1
|
Yue H, Bai L. Progress, implications, and challenges in using humanized immune system mice in CAR-T therapy-Application evaluation and improvement. Animal Model Exp Med 2024; 7:3-11. [PMID: 37823214 PMCID: PMC10961865 DOI: 10.1002/ame2.12353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023] Open
Abstract
In recent years, humanized immune system (HIS) mice have been gradually used as models for preclinical research in pharmacotherapies and cell therapies with major breakthroughs in tumor and other fields, better mimicking the human immune system and the tumor immune microenvironment, compared to traditional immunodeficient mice. To better promote the application of HIS mice in preclinical research, we selectively summarize the current prevalent and breakthrough research and evaluation of chimeric antigen receptor (CAR) -T cells in various antiviral and antitumor treatments. By exploring its application in preclinical research, we find that it can better reflect the actual clinical patient condition, with the advantages of providing high-efficiency detection indicators, even for progressive research and development. We believe that it has better clinical patient simulation and promotion for the updated design of CAR-T cell therapy than directly transplanted immunodeficient mice. The characteristics of the main models are proposed to improve the use defects of the existing models by reducing the limitation of antihost reaction, combining multiple models, and unifying sources and organoid substitution. Strategy study of relapse and toxicity after CAR-T treatment also provides more possibilities for application and development.
Collapse
Affiliation(s)
- Hanwei Yue
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal SciencesCAMS and PUMCChao‐yang District, BeijingChina
| | - Lin Bai
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal SciencesCAMS and PUMCChao‐yang District, BeijingChina
| |
Collapse
|
2
|
Zhou XY, Li JY, Tan JT, HuangLi YL, Nie XC, Xia P. Clinical significance of the CD98hc-CD147 complex in ovarian cancer: a bioinformatics analysis. J OBSTET GYNAECOL 2023; 43:2188085. [PMID: 36930892 DOI: 10.1080/01443615.2023.2188085] [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] [Indexed: 03/19/2023]
Abstract
Ovarian cancer is one of the most common malignant tumours affecting the female reproductive organs. CD147 (BSG) and CD98hc (SLC3A2) are oncogenes that form the CD98hc-CD147 complex, which regulates the proliferation, metastasis, metabolism, and cell cycle of cancer cells. The roles of the CD98hc-CD147 complex in ovarian cancer remain unclear. We analysed the expression and prognostic value of CD147 and CD98hc in ovarian cancer using the TCGA and ICGC databases. The effect of CD147 and CD98hc on the tumour immune response was analysed using the TIMER database. CD98hc was more highly expressed in normal tissues than primary tumour tissues, while CD147 was more highly expressed in primary tumour tissues than normal tissues. CD98hc expression was significantly associated with neutrophil and dendritic cell levels. CD147 and CD98hc were correlated with DNA repair, the cell cycle, and DNA replication. The CD98hc-CD147 complex could serve as a target for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xin-Yue Zhou
- School of Stomatology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Jin-Yao Li
- School of Stomatology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Jing-Tong Tan
- School of Stomatology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Yi-Li HuangLi
- School of Stomatology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Xiao-Cui Nie
- Department of Gynaecology, Shenyang Women's and Children's Hospital, Shenyang, P.R. China
| | - Pu Xia
- Biological Anthropology Institute, College of Basic Medical Science, Jinzhou Medical University, Jinzhou, P.R. China
| |
Collapse
|
3
|
Xia P, Dubrovska A. CD98 heavy chain as a prognostic biomarker and target for cancer treatment. Front Oncol 2023; 13:1251100. [PMID: 37823053 PMCID: PMC10562705 DOI: 10.3389/fonc.2023.1251100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
The SLC3A2 gene encodes for a cell-surface transmembrane protein CD98hc (4F2). CD98hc serves as a chaperone for LAT1 (SLC7A5), LAT2 (SLC7A8), y+LAT1 (SLC7A7), y+LAT2 (SLC7A6), xCT (SLC7A11) and Asc1 (SLC7A10) providing their recruitment to the plasma membrane. Together with the light subunits, it constitutes heterodimeric transmembrane amino acid transporters. CD98hc interacts with other surface molecules, such as extracellular matrix metalloproteinase inducer CD147 (EMMPRIN) and adhesion receptors integrins, and regulates glucose uptake. In this way, CD98hc connects the signaling pathways sustaining cell proliferation and migration, biosynthesis and antioxidant defense, energy production, and stem cell properties. This multifaceted role makes CD98hc one of the critical regulators of tumor growth, therapy resistance, and metastases. Indeed, the high expression levels of CD98hc were confirmed in various tumor tissues, including head and neck squamous cell carcinoma, glioblastoma, colon adenocarcinoma, pancreatic ductal adenocarcinoma, and others. A high expression of CD98hc has been linked to clinical prognosis and response to chemo- and radiotherapy in several types of cancer. In this mini-review, we discuss the physiological functions of CD98hc, its role in regulating tumor stemness, metastases, and therapy resistance, and the clinical significance of CD98hc as a tumor marker and therapeutic target.
Collapse
Affiliation(s)
- Pu Xia
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
4
|
Zhang W, Zhou M, Chen C, Wu S, Wang L, Xia B, Liu J, Ma X, Pan T, Zhang H, Li L, Liu B. Identification of CD98 as a Novel Biomarker for HIV-1 Permissiveness and Latent Infection. mBio 2022; 13:e0249622. [PMID: 36214569 PMCID: PMC9765422 DOI: 10.1128/mbio.02496-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) can integrate viral DNA into host cell chromosomes to establish a long-term stable latent reservoir, which is a major obstacle to cure HIV-1 infection. The characteristics of the HIV-1 latent reservoir have not been fully understood. Here, we identified 126 upregulated plasma membrane proteins in HIV-1 latently infected cells by a label-free liquid chromatography-tandem mass spectrometry analysis. The higher levels of CD98 expression in multiple HIV-1 latently infected cell lines and primary CD4+ T cells compared to uninfected cells were further confirmed by quantitative reverse transcription PCR (RT-qPCR) and flow cytometry analyses. In addition, CD98high CD4+ T cells displayed hyper-permissiveness to HIV-1 infection and possessed distinct immune phenotypic profiles associated with Th17 and peripheral follicular T helper (pTFH) characteristics. Notably, the CD98high resting memory CD4+ T cells harbored significantly higher cell-associated viral RNA and intact provirus than CD98low counterparts in HIV-1-infected individuals receiving combined antiretroviral therapy. Furthermore, CD98high CD4+ T cells exhibited a robust proliferative capacity and significantly contributed to the clonal expansion of the HIV-1 latent reservoir. Our study demonstrates that CD98 can be used as a novel biomarker of HIV-1 latently infected cells to indicate the effect of various strategies to reduce the viral reservoir. IMPORTANCE Identification of cellular biomarkers is the crucial challenge to eradicate the HIV-1 latent reservoir. In our study, we identified CD98 as a novel plasma membrane biomarker for HIV-1 permissiveness and latent infection. Importantly, CD98high CD4+ T cells exhibited a hyper-permissiveness to HIV-1 infection and significantly contributed to the clonal expansion of the HIV-1 latent reservoir. CD98 could be targeted to develop therapeutic strategies to reduce the HIV-1 latent reservoir in further research.
Collapse
Affiliation(s)
- Wanying Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Infectious Diseases Center, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mo Zhou
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cancan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shiyu Wu
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lilin Wang
- Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Baijin Xia
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Liu
- Qianyang Biomedical Research Institute, Guangzhou, Guangdong, China
| | - Xiancai Ma
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ting Pan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linghua Li
- Infectious Diseases Center, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bingfeng Liu
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Tran DT, Sundararaj K, Atkinson C, Nadig SN. T-cell Immunometabolism: Therapeutic Implications in Organ Transplantation. Transplantation 2021; 105:e191-e201. [PMID: 33795597 PMCID: PMC8464628 DOI: 10.1097/tp.0000000000003767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although solid-organ transplantation has evolved steadily with many breakthroughs in the past 110 y, many problems remain to be addressed, and advanced therapeutic strategies need to be considered. T-cell immunometabolism is a rapidly advancing field that has gathered much attention recently, providing ample mechanistic insight from which many novel therapeutic approaches have been developed. Applications from the field include antitumor and antimicrobial therapies, as well as for reversing graft-versus-host disease and autoimmune diseases. However, the immunometabolism of T cells remains underexplored in solid-organ transplantation. In this review, we will highlight key findings from hallmark studies centered around various metabolic modes preferred by different T-cell subtypes (categorized into naive, effector, regulatory, and memory T cells), including glycolysis, glutaminolysis, oxidative phosphorylation, fatty acid synthesis, and oxidation. This review will discuss the underlying cellular signaling components that affect these processes, including the transcription factors myelocytomatosis oncogene, hypoxia-inducible factor 1-alpha, estrogen-related receptor alpha, and sterol regulatory element-binding proteins, along with the mechanistic target of rapamycin and adenosine monophosphate-activated protein kinase signaling. We will also explore potential therapeutic strategies targeting these pathways, as applied to the potential for tolerance induction in solid-organ transplantation.
Collapse
Affiliation(s)
- Danh T. Tran
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
| | - Kamala Sundararaj
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Carl Atkinson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Satish N. Nadig
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
6
|
Zhao J, Que W, Du X, Fujino M, Ichimaru N, Ueta H, Tokuda N, Guo WZ, Zabrocki P, de Haard H, Nonomura N, Li XK. Monotherapy With Anti-CD70 Antibody Causes Long-Term Mouse Cardiac Allograft Acceptance With Induction of Tolerogenic Dendritic Cells. Front Immunol 2021; 11:555996. [PMID: 33737923 PMCID: PMC7961176 DOI: 10.3389/fimmu.2020.555996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/31/2020] [Indexed: 01/02/2023] Open
Abstract
Allograft rejection has been an obstacle for the long-term survival of patients. CD70, a tumor necrosis factor (TNF) family member critically expressed on antigen-presenting cells and strongly but transiently up-regulated during lymphocyte activation, represents an important co-stimulatory molecule that induces effective T cell responses. We used a mouse heterotopic cardiac transplantation model to evaluate the effects of monotherapy with the antibody targeting mouse CD70 (FR70) on transplantation tolerance and its immunoregulatory activity. FR70-treated C3H recipient mice permanently accepted B6 fully mismatched cardiac allografts. Consistent with the graft survival, the infiltration of CD8+ T cells in the graft was reduced, dendritic cells were differentiated into a tolerogenic status, and the number of regulatory T cells was elevated both in the graft and the recipient’s spleen. In addition, naïve C3H given an adoptive transfer of spleen cells from the primary recipients with FR70 treatment accepted a heart graft from a matching B6 donor but not third-party BALB/c mice. Our findings show that treatment with FR70 induced regulatory cells and inhibited cytotoxic T cell proliferation, which led to long-term acceptance of mouse cardiac allografts. These findings highlight the potential role of anti-CD70 antibodies as a clinically effective treatment for allograft rejection.
Collapse
Affiliation(s)
- Jing Zhao
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan.,Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weitao Que
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Xiaoxiao Du
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naotsugu Ichimaru
- Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisashi Ueta
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Nobuko Tokuda
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | - Norio Nonomura
- Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Ikeda K, Kinoshita M, Kayama H, Nagamori S, Kongpracha P, Umemoto E, Okumura R, Kurakawa T, Murakami M, Mikami N, Shintani Y, Ueno S, Andou A, Ito M, Tsumura H, Yasutomo K, Ozono K, Takashima S, Sakaguchi S, Kanai Y, Takeda K. Slc3a2 Mediates Branched-Chain Amino-Acid-Dependent Maintenance of Regulatory T Cells. Cell Rep 2017; 21:1824-1838. [DOI: 10.1016/j.celrep.2017.10.082] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 08/06/2017] [Accepted: 10/20/2017] [Indexed: 12/23/2022] Open
|
8
|
Cai S, Hou J, Fujino M, Zhang Q, Ichimaru N, Takahara S, Araki R, Lu L, Chen JM, Zhuang J, Zhu P, Li XK. iPSC-Derived Regulatory Dendritic Cells Inhibit Allograft Rejection by Generating Alloantigen-Specific Regulatory T Cells. Stem Cell Reports 2017; 8:1174-1189. [PMID: 28434942 PMCID: PMC5425686 DOI: 10.1016/j.stemcr.2017.03.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023] Open
Abstract
Regulatory dendritic cell (DCregs)-based immunotherapy is a potential therapeutic tool for transplant rejection. We generated DCregs from murine induced pluripotent stem cells (iPSCs), which could remain in a “stable immature stage” even under strong stimulation. Harnessing this characteristic, we hypothesized that iPS-DCregs worked as a negative vaccine to generate regulatory T cells (Tregs), and induced donor-specific allograft acceptance. We immunized naive CBA (H-2Kk) mice with B6 (H-2Kb) iPS-DCregs and found that Tregs (CD4+CD25+FOXP3+) significantly increased in CBA splenocytes. Moreover, immunized CBA recipients permanently accepted B6 cardiac grafts in a donor-specific pattern. We demonstrated mechanistically that donor-type iPS-DCregs triggered transforming growth factor β1 secretion, under which the donor-antigen peptides directed naive CD4+ T cells to differentiate into donor-specific FOXP3+ Tregs instead of into effector T cells in vivo. These findings highlight the potential of iPS-DCregs as a key cell therapy resource in clinical transplantation. iPS-DCregs keep in stable immature stage that makes them a powerful cellular vaccine Donor-type iPS-DCregs lead to permanent acceptance of allogeneic cardiac grafts iPS-DCregs reduce CTL and downregulate proinflammatory cytokine iPS-DCregs enhance Tregs transmigration capability in a TGF-β1-dependent manner
Collapse
Affiliation(s)
- Songjie Cai
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Jiangang Hou
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Qi Zhang
- Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Naotsugu Ichimaru
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shiro Takahara
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Ryoko Araki
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ji-Mei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ping Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
9
|
Ren W, Liu G, Yin J, Tan B, Wu G, Bazer FW, Peng Y, Yin Y. Amino-acid transporters in T-cell activation and differentiation. Cell Death Dis 2017; 8:e2655. [PMID: 28252650 PMCID: PMC5386510 DOI: 10.1038/cddis.2016.222] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/25/2022]
Abstract
T-cell-mediated immune responses aim to protect mammals against cancers and infections, and are also involved in the pathogenesis of various inflammatory or autoimmune diseases. Cellular uptake and the utilization of nutrients is closely related to the T-cell fate decision and function. Research in this area has yielded surprising findings in the importance of amino-acid transporters for T-cell development, homeostasis, activation, differentiation and memory. In this review, we present current information on amino-acid transporters, such as LAT1 (l-leucine transporter), ASCT2 (l-glutamine transporter) and GAT-1 (γ-aminobutyric acid transporter-1), which are critically important for mediating peripheral naive T-cell homeostasis, activation and differentiation, especially for Th1 and Th17 cells, and even memory T cells. Mechanically, the influence of amino-acid transporters on T-cell fate decision may largely depend on the mechanistic target of rapamycin complex 1 (mTORC1) signaling. These discoveries remarkably demonstrate the role of amino-acid transporters in T-cell fate determination, and strongly indicate that manipulation of the amino-acid transporter-mTORC1 axis could ameliorate many inflammatory or autoimmune diseases associated with T-cell-based immune responses.
Collapse
Affiliation(s)
- Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.,University of the Chinese Academy of Sciences, Beijing 10008, China
| | - Gang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843-2471, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX 77843-2471, USA
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage and Herbivore, College of Animal Science and Technology, Southwest University, Chongqing 400716, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Observation and Experiment Station of Animal Nutrition and Feed Science in South-Central China, Ministry of Agriculture; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China
| |
Collapse
|
10
|
Fujino M, Zhu P, Cai S, Nishio Y, Zhuang J, Li XK. MicroRNAs Involved in Acute Rejection and Tolerance in Murine Cardiac Allografts. EXP CLIN TRANSPLANT 2016; 14:424-30. [PMID: 27227980 DOI: 10.6002/ect.2015.0251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Induction of immunologic tolerance is the ultimate goal of organ transplant. To investigate the involvement of microRNA in tolerance induction after organ transplant, murine cardiac allografts were performed and the expression of microRNA in the grafts was analyzed. MATERIALS AND METHODS Cardiac allografts were performed using C57BL/10 (H2-Kb) to CBA/N (H2-Kk) fully mismatched combination with or without eicosapentaenoic acid for tolerance induction. Ten microRNA, mir-146a, 15b, 223, 23a, 27a, 34a, 451, 101a, 101b, 148a, discovered in hepatic grafts were examined by quantitative reverse transcription polymerase chain reaction using RNA from the cardiac allografts. RESULTS The administration of eicosapentaenoic acid markedly prolonged the cardiac allograft survival (median survival time > 100 days) and decreased the pathological score. Quantitative reverse transcription polymerase chain reaction revealed that mir-223 was up-regulated in accordance with pathological deterioration as compared with the expression observed in the syngeneic grafts. In contrast, the other microRNA was down-regulated. Pearson product moment correlation analysis demonstrated that the expression patterns of mir-223 and mir-146a had high or moderate positive associations between the cardiac and haptic allografts in mice. CONCLUSIONS The change in the microRNA expression in the allografts suggests that microRNA plays a role in the induction and/or maintenance of tolerance after allograft transplant. Our findings suggest that mir-223 may be associated with rejection while mir-146a, -15b, -23a, -27a, -34a, -451, -101a, -101b, -148a may be involved in tolerance. A superior grasp of the mechanism for rejection and tolerance observed in the murine heart allotransplant model may provide a better curative treatment strategy to mitigate allograft rejection.
Collapse
Affiliation(s)
- Masayuki Fujino
- rom the Division of Transplantation Immunology National Research Institute for Child Health and Development; and the AIDS Research Center National Institute of Infectious Diseases Tokyo Japan
| | | | | | | | | | | |
Collapse
|
11
|
Nishio Y, Fujino M, Cai S, Kitajima Y, Saito T, Tsumura H, Ito M, Ito Y, Nagahara Y, Li XK. Impaired CD98 signaling protects against graft-versus-host disease by increasing regulatory T cells. Transpl Immunol 2016; 35:34-9. [PMID: 26836475 DOI: 10.1016/j.trim.2016.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 01/08/2023]
Abstract
Graft-versus-host disease (GvHD) is a major barrier to the broader use of allogenic hematopoietic stem cell transplantation for non-malignant clinical applications. A murine model of C57BL/6 to B6D2F1 acute GvHD was employed with T lymphocytes harboring a deletion of the CD98 heavy chain (CD98hc(-/-)) as donor cells. The CD98hc(-/-) resulted in lower responses to alloantigen stimulation in a mixed leukocyte reaction assay, and prevented the mortality associated with disease progression. The percentage of donor CD8 T lymphocytes was significantly decreased, while the percentage of Foxp3-positive regulatory T cells (Tregs) in recipients was increased by CD98hc(-/-). Decreased expression of FAS, FASL, ICOS, ICOSL, PD-1 and PD-L1 by donor CD8 T cells, and mRNA expression of cytotoxic T cell-related cytokines in the recipients were shown in those with CD98hc(-/-). Fewer infiltrated cells are found in the lungs, liver, tongue and skin of recipients with CD98hc(-/-) compared with the wild type recipients. Taken together, our data indicate that T cell-specific deletion of CD98hc can contribute to the prevention of GvHD development due to the attenuation of lymphocyte migration and by increasing the generation of Treg cells. These findings are expected to make it possible to develop novel approaches for the prevention of GvHD.
Collapse
Affiliation(s)
- Yoshiaki Nishio
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Songjie Cai
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yuya Kitajima
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Division of Life Science and Engineering, Tokyo Denki University, Saitama, Japan
| | - Taro Saito
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Division of Life Science and Engineering, Tokyo Denki University, Saitama, Japan
| | - Hideki Tsumura
- Division of Laboratory Animal Resources, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Morihiro Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Yasuhiko Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Yukitoshi Nagahara
- Division of Life Science and Engineering, Tokyo Denki University, Saitama, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
12
|
Conditional deletion of CD98hc inhibits osteoclast development. Biochem Biophys Rep 2015; 5:203-210. [PMID: 28955825 PMCID: PMC5600448 DOI: 10.1016/j.bbrep.2015.11.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 10/31/2015] [Accepted: 11/30/2015] [Indexed: 11/22/2022] Open
Abstract
The CD98 heavy chain (CD98hc) regulates virus-induced cell fusion and monocyte fusion, and is involved in amino acid transportation. Here, we examined the role that CD98hc plays in the formation of osteoclasts using CD98hcflox/floxLysM-cre peritoneal macrophages (CD98hc-defect macrophages). Peritoneal macrophages were stimulated with co-cultured with osteoblasts in the presence of 1,25(OH)2 vitamin D3, and thereafter stained with tartrate-resistant acid phosphatase staining solution. The multinucleated osteoclast formation was severely impaired in the peritoneal macrophages isolated from the CD98hc-defect mice compared with those from wild-type mice. CD98hc mediates integrin signaling and amino acid transport through the CD98 light chain (CD98lc). In integrin signaling, suppression of the M-CSF-RANKL-induced phosphorylation of ERK, Akt, JNK and p130Cas were observed at the triggering phase in the CD98h-defect peritoneal macrophages. Moreover, we showed that the general control non-derepressible (GCN) pathway, which was activated by amino acid starvation, was induced by the CD98hc-defect peritoneal macrophages stimulated with RANKL. These results indicate that CD98 plays two important roles in osteoclast formation through integrin signaling and amino acid transport. The osteoclastogenesis was severely impaired in the CD98hc-defect macrophages. CD98hc-defect peritoneal macrophages fall into amino acid starvation, resulting in inducing the general control non-derepressible (GCN) pathway in the osteoclastogenesis.
Collapse
|
13
|
Kurihara T, Arimochi H, Bhuyan ZA, Ishifune C, Tsumura H, Ito M, Ito Y, Kitamura A, Maekawa Y, Yasutomo K. CD98 Heavy Chain Is a Potent Positive Regulator of CD4+ T Cell Proliferation and Interferon-γ Production In Vivo. PLoS One 2015; 10:e0139692. [PMID: 26444422 PMCID: PMC4596652 DOI: 10.1371/journal.pone.0139692] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 09/16/2015] [Indexed: 12/24/2022] Open
Abstract
Upon their recognition of antigens presented by the MHC, T cell proliferation is vital for clonal expansion and the acquisition of effector functions, which are essential for mounting adaptive immune responses. The CD98 heavy chain (CD98hc, Slc3a2) plays a crucial role in the proliferation of both CD4+ and CD8+ T cells, although it is unclear if CD98hc directly regulates the T cell effector functions that are not linked with T cell proliferation in vivo. Here, we demonstrate that CD98hc is required for both CD4+ T cell proliferation and Th1 functional differentiation. T cell-specific deletion of CD98hc did not affect T cell development in the thymus. CD98hc-deficient CD4+ T cells proliferated in vivo more slowly as compared with control T cells. C57BL/6 mice lacking CD98hc in their CD4+ T cells could not control Leishmania major infections due to lowered IFN-γ production, even with massive CD4+ T cell proliferation. CD98hc-deficient CD4+ T cells exhibited lower IFN-γ production compared with wild-type T cells, even when comparing IFN-γ expression in cells that underwent the same number of cell divisions. Therefore, these data indicate that CD98hc is required for CD4+ T cell expansion and functional Th1 differentiation in vivo, and suggest that CD98hc might be a good target for treating Th1-mediated immune disorders.
Collapse
Affiliation(s)
- Takeshi Kurihara
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Hideki Arimochi
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Zaied Ahmed Bhuyan
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Chieko Ishifune
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Hideki Tsumura
- Division of Laboratory Animal Resources, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Morihiro Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Yasuhiko Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Akiko Kitamura
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Yoichi Maekawa
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Koji Yasutomo
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima, Japan
- * E-mail:
| |
Collapse
|
14
|
Zhang Q, Ichimaru N, Higuchi S, Cai S, Hou J, Fujino M, Nonomura N, Kobayashi M, Ando H, Uno A, Sakurai K, Mochizuki S, Adachi Y, Ohno N, Zou H, Xu J, Li XK, Takahara S. Permanent acceptance of mouse cardiac allografts with CD40 siRNA to induce regulatory myeloid cells by use of a novel polysaccharide siRNA delivery system. Gene Ther 2015; 22:217-26. [DOI: 10.1038/gt.2014.119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 11/06/2014] [Accepted: 11/17/2014] [Indexed: 01/27/2023]
|
15
|
5-Aminolevulinic acid with ferrous iron induces permanent cardiac allograft acceptance in mice via induction of regulatory cells. J Heart Lung Transplant 2014; 34:254-63. [PMID: 25455753 DOI: 10.1016/j.healun.2014.09.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 08/22/2014] [Accepted: 09/24/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND 5-Aminolevulinic acid (5-ALA), a precursor of heme biosynthesis, plays a fundamentally important role in aerobic energy metabolism. Heme oxygenase (HO)-1 cleaves heme to form biliverdin, carbon monoxide (CO) and iron (Fe(2+)). The anti-inflammatory properties of biliverdin and CO help to alleviate ischemia/reperfusion injury as well as acute and/or chronic allograft rejection. We investigated whether 5-ALA and Fe(2+) exerts salutary effects in the setting of organ transplantation. METHODS An in vitro mixed-lymphocyte reaction (MLR) assay and cardiac allotransplantation model (CBA to C57BL/10) were used to evaluate the effects of 5-ALA and Fe(2+) on transplantation tolerance. RESULTS Treatment with 5-ALA and sodium ferrous citrate (SFC) resulted in permanent acceptance in the murine cardiac allografts in a dose-, SFC- and HO-1-dependent manner. The number of graft-infiltrating CD8 T cells was lower and the survival response of recipient spleen T cells to donor-type alloantigens was less compared with control recipients; however, numbers of both regulatory T cells and dendritic cells were significantly increased in 5-ALA/SFC-treated recipients. CONCLUSIONS Our findings show that 5-ALA/SFC inhibits T-cell proliferation in response to alloantigens and an increased number of regulatory cells, resulting in permanent cardiac allograft acceptance in mice. These findings highlight the major roles of CO and/or HO-1 in inducing tolerance and suggest that 5-ALA/SFC may be a clinically effective treatment for allograft rejection.
Collapse
|
16
|
Bhuyan ZA, Arimochi H, Nishida J, Kataoka K, Kurihara T, Ishifune C, Tsumura H, Ito M, Ito Y, Kitamura A, Yasutomo K. CD98hc regulates the development of experimental colitis by controlling effector and regulatory CD4(+) T cells. Biochem Biophys Res Commun 2014; 444:628-33. [PMID: 24491544 DOI: 10.1016/j.bbrc.2014.01.144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/24/2014] [Indexed: 01/06/2023]
Abstract
CD4(+) T cell activation is controlled by signaling through the T cell receptor in addition to various co-receptors, and is also affected by their interactions with effector and regulatory T cells in the microenvironment. Inflammatory bowel diseases (IBD) are caused by the persistent activation and expansion of auto-aggressive CD4(+) T cells that attack intestinal epithelial cells. However, the molecular basis for the persistent activation of CD4(+) T cells in IBD remains unclear. In this study, we investigated how the CD98 heavy chain (CD98hc, Slc3a2) affected the development of colitis in an experimental animal model. Transferring CD98hc-deficient CD4(+)CD25(-) T cells into Rag2(-/-) mice did not cause colitis accompanied by increasing Foxp3(+) inducible regulatory T cells. By comparison, CD98hc-deficient naturally occurring regulatory T cells (nTregs) had a decreased capability to suppress colitis induced by CD4(+)CD25(-) T cells, although CD98hc-deficient mice did not have a defect in the development of nTregs. Blocking CD98hc with an anti-CD98 blocking antibody prevented the development of colitis. Our results indicate that CD98hc regulates the expansion of autoimmune CD4(+) T cells in addition to controlling nTregs functions, which suggests the CD98hc as an important target molecule for establishing strategies for treating colitis.
Collapse
Affiliation(s)
- Zaied Ahmed Bhuyan
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Hideki Arimochi
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Jun Nishida
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Keiko Kataoka
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Takeshi Kurihara
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Chieko Ishifune
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Hideki Tsumura
- Division of Laboratory Animal Resources, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Morihiro Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Yasuhiko Ito
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Akiko Kitamura
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Koji Yasutomo
- Department of Immunology & Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan.
| |
Collapse
|
17
|
Sinclair LV, Rolf J, Emslie E, Shi YB, Taylor PM, Cantrell DA. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat Immunol 2013; 14:500-8. [PMID: 23525088 PMCID: PMC3672957 DOI: 10.1038/ni.2556] [Citation(s) in RCA: 656] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/24/2013] [Indexed: 12/13/2022]
Abstract
T lymphocytes must regulate nutrient uptake to meet the metabolic demands of an immune response. Here we show that the intracellular supply of large neutral amino acids (LNAAs) in T cells was regulated by pathogens and the T cell antigen receptor (TCR). T cells responded to antigen by upregulating expression of many amino-acid transporters, but a single System L ('leucine-preferring system') transporter, Slc7a5, mediated uptake of LNAAs in activated T cells. Slc7a5-null T cells were unable to metabolically reprogram in response to antigen and did not undergo clonal expansion or effector differentiation. The metabolic catastrophe caused by loss of Slc7a5 reflected the requirement for sustained uptake of the LNAA leucine for activation of the serine-threonine kinase complex mTORC1 and for expression of the transcription factor c-Myc. Control of expression of the System L transporter by pathogens is thus a critical metabolic checkpoint for T cells.
Collapse
Affiliation(s)
- Linda V Sinclair
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | | | | | | | | | | |
Collapse
|