonstrate that DNTregs effectively suppress CD4+/CD8+Teff and CD8+Tm but not CD4+Tm cells, whereas the suppression on CD8+Tm is abrogated by perforin (PFN) deficiency in DNTregs. Consistently, in a BALB/c to B6-Rag1-/-skin transplantation, transfer of DN Tregs suppressed the rejection mediated by CD4þ/CD8+Teff and CD8+Tmcells (76.0±4.9, 87.5±5.0 and 63.0±4.7 days, respectively)but not CD4þTmcells (25.3±1.4 days). Both CD8þ effector memory T and central memory T compartments significantly reduced after DN Treg transfer. CD4+Tm highly expresses granzyme B (GzmB) inhibitor serine protease inhibitor-6 (Spi6). Spi6 deficiency renders CD4þTm susceptible to DN Treg suppression. In addition,transfer of WT DN Tregs, but not PFN-/-DN Tregs,inhibited the skin allograft rejection mediated by Spi6-/-CD4þTm(75.5±7.9 days). In conclusion, CD4+ and CD8+Tm cells differentially respond toDNTregs’ suppression.The GzmB resistance conferred by Spi6 in CD4þTm cells might hint at the physiological significance of Tmpersistence
Collapse
|
35
|
Liang Q, Jiao Y, Zhang T, Wang R, Li W, Zhang H, Huang X, Tang Z, Wu H. Double Negative (DN) [CD3⁺CD4⁻CD8⁻] T cells correlate with disease progression during HIV infection. Immunol Invest 2014; 42:431-7. [PMID: 23802173 DOI: 10.3109/08820139.2013.805763] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Although double negative T (DNT) cells (CD3⁺CD4⁻CD8⁻) share some characteristics with T regulatory cells, the relationship between DNT cells and disease progression in HIV infection is unclear. In this study, we analyzed the relationship between DNT cells and disease progression during the first 2 years of HIV infection. We found that DNT cell numbers tended to decrease with disease progression. There was a positive correlation between DNT cells and CD4 counts. The DNT cell numbers were significantly lower in the high viral load group compared with the low viral load group. Therefore, we conclude that DNT cells correlated with disease progression in HIV infection. These data provide valuable information for further understanding of the role of DNT cells during HIV infection.
Collapse
Affiliation(s)
- Qi Liang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Gao J, McIntyre MSF, D'Souza CA, Zhang L. Pretransplant infusion of donor B cells enhances donor-specific skin allograft survival. PLoS One 2013; 8:e77761. [PMID: 24204953 PMCID: PMC3810130 DOI: 10.1371/journal.pone.0077761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 09/04/2013] [Indexed: 01/06/2023] Open
Abstract
Pretransplant donor lymphocyte infusion (DLI) has been shown to enhance donor-specific allograft survival in rodents, primates and humans. However, the cell subset that is critical for the DLI effect and the mechanisms involved remain elusive. In this study, we monitored donor cell subsets after DLI in a murine MHC class I Ld-mismatched skin transplantation model. We found that donor B cells, but not DCs, are the major surviving donor APCs in recipients following DLI. Infusing donor B, but not non-B, cells resulted in significantly enhanced donor-specific skin allograft survival. Furthermore, mice that had received donor B cells showed higher expression of Ly6A and CD62L on antigen-specific TCRαβ+CD3+CD4−CD8−NK1.1− double negative (DN) regulatory T cells (Tregs). B cells presented alloantigen to DN Tregs and primed their proliferation in an antigen-specific fashion. Importantly, DN Tregs, activated by donor B cells, showed increased cytotoxicity toward anti-donor CD8+ T cells. These data demonstrate that donor B cells can enhance skin allograft survival, at least partially, by increasing recipient DN Treg-mediated killing of anti-donor CD8+ T cells. These findings provide novel insights into the mechanisms underlying DLI-induced transplant tolerance and suggest that DN Tregs have great potential as an antigen-specific immune therapy to enhance allograft survival.
Collapse
Affiliation(s)
- Julia Gao
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Megan S. Ford. McIntyre
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl A. D'Souza
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Li Zhang
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
37
|
Helper T cells down-regulate CD4 expression upon chronic stimulation giving rise to double-negative T cells. Cell Immunol 2013; 284:68-74. [PMID: 23933188 DOI: 10.1016/j.cellimm.2013.06.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/13/2013] [Accepted: 06/24/2013] [Indexed: 11/23/2022]
Abstract
Double-negative T (DNT) cells are αβTCR(+)CD3(+)CD4(-)CD8(-)NK1.1(-) cells that constitute a small but significant proportion of the αβTCR(+) T cells. Their developmental pathway and pathological significance remain unclear. In the present study, we utilized chronic in vitro stimulation of CD4(+) T cells to mimic immune hyper-activation of autoimmune lymphoproliferative syndrome and systemic lupus erythematosus, conditions characterized by DNT cells accumulation. After approximately 4-5 rounds of stimulation, the CD3(+)CD4(-) population became apparent. These cells did not express CD8, NK1.1, γδTCR, or B220, exhibited a highly proliferative effector phenotype, and were dependent on T cell receptor (TCR) stimulation for survival. Moreover, CD3(+)CD4(-) cells expressed MHC class II-restricted αβTCR, indicative of their origin from a CD4(+) T cell population. The results presented herein illustrate a novel method of DNT cell generation in vitro and suggest that immune hyper-activation could also be implicated in the genesis of the disease-associated DNT cells in vivo.
Collapse
|
38
|
Juvet SC, Thomson CW, Kim EY, Han M, Zhang L. FcRγ controls the fas-dependent regulatory function of lymphoproliferative double negative T cells. PLoS One 2013; 8:e65253. [PMID: 23762329 PMCID: PMC3675138 DOI: 10.1371/journal.pone.0065253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/24/2013] [Indexed: 12/21/2022] Open
Abstract
Patients with autoimmune lymphoproliferative syndrome (ALPS) and lymphoproliferation (LPR) mice are deficient in Fas, and accumulate large numbers of αβ-TCR+, CD4−, CD8− double negative (DN) T cells. The function of these DN T cells remains largely unknown. The common γ subunit of the activating Fc receptors, FcRγ, plays an important role in mediating innate immune responses. We have shown previously that a significant proportion of DN T cells express FcRγ, and that this molecule is required for TCR transgenic DN T cells to suppress allogeneic immune responses. Whether FcRγ plays a critical role in LPR DN T cell-mediated suppression of immune responses to auto and allo-antigens is not known. Here, we demonstrated that FcRγ+, but not FcRγ− LPR DN T cells could suppress Fas+ CD4+ and CD8+ T cell proliferation in vitro and attenuated CD4+ T cell-mediated graft-versus host disease. Although FcRγ expression did not allow LPR DN T cells to inhibit the expansion of Fas-deficient cells within the LPR context, adoptive transfer of FcRγ+, but not FcRγ−, DN T cells inhibited lymphoproliferation in generalized lymphoproliferative disease (GLD) mice. Furthermore, FcRγ acted in a cell-intrinsic fashion to limit DN T cell accumulation by increasing the rate of apoptosis in proliferated cells. These results indicate that FcRγ can confer Fas-dependent regulatory properties on LPR DN T cells, and suggest that FcRγ may be a novel marker for functional DN Tregs.
Collapse
Affiliation(s)
- Stephen C. Juvet
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Respirology and Toronto Lung Transplant Program, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Clinician-Scientist Training Program, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Christopher W. Thomson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Edward Y. Kim
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mei Han
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Li Zhang
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
39
|
Identification of CD3+CD4-CD8- T cells as potential regulatory cells in an experimental murine model of graft-versus-host skin disease (GVHD). J Invest Dermatol 2013; 133:2538-2545. [PMID: 23648548 PMCID: PMC3795811 DOI: 10.1038/jid.2013.212] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 11/11/2022]
Abstract
We have developed K14-mOVA transgenic (Tg) mice that express membrane-associated ovalbumin (mOVA) under the control of a K14 promoter as well as double Tg mice by crossing them with OT-I mice that have a T cell receptor (TCR) recognizing OVA peptide. When injected with CD8+ OT-I cells, K14-mOVATg mice develop graft-vs-host disease (GvHD), whereas double Tg mice are protected. This suggests that, in double Tg mice, regulatory mechanisms may prevent infused OT-I cells from inducing GvHD. We demonstrated that, after adoptive transfer, TCRαβ+CD3+CD4-CD8-NK1.1- double negative (DN) T cells are increased in the peripheral lymphoid organs and skin of double Tg mice and exhibit a Vα2+Vβ5+TCR that is the same TCR specificity as OT-I cells. These DN T cells isolated from tolerant double Tg mice proliferated in response to OVA peptide and produced IFN-γ in the presence of IL-2. These cells could also suppress the proliferation of OT-I cells and were able to specifically kill activated OT-I cells through Fas/Fas ligand interaction. These findings suggest that DN T cells that accumulate in double Tg mice have regulatory functions and may play a role in the maintenance of peripheral tolerance in vivo.
Collapse
|
40
|
Hillhouse EE, Delisle JS, Lesage S. Immunoregulatory CD4(-)CD8(-) T cells as a potential therapeutic tool for transplantation, autoimmunity, and cancer. Front Immunol 2013; 4:6. [PMID: 23355840 PMCID: PMC3553425 DOI: 10.3389/fimmu.2013.00006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 01/05/2013] [Indexed: 11/17/2022] Open
Abstract
A central objective in organ transplantation and the treatment or prevention of autoimmune disease is the achievement of antigen-specific immune tolerance. An additional challenge in bone marrow transplantation for the treatment of hematological malignancies is the prevention of graft-vs-host disease (GVHD) while maintaining graft-vs-tumor activity. Interestingly, CD4-CD8- (double negative, DN) T cells, which exhibit a unique antigen-specific immunoregulatory potential, appear to exhibit all of the properties to respond to these challenges. Herein, we review the therapeutic potential of immunoregulatory DN T cells in various immunopathological settings, including graft tolerance, GVHD, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Erin E Hillhouse
- Department of Microbiology and Immunology, University of Montreal Montreal, QC, Canada ; Research Center, Maisonneuve-Rosemont Hospital Montreal, QC, Canada
| | | | | |
Collapse
|
41
|
Trivedi HL, Vanikar AV, Kute VB, Patel HV, Gumber MR, Shah PR, Dave SD, Trivedi VB. The effect of stem cell transplantation on immunosuppression in living donor renal transplantation: a clinical trial. Int J Organ Transplant Med 2013; 4:155-62. [PMID: 25013669 PMCID: PMC4089327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND/OBJECTIVE We designed a clinical trial on a group of live-donor renal transplantation (LDRT) patients subjected to pre-transplant stem cell transplantation (SCT) to minimize immunosuppression to low-dose steroid monotherapy. METHODS LDRT patients subjected to pretransplant SCT who had stable graft function for ≥2 years and serum creatinine (SCr) <2 mg/dL were recruited. Patients with diabetes, hepatitis C/B, rejections, or unwilling to participate, were excluded. They had been subjected to non-myeloablative conditioning of total lymphoid irradiation (TLI)/bortezomib and cyclophosphamide, rabbit-antithymocyte globulin (r-ATG) and rituximab with SCT. The maintenance immunosuppression consisted of calcineurin inhibitors (CNI) and/or anti-proliferative agents and prednisone. Donor-specific antibodies (DSA) and peripheral T-regulatory cells (CD127(low/-)/4(+)/25(high)) (p-Tregs) were studied before and after withdrawal of major immunosuppressants; graft biopsy was taken after 100 days of withdrawal in willing patients. Rejections were planned to be treated by anti-rejection therapy followed by rescue immunosuppression. RESULTS All immunosuppression but prednisone, 5-10 mg/day has been successfully withdrawn for a mean of 2.2 years in 76 patients with a mean age of 31.4 years and a mean donor-recipient HLA match of 2.9. The mean SCr of 1.4 mg/dL and p-Tregs of 3.5% was remained stable after withdrawal; DSA status was negative in 35.5% and positive in 47.4% patients. Protocol biopsies in all 10 patients who gave the consent were unremarkable. CONCLUSION Stable graft function in LDRT on low-dose steroid monotherapy using pre-transplant SCT under non-myeloablative conditioning with generation of p-Tregs can be achieved successfully and safely.
Collapse
Affiliation(s)
- H. L. Trivedi
- Department of Nephrology and Transplantation Medicine,
| | - A. V. Vanikar
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre and Dr. H. L. Trivedi Institute of Transplantation Sciences (ITS), Asarwa, Ahmedabad-380016, Gujarat, India
| | - V. B. Kute
- Department of Nephrology and Transplantation Medicine,
| | - H. V. Patel
- Department of Nephrology and Transplantation Medicine,
| | - M. R. Gumber
- Department of Nephrology and Transplantation Medicine,
| | - P. R. Shah
- Department of Nephrology and Transplantation Medicine,
| | - S. D. Dave
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre and Dr. H. L. Trivedi Institute of Transplantation Sciences (ITS), Asarwa, Ahmedabad-380016, Gujarat, India
| | - V. B. Trivedi
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre and Dr. H. L. Trivedi Institute of Transplantation Sciences (ITS), Asarwa, Ahmedabad-380016, Gujarat, India
| |
Collapse
|
42
|
Le Gall SM, Legrand J, Benbijja M, Safya H, Benihoud K, Kanellopoulos JM, Bobé P. Loss of P2X7 receptor plasma membrane expression and function in pathogenic B220+ double-negative T lymphocytes of autoimmune MRL/lpr mice. PLoS One 2012; 7:e52161. [PMID: 23284917 PMCID: PMC3528777 DOI: 10.1371/journal.pone.0052161] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/12/2012] [Indexed: 12/12/2022] Open
Abstract
Lupus is a chronic inflammatory autoimmune disease influenced by multiple genetic loci including Fas Ligand (FasL) and P2X7 receptor (P2X7R). The Fas/Fas Ligand apoptotic pathway is critical for immune homeostasis and peripheral tolerance. Normal effector T lymphocytes up-regulate the transmembrane tyrosine phosphatase B220 before undergoing apoptosis. Fas-deficient MRL/lpr mice (lpr mutation) exhibit lupus and lymphoproliferative syndromes due to the massive accumulation of B220+ CD4–CD8– (DN) T lymphocytes. The precise ontogeny of B220+ DN T cells is unknown. B220+ DN T lymphocytes could be derived from effector CD4+ and CD8+ T lymphocytes, which have not undergone activation-induced cell death due to inactivation of Fas, or from a special cell lineage. P2X7R is an extracellular ATP-gated cell membrane receptor involved in the release of proinflammatory cytokines and TNFR1/Fas-independent cell death. P2X7R also regulate early signaling events involved in T-cell activation. We show herein that MRL/lpr mice carry a P2X7R allele, which confers a high sensitivity to ATP. However, during aging, the MRL/lpr T-cell population exhibits a drastically reduced sensitivity to ATP- or NAD-mediated stimulation of P2X7R, which parallels the increase in B220+ DN T-cell numbers in lymphoid organs. Importantly, we found that this B220+ DN T-cell subpopulation has a defect in P2X7R-mediated responses. The few B220+ T cells observed in normal MRL+/+ and C57BL/6 mice are also resistant to ATP or NAD treatment. Unexpectedly, while P2X7R mRNA and proteins are present inside of B220+ T cells, P2X7R are undetectable on the plasma membrane of these T cells. Our results prompt the conclusion that cell surface expression of B220 strongly correlates with the negative regulation of the P2X7R pathway in T cells.
Collapse
Affiliation(s)
- Sylvain M Le Gall
- Institut Jacques Monod, Centre National de la Recherche Scientifique, Université Paris Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
43
|
Autocrine IFNγ controls the regulatory function of lymphoproliferative double negative T cells. PLoS One 2012; 7:e47732. [PMID: 23077665 PMCID: PMC3471870 DOI: 10.1371/journal.pone.0047732] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 09/14/2012] [Indexed: 11/19/2022] Open
Abstract
TCRαβ+ CD4−CD8−NK− double negative T cells (DN T cells) can act as regulatory T cells to inhibit allograft rejection and autoimmunity. Their role in graft-versus-host disease and mechanisms of suppression remain elusive. In this study, we demonstrate that DN T cells can inhibit CD4+ T cell-mediated GVHD in a semi-allogeneic model of bone marrow transplantation. Furthermore, we present evidence of a novel autocrine IFNγ signaling pathway in Fas-deficient C57BL/6.lpr (B6.lpr) DN T cells. B6.lpr DN T cells lacking IFNγ or its receptor were impaired in their ability to suppress syngeneic CD4+ T cells responding to alloantigen stimulation both in vitro and in vivo. Autocrine IFNγ signaling was required for sustained B6.lpr DN T cell IFNγ secretion in vivo and for upregulation of surface Fas ligand expression during TCR stimulation. Fas ligand (FasL) expression by B6.lpr DN T cells permitted lysis of activated CD4+ T cells and was required for suppression of GVHD. Collectively, our data indicate that DN T cells can inhibit GVHD and that IFNγ plays a critical autocrine role in controlling the regulatory function of B6.lpr DN T cells.
Collapse
|
44
|
Hillhouse EE, Lesage S. A comprehensive review of the phenotype and function of antigen-specific immunoregulatory double negative T cells. J Autoimmun 2012; 40:58-65. [PMID: 22910322 DOI: 10.1016/j.jaut.2012.07.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/31/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
Abstract
Double negative T cells that lack the expression of both CD4 and CD8 T cell co-receptors exhibit a most unique antigen-specific immunoregulatory potential first described over a decade ago. Due to their immunoregulatory function, this rare T cell population has been studied in both mice and humans for their contribution to peripheral tolerance and disease prevention. Consequently, double negative cells are gaining interest as a potential cellular therapeutic. Herein, we review the phenotype and function of double negative T cells with emphasis on their capacity to induce antigen-specific immune tolerance. While the phenotypic and functional similarities between double negative T cells identified in mouse and humans are highlighted, we also call attention to the need for a specific marker of double negative T cells, which will facilitate future studies in humans. Altogether, due to their unique properties, double negative T cells present a promising therapeutic potential in the context of various disease settings.
Collapse
Affiliation(s)
- Erin E Hillhouse
- Department of Microbiology and Immunology, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.
| | | |
Collapse
|
45
|
Wood KJ, Bushell A, Hester J. Regulatory immune cells in transplantation. Nat Rev Immunol 2012; 12:417-30. [DOI: 10.1038/nri3227] [Citation(s) in RCA: 334] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
Juvet SC, Zhang L. Double negative regulatory T cells in transplantation and autoimmunity: recent progress and future directions. J Mol Cell Biol 2012; 4:48-58. [PMID: 22294241 DOI: 10.1093/jmcb/mjr043] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
T lymphocytes bearing the αβ T cell receptor (TCR) but lacking CD4, CD8, and markers of natural killer (NK) cell differentiation are known as 'double-negative' (DN) T cells and have been described in both humans and rodent models. We and others have shown that DN T cells can act as regulatory T cells (Tregs) that are able to prevent allograft rejection, graft-versus-host disease, and autoimmune diabetes. In the last few years, new data have revealed evidence of DN Treg function in vivo in rodents and humans. Moreover, significant advances have been made in the mechanisms by which DN Tregs target antigen-specific T cells. One major limitation of the field is the lack of a specific marker that can be used to distinguish truly regulatory DN T cells (DN Tregs) from non-regulatory ones, and this is the central challenge in the coming years. Here, we review recent progress on the role of DN Tregs in transplantation and autoimmunity, and their mechanisms of action. We also provide some perspectives on how DN Tregs compare with Foxp3(+) Tregs.
Collapse
Affiliation(s)
- Stephen C Juvet
- Division of Respirology and Clinician-Scientist Training Program, Department of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
47
|
Su Y, Huang X, Wang S, Min WP, Yin Z, Jevnikar AM, Zhang ZX. Double negative Treg cells promote nonmyeloablative bone marrow chimerism by inducing T-cell clonal deletion and suppressing NK cell function. Eur J Immunol 2012; 42:1216-1225. [PMID: 22539294 DOI: 10.1002/eji.201141808] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The establishment of immune tolerance and prevention of chronic rejection remain major goals in clinical transplantation. In bone marrow (BM) transplantation, T cells and NK cells play important roles for graft rejection. In addition, graft-versus-host-disease (GVHD) remains a major obstacle for BM transplantation. In this study, we aimed to establish mixed chimerism in an irradiation-free condition. Our data indicate that adoptive transfer of donor-derived T-cell receptor (TCR) αβ(+) CD3(+) CD4(-) CD8(-) NK1.1(-) (double negative, DN) Treg cells prior to C57BL/6 to BALB/c BM transplantation, in combination with cyclophosphamide, induced a stable-mixed chimerism and acceptance of C57BL/6 skin allografts but rejection of third-party C3H (H-2k) skin grafts. Adoptive transfer of CD4(+) and CD8(+) T cells, but not DN Treg cells, induced GVHD in this regimen. The recipient T-cell alloreactive responsiveness was reduced in the DN Treg cell-treated group and clonal deletions of TCRVβ2, 7, 8.1/2, and 8.3 were observed in both CD4(+) and CD8(+) T cells. Furthermore, DN Treg-cell treatment suppressed NK cell-mediated BM rejection in a perforin-dependent manner. Taken together, our results suggest that adoptive transfer of DN Treg cells can control both adoptive and innate immunities and promote stable-mixed chimerism and donor-specific tolerance in the irradiation-free regimen.
Collapse
Affiliation(s)
- Ye Su
- The Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
48
|
Ye H, Chang Y, Zhao X, Huang X. Characterization of CD3+CD4-CD8- (double negative) T cells reconstitution in patients following hematopoietic stem-cell transplantation. Transpl Immunol 2011; 25:180-6. [PMID: 21911061 DOI: 10.1016/j.trim.2011.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2011] [Revised: 08/04/2011] [Accepted: 08/21/2011] [Indexed: 11/16/2022]
Abstract
BACKGROUND CD3+CD4-CD8-double negative (DN) T cells, as a distinct subset of regulatory T cells (Tregs), played a pivotal role in patients following hematopoietic stem-cell transplantation. METHODS This study examines the behavior of CD3+CD4-CD8- double negative (DN) T cells in 73 patients at days 30, 60, 90 and 180 after allo-HSCT. RESULTS There was no significant difference in neutrophil and platelet engraftment between the higher and lower absolute counts of 30days DN Tregs (p=0.674, 0.863, respectively). The reconstitution of DN Tregs was significantly slower than that of CD8+, CD4+, and CD3+CD8+CD28- T cells (p<0.001), but significantly faster than that of CD19+ and CD4+CD25+ T cells (p<0.001, p=0.032, respectively). Importantly, in the HLA mismatched group, DN Tregs reconstitution had significant effect on aGVHD (p=0.027) and there was significant correlation between aGVHD and DN Tregs reconstitution (p=0.035). DN Tregs reconstitution was significantly faster in the patients who were devoid of aGVHD than that of patients who developed aGVHD. Furthermore, we compared the absolute value of DN Tregs at 30days, 60days, 90days and 180days after allo-HSCT with grade aGVHD and found an inverse linear relationship in the HLA mismatched group (n=37, P<0.001, r=-0.573). CONCLUSIONS The successful expansion of DN Tregs at 60days after allo-HCST may help avoid severe manifestations of aGVHD in the HLA mismatched group, suggesting that DN Tregs have potential protection effect against aGVHD.
Collapse
Affiliation(s)
- Haige Ye
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | | | | | | |
Collapse
|
49
|
Gao JF, McIntyre MSF, Juvet SC, Diao J, Li X, Vanama RB, Mak TW, Cattral MS, Zhang L. Regulation of antigen-expressing dendritic cells by double negative regulatory T cells. Eur J Immunol 2011; 41:2699-708. [PMID: 21660936 DOI: 10.1002/eji.201141428] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 05/05/2011] [Accepted: 06/01/2011] [Indexed: 12/25/2022]
Abstract
TCRαβ(+) CD3(+) CD4(-) CD8(-) NK1.1(-) double negative (DN) Tregs comprise 1-3% of peripheral T lymphocytes in mice and humans. It has been demonstrated that DN Tregs can suppress allo-, xeno- and auto-immune responses in an Ag-specific fashion. However, the mechanisms by which DN Tregs regulate immune responses remain elusive. Whether DN Tregs can regulate DCs has not been investigated previously. In this study, we demonstrate that DN Tregs express a high level of CTLA4 and are able to down-regulate costimulatory molecules CD80 and CD86 expressed on Ag-expressing mature DCs (mDCs). DN Tregs from CTLA4 KO mice were not able to downregulate CD80 and CD86 expression, indicating that CTLA4 is critical for DN Treg-mediated downregulation of costimulatory molecule expression on Ag-expressing mature DCs. Furthermore, DN Tregs could kill both immature and mature allogeneic DCs, as well as Ag-loaded syngeneic DCs, in an Ag-specific manner in vitro and in vivo, mainly through the Fas-FasL pathway. These data demonstrate, for the first time, that DN Tregs are potent regulators of DCs and may have the potential to be developed as a novel immune suppression treatment.
Collapse
Affiliation(s)
- Julia Fang Gao
- University of Toronto Transplant Institute, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang D, Zhang W, Ng TW, Wang Y, Liu Q, Gorantla V, Lakkis F, Zheng XX. Adoptive cell therapy using antigen-specific CD4⁻CD8⁻T regulatory cells to prevent autoimmune diabetes and promote islet allograft survival in NOD mice. Diabetologia 2011; 54:2082-92. [PMID: 21594554 DOI: 10.1007/s00125-011-2179-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 04/04/2011] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS A new differentiation pathway for CD4(-)CD8(-) (DN) T cells has recently been identified that exhibits the potent function of peripheral converted DN T cells in suppressing immune responses and provides the potential to treat autoimmune diseases. The aim of this study was to determine if the DN T cells converted from CD4(+) T cells of NOD mice retain the antigen-specific regulatory capacity and prevent autoimmune diabetes in vivo. We also sought to determine if the combination of DN T cells with rapamycin promotes islet allograft survival in autoimmune diabetic NOD recipients. METHODS NOD CD4(+) T cells were converted to DN T cells in an in vitro mixed-lymphocyte reaction, with or without GAD65 peptide, as previously reported. The antigen-specific DN T cells were adoptively transferred to NOD/SCID mice, new-onset diabetic NOD mice or islet-allograft-recipient NOD mice as the part of cell-based therapy. The development of diabetes and allograft survival was assessed by monitoring blood glucose levels. RESULTS NOD CD4(+) T cells were converted in vitro to DN T cells at a rate of 50% and expressed unique cell features. The DN T cells from NOD donors blocked autoimmunity and prevented diabetes in NOD models, and these effects were even greater for GAD65-peptide-primed DN T cells. DN T cells acted in conjunction with rapamycin to suppress alloantigen-triggered T cell proliferation, promoted apoptosis and prolonged islet allograft survival in NOD recipients. CONCLUSIONS/INTERPRETATION Administration of the islet beta cell antigen-specific DN T cells can prevent the development of autoimmune diabetes and promote islet allograft survival in NOD mice.
Collapse
Affiliation(s)
- D Zhang
- Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|