1
|
Omar I, Alakhras A, Mutwali S, Bakhiet M. Molecular insights into T cell development, activation and signal transduction (Review). Biomed Rep 2025; 22:94. [PMID: 40247929 PMCID: PMC12001230 DOI: 10.3892/br.2025.1972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
T cell modulation plays a fundamental role to adaptive and innate immunity, which aids the recognition and defense against pathogens while also maintaining self-tolerance. Numerous molecular pathways participate in this process including thymic selection, T cell receptor and antigen-presenting cells cross linkage, along with co-stimulatory signaling cascades. The present review demonstrates a holistic analysis of various classic and novel mechanisms that govern T cell regulation and emerging therapeutic applications. Recent advancements have introduced novel roles in the journey of T cell modulation that can have a pivotal impact on the understanding of this process; for example, phase separation of the linker for activation of T cells, and the newer application of chimeric antigen receptor (CAR) T cell therapy in autoimmune diseases. While discoveries of proximal and distal signal transduction pathways have contributed to the comprehension of T cell anergy, cytokine-mediated differentiation and the delicate balance between immune activation and tolerance, there are still unresolved debates about further molecular mechanisms. There are also still questions about the long-term side effects of CAR-T cell therapy. Deeper research and analysis are required to further aid the understanding and use of this novel therapeutic approach.
Collapse
Affiliation(s)
- Isra Omar
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
- Department of Clinical Medicine, College of Medicine, University of Medical Sciences and Technology, 3523 Kigali, Rwanda
- Royal College of Physicians of Ireland, Dublin D02 E434, Ireland
| | - Ahmed Alakhras
- Department of Clinical Medicine, College of Medicine, Almaarefa University, 11597 Riyadh, Kingdom of Saudi Arabia
| | - Samahir Mutwali
- TeleGeriatric Research Fellowship Program, Michigan State University, MI 48824, USA
| | - Moiz Bakhiet
- Department of Molecular Medicine and Medical Science, Arabian Gulf University, Manama 328, Kingdom of Bahrain
| |
Collapse
|
2
|
Kureshi R, Bello E, Kureshi CT, Walsh MJ, Lippert V, Hoffman MT, Dougan M, Longmire T, Wichroski M, Dougan SK. DGKα/ζ inhibition lowers the TCR affinity threshold and potentiates antitumor immunity. SCIENCE ADVANCES 2023; 9:eadk1853. [PMID: 38000024 PMCID: PMC10672170 DOI: 10.1126/sciadv.adk1853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023]
Abstract
Diacylglycerol kinases (DGKs) attenuate diacylglycerol (DAG) signaling by converting DAG to phosphatidic acid, thereby suppressing pathways downstream of T cell receptor signaling. Using a dual DGKα/ζ inhibitor (DGKi), tumor-specific CD8 T cells with different affinities (TRP1high and TRP1low), and altered peptide ligands, we demonstrate that inhibition of DGKα/ζ can lower the signaling threshold for T cell priming. TRP1high and TRP1low CD8 T cells produced more effector cytokines in the presence of cognate antigen and DGKi. Effector TRP1high- and TRP1low-mediated cytolysis of tumor cells with low antigen load required antigen recognition, was mediated by interferon-γ, and augmented by DGKi. Adoptive T cell transfer into mice bearing pancreatic or melanoma tumors synergized with single-agent DGKi or DGKi and antiprogrammed cell death protein 1 (PD-1), with increased expansion of low-affinity T cells and increased cytokine production observed in tumors of treated mice. Collectively, our findings highlight DGKα/ζ as therapeutic targets for augmenting tumor-specific CD8 T cell function.
Collapse
Affiliation(s)
- Rakeeb Kureshi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elisa Bello
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Courtney T.S. Kureshi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael J. Walsh
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Victoria Lippert
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Megan T. Hoffman
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Dougan
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Stephanie K. Dougan
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
3
|
Shorer O, Yizhak K. Metabolic predictors of response to immune checkpoint blockade therapy. iScience 2023; 26:108188. [PMID: 37965137 PMCID: PMC10641254 DOI: 10.1016/j.isci.2023.108188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/23/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
Metabolism of immune cells in the tumor microenvironment (TME) plays a critical role in cancer patient response to immune checkpoint inhibitors (ICI). Yet, a metabolic characterization of immune cells in the TME of patients treated with ICI is lacking. To bridge this gap we performed a semi-supervised analysis of ∼1700 metabolic genes using single-cell RNA-seq data of > 1 million immune cells from ∼230 samples of cancer patients treated with ICI. When clustering cells based on their metabolic gene expression, we found that similar immunological cellular states are found in different metabolic states. Most importantly, we found metabolic states that are significantly associated with patient response. We then built a metabolic predictor based on a dozen gene signature, which significantly differentiates between responding and non-responding patients across different cancer types (AUC = 0.8-0.92). Taken together, our results demonstrate the power of metabolism in predicting patient response to ICI.
Collapse
Affiliation(s)
- Ofir Shorer
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Keren Yizhak
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- The Taub Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
4
|
Renauer P, Park JJ, Bai M, Acosta A, Lee WH, Lin GH, Zhang Y, Dai X, Wang G, Errami Y, Wu T, Clark P, Ye L, Yang Q, Chen S. Immunogenetic Metabolomics Reveals Key Enzymes That Modulate CAR T-cell Metabolism and Function. Cancer Immunol Res 2023; 11:1068-1084. [PMID: 37253111 PMCID: PMC10527769 DOI: 10.1158/2326-6066.cir-22-0565] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/26/2023] [Accepted: 05/23/2023] [Indexed: 06/01/2023]
Abstract
Immune evasion is a critical step of cancer progression that remains a major obstacle for current T cell-based immunotherapies. Hence, we investigated whether it is possible to genetically reprogram T cells to exploit a common tumor-intrinsic evasion mechanism whereby cancer cells suppress T-cell function by generating a metabolically unfavorable tumor microenvironment (TME). In an in silico screen, we identified ADA and PDK1 as metabolic regulators. We then showed that overexpression (OE) of these genes enhanced the cytolysis of CD19-specific chimeric antigen receptor (CAR) T cells against cognate leukemia cells, and conversely, ADA or PDK1 deficiency dampened this effect. ADA-OE in CAR T cells improved cancer cytolysis under high concentrations of adenosine, the ADA substrate, and an immunosuppressive metabolite in the TME. High-throughput transcriptomics and metabolomics analysis of these CAR T cells revealed alterations of global gene expression and metabolic signatures in both ADA- and PDK1-engineered CAR T cells. Functional and immunologic analyses demonstrated that ADA-OE increased proliferation and decreased exhaustion in CD19-specific and HER2-specific CAR T cells. ADA-OE improved tumor infiltration and clearance by HER2-specific CAR T cells in an in vivo colorectal cancer model. Collectively, these data unveil systematic knowledge of metabolic reprogramming directly in CAR T cells and reveal potential targets for improving CAR T-cell therapy.
Collapse
Affiliation(s)
- Paul Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, Connecticut, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut, USA
| | - Jonathan J. Park
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut, USA
- M.D.-Ph.D. Program, Yale University, West Haven, Connecticut, USA
| | - Meizhu Bai
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
| | - Arianny Acosta
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Won-Ho Lee
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Guang Han Lin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Yueqi Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
| | - Xiaoyun Dai
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
| | - Guangchuan Wang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Present Address: Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Youssef Errami
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Present Address: Tulane University, New Orleans, LA, USA
| | - Terence Wu
- West Campus Analytical Core, Mass Spectrometry/Proteomics Facility, West Haven, Connecticut, USA
| | - Paul Clark
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
| | - Lupeng Ye
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Present Address: Nanjing University, Nanjing, Jiangsu, China
| | - Quanjun Yang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Present Address: Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Center for Cancer Systems Biology, Yale University, West Haven, Connecticut, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, Connecticut, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut, USA
- M.D.-Ph.D. Program, Yale University, West Haven, Connecticut, USA
- Immunobiology Program, Yale University, New Haven, Connecticut, USA
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, USA
- Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Biomedical Data Science, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Wu-Tsai Center, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Kudek MR, Xin G, Alson D(T, Holzhauer S, Shen J, Kasmani MY, Riese M, Cui W. Lymphocytic Choriomeningitis Virus Clone 13 Infection Results in CD8 T Cell-Mediated Host Mortality in Diacylglycerol Kinase α-Deficient Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1281-1291. [PMID: 36920384 PMCID: PMC10121876 DOI: 10.4049/jimmunol.2101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
Diacylglycerol is a potent element of intracellular secondary signaling cascades whose production is enhanced by cell-surface receptor agonism and function is regulated by enzymatic degradation by diacylglycerol kinases (DGKs). In T cells, stringent regulation of the activity of this second messenger maintains an appropriate balance between effector function and anergy. In this article, we demonstrate that DGKα is an indispensable regulator of TCR-mediated activation of CD8 T cells in lymphocytic choriomeningitis virus Clone 13 viral infection. In the absence of DGKα, Clone 13 infection in a murine model results in a pathologic, proinflammatory state and a multicellular immunopathologic host death that is predominantly driven by CD8 effector T cells.
Collapse
Affiliation(s)
- Matthew R. Kudek
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and BMT. Medical College of Wisconsin, Milwaukee, WI, USA
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Gang Xin
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Current address: Department of Microbial Infection and Immunity. Ohio State University, Columbus, OH, USA
| | | | | | - Jian Shen
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Microbiology and Immunology. Medical College of Wisconsin, Milwaukee, WI USA
| | - Moujtaba Y. Kasmani
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Microbiology and Immunology. Medical College of Wisconsin, Milwaukee, WI USA
| | - Matthew Riese
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Medicine, Division of Oncology. Medical College of Wisconsin, Milwaukee, WI USA
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Microbiology and Immunology. Medical College of Wisconsin, Milwaukee, WI USA
| |
Collapse
|
6
|
Harabuchi S, Khan O, Bassiri H, Yoshida T, Okada Y, Takizawa M, Ikeda O, Katada A, Kambayashi T. Manipulation of diacylglycerol and ERK-mediated signaling differentially controls CD8 + T cell responses during chronic viral infection. Front Immunol 2022; 13:1032113. [PMID: 36846018 PMCID: PMC9951774 DOI: 10.3389/fimmu.2022.1032113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Activation of T cell receptor (TCR) signaling is critical for clonal expansion of CD8+ T cells. However, the effects of augmenting TCR signaling during chronic antigen exposure is less understood. Here, we investigated the role of diacylglycerol (DAG)-mediated signaling downstream of the TCR during chronic lymphocytic choriomeningitis virus clone 13 (LCMV CL13) infection by blocking DAG kinase zeta (DGKζ), a negative regulator of DAG. Methods We examined the activation, survival, expansion, and phenotype of virus-specific T cell in the acute and chronic phases of LCMV CL13-infected in mice after DGKζ blockade or selective activation of ERK. Results Upon LCMV CL13 infection, DGKζ deficiency promoted early short-lived effector cell (SLEC) differentiation of LCMV-specific CD8+ T cells, but this was followed by abrupt cell death. Short-term inhibition of DGKζ with ASP1570, a DGKζ-selective pharmacological inhibitor, augmented CD8+ T cell activation without causing cell death, which reduced virus titers both in the acute and chronic phases of LCMV CL13 infection. Unexpectedly, the selective enhancement of ERK, one key signaling pathway downstream of DAG, lowered viral titers and promoted expansion, survival, and a memory phenotype of LCMV-specific CD8+ T cells in the acute phase with fewer exhausted T cells in the chronic phase. The difference seen between DGKζ deficiency and selective ERK enhancement could be potentially explained by the activation of the AKT/mTOR pathway by DGKζ deficiency, since the mTOR inhibitor rapamycin rescued the abrupt cell death seen in virus-specific DGKζ KO CD8+ T cells. Discussion Thus, while ERK is downstream of DAG signaling, the two pathways lead to distinct outcomes in the context of chronic CD8+ T cell activation, whereby DAG promotes SLEC differentiation and ERK promotes a memory phenotype.
Collapse
Affiliation(s)
- Shohei Harabuchi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Omar Khan
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Taku Yoshida
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Yohei Okada
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Masaomi Takizawa
- Research Program Management-Applied Research Management, Astellas Pharma Inc., Tokyo, Japan
| | - Osamu Ikeda
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | - Akihiro Katada
- Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Lim SA, Su W, Chapman NM, Chi H. Lipid metabolism in T cell signaling and function. Nat Chem Biol 2022; 18:470-481. [PMID: 35484263 PMCID: PMC11103273 DOI: 10.1038/s41589-022-01017-3] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/17/2022] [Indexed: 12/19/2022]
Abstract
T cells orchestrate adaptive immunity against pathogens and other immune challenges, but their dysfunction can also mediate the pathogenesis of cancer and autoimmunity. Metabolic adaptation in response to immunological and microenvironmental signals contributes to T cell function and fate decision. Lipid metabolism has emerged as a key regulator of T cell responses, with selective lipid metabolites serving as metabolic rheostats to integrate environmental cues and interplay with intracellular signaling processes. Here, we discuss how extracellular, de novo synthesized and membrane lipids orchestrate T cell biology. We also describe the roles of lipids as regulators of intracellular signaling at the levels of transcriptional, epigenetic and post-translational regulation in T cells. Finally, we summarize therapeutic targeting of lipid metabolism and signaling, and conclude with a discussion of important future directions. Understanding the molecular and functional interplay between lipid metabolism and T cell biology will ultimately inform therapeutic intervention for human disease.
Collapse
Affiliation(s)
- Seon Ah Lim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wei Su
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
8
|
Li S, Zhao F, Ye J, Li K, Wang Q, Du Z, Yue Q, Wang S, Wu Q, Chen H. Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19. Med Microbiol Immunol 2022; 211:49-69. [PMID: 35022857 PMCID: PMC8755516 DOI: 10.1007/s00430-021-00727-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023]
Abstract
Metabolic pathways drive cellular behavior. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes lung tissue damage directly by targeting cells or indirectly by producing inflammatory cytokines. However, whether functional alterations are related to metabolic changes in lung cells after SARS-CoV-2 infection remains unknown. Here, we analyzed the lung single-nucleus RNA-sequencing (snRNA-seq) data of several deceased COVID-19 patients and focused on changes in transcripts associated with cellular metabolism. We observed upregulated glycolysis and oxidative phosphorylation in alveolar type 2 progenitor cells, which may block alveolar epithelial differentiation and surfactant secretion. Elevated inositol phosphate metabolism in airway progenitor cells may promote neutrophil infiltration and damage the lung barrier. Further, multiple metabolic alterations in the airway goblet cells are associated with impaired muco-ciliary clearance. Increased glycolysis, oxidative phosphorylation, and inositol phosphate metabolism not only enhance macrophage activation but also contribute to SARS-CoV-2 induced lung injury. The cytotoxicity of natural killer cells and CD8+ T cells may be enhanced by glycerolipid and inositol phosphate metabolism. Glycolytic activation in fibroblasts is related to myofibroblast differentiation and fibrogenesis. Glycolysis, oxidative phosphorylation, and glutathione metabolism may also boost the aging, apoptosis and proliferation of vascular smooth muscle cells, resulting in pulmonary arterial hypertension. In conclusion, this preliminary study revealed a possible cellular metabolic basis for the altered innate immunity, adaptive immunity, and niche cell function in the lung after SARS-CoV-2 infection. Therefore, patients with COVID-19 may benefit from therapeutic strategies targeting cellular metabolism in future.
Collapse
Affiliation(s)
- Shuangyan Li
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Fuxiaonan Zhao
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Jing Ye
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Kuan Li
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Qi Wang
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Zhongchao Du
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China
| | - Qing Yue
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Sisi Wang
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China
| | - Qi Wu
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, 890 Jingu Road, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, 890 Jingu Road, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe Hospital, Tianjin University, 890 Jingu Road, Tianjin, 300350, China.
| |
Collapse
|
9
|
Gu J, Wang C, Cao C, Huang J, Holzhauer S, Desilva H, Wesley EM, Evans DB, Benci J, Wichroski M, Wee S, Riese MJ. DGKζ exerts greater control than DGKα over CD8 + T cell activity and tumor inhibition. Oncoimmunology 2021; 10:1941566. [PMID: 34350062 PMCID: PMC8296965 DOI: 10.1080/2162402x.2021.1941566] [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] [Indexed: 12/04/2022] Open
Abstract
Two isoforms of diacylglycerol kinases (DGKs), DGKα and DGKζ, are primarily responsible for terminating DAG-mediated activation of Ras and PKCθ pathways in T cells. A direct comparison of tumor growth between mice lacking each isoform has not been undertaken. We evaluated the growth of three syngeneic tumor cell lines in mice lacking either DGKα or DGKζ in the presence or absence of treatment with anti-PD1 and determined that (i) mice deficient in DGKζ conferred enhanced control of tumor relative to mice deficient in DGKα and (ii) deficiency of DGKζ acted additively with anti-PD1 in tumor control. Consistent with this finding, functional and RNA-sequencing analyses revealed greater changes in stimulated DGKζ-deficient T cells compared with DGKα-deficient T cells, which were enhanced relative to wildtype T cells. DGKζ also imparted greater regulation than DGKα in human T cells. Together, these data support targeting the ζ isoform of DGKs to therapeutically enhance T cell anti-tumor activity.
Collapse
Affiliation(s)
- Junchen Gu
- Oncology Translational Research, Janssen Research & Development, PA
| | - Cindy Wang
- Oncology Research, Janssen Pharmaceutical, Raritan, NJ
| | - Carolyn Cao
- Oncology Discovery, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Jinwen Huang
- Oncology Discovery, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Sandra Holzhauer
- A Division of Versiti, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - Heshani Desilva
- Oncology Discovery, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Erin M Wesley
- A Division of Versiti, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Douglas B Evans
- Blood Research Institute, Blood Center of Wisconsin, a Division of Versiti, Milwaukee
| | - Joseph Benci
- Oncology Discovery, Bristol Myers Squibb, Princeton, NJ
| | - Michael Wichroski
- Oncology Discovery, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Susan Wee
- Oncology Discovery, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Matthew J Riese
- A Division of Versiti, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
10
|
Potential role of diacylglycerol kinases in immune-mediated diseases. Clin Sci (Lond) 2021; 134:1637-1658. [PMID: 32608491 DOI: 10.1042/cs20200389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
The mechanism promoting exacerbated immune responses in allergy and autoimmunity as well as those blunting the immune control of cancer cells are of primary interest in medicine. Diacylglycerol kinases (DGKs) are key modulators of signal transduction, which blunt diacylglycerol (DAG) signals and produce phosphatidic acid (PA). By modulating lipid second messengers, DGK modulate the activity of downstream signaling proteins, vesicle trafficking and membrane shape. The biological role of the DGK α and ζ isoforms in immune cells differentiation and effector function was subjected to in deep investigations. DGK α and ζ resulted in negatively regulating synergistic way basal and receptor induced DAG signals in T cells as well as leukocytes. In this way, they contributed to keep under control the immune response but also downmodulate immune response against tumors. Alteration in DGKα activity is also implicated in the pathogenesis of genetic perturbations of the immune function such as the X-linked lymphoproliferative disease 1 and localized juvenile periodontitis. These findings suggested a participation of DGK to the pathogenetic mechanisms underlying several immune-mediated diseases and prompted several researches aiming to target DGK with pharmacologic and molecular strategies. Those findings are discussed inhere together with experimental applications in tumors as well as in other immune-mediated diseases such as asthma.
Collapse
|
11
|
Kambayashi T, Deshpande DA. The role of diacylglycerol kinases in allergic airway disease. Curr Opin Pharmacol 2020; 51:50-58. [PMID: 32836013 DOI: 10.1016/j.coph.2020.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022]
Abstract
Asthma is an obstructive inflammatory airway disease. Airway obstruction is mediated by hyperresponsive airway smooth muscle cell contraction, which is induced and compounded by inflammation caused by T lymphocytes. One important signal transduction pathway that is involved in the activation of these cell types involves the generation of a lipid second messenger known as diacylglycerol (DAG). DAG levels are controlled in cells by a negative regulator known as DAG kinase (DGK). In this review, we discuss how the DAG signaling pathway attenuates the pathological function of immune cells and airway smooth muscle cells in allergic airway disease and asthma. Furthermore, we discuss how the enhancement of the DAG signaling pathway through the inhibition of DGK may represent a novel therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Deepak A Deshpande
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Xie D, Zhang S, Chen P, Deng W, Pan Y, Xie J, Wang J, Liao B, Sleasman JW, Zhong XP. Negative control of diacylglycerol kinase ζ-mediated inhibition of T cell receptor signaling by nuclear sequestration in mice. Eur J Immunol 2020; 50:1729-1745. [PMID: 32525220 DOI: 10.1002/eji.201948442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 04/17/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
Diacylglycerol kinases (DGKs) play important roles in restraining diacylglycerol (DAG)-mediated signaling. Within the DGK family, the ζ isoform appears to be the most important isoform in T cells for controlling their development and function. DGKζ has been demonstrated to regulate T cell maturation, activation, anergy, effector/memory differentiation, defense against microbial infection, and antitumor immunity. Given its critical functions, DGKζ function should be tightly regulated to ensure proper signal transduction; however, mechanisms that control DGKζ function are still poorly understood. We report here that DGKζ dynamically translocates from the cytosol into the nuclei in T cells after TCR stimulation. In mice, DGKζ mutant defective in nuclear localization displayed enhanced ability to inhibit TCR-induced DAG-mediated signaling in primary T cells, maturation of conventional αβT and iNKT cells, and activation of peripheral T cells compared with WT DGKζ. Our study reveals for the first time nuclear sequestration of DGKζ as a negative control mechanism to spatially restrain it from terminating DAG mediated signaling in T cells. Our data suggest that manipulation of DGKζ nucleus-cytosol shuttling as a novel strategy to modulate DGKζ activity and immune responses for treatment of autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Danli Xie
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Shimeng Zhang
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Pengcheng Chen
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Wenhai Deng
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Yun Pan
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Jinhai Xie
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Jinli Wang
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Bryce Liao
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - John W Sleasman
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Xiao-Ping Zhong
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina.,Department of Immunology, Duke University Medical Center, Durham, North Carolina.,Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
13
|
Recent insights of T cell receptor-mediated signaling pathways for T cell activation and development. Exp Mol Med 2020; 52:750-761. [PMID: 32439954 PMCID: PMC7272404 DOI: 10.1038/s12276-020-0435-8] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
T cell activation requires extracellular stimulatory signals that are mainly mediated by T cell receptor (TCR) complexes. The TCR recognizes antigens on major histocompatibility complex molecules with the cooperation of CD4 or CD8 coreceptors. After recognition, TCR-induced signaling cascades that propagate signals via various molecules and second messengers are induced. Consequently, many features of T cell-mediated immune responses are determined by these intracellular signaling cascades. Furthermore, differences in the magnitude of TCR signaling direct T cells toward distinct effector linages. Therefore, stringent regulation of T cell activation is crucial for T cell homeostasis and proper immune responses. Dysregulation of TCR signaling can result in anergy or autoimmunity. In this review, we summarize current knowledge on the pathways that govern how the TCR complex transmits signals into cells and the roles of effector molecules that are involved in these pathways.
Collapse
|
14
|
Saravia J, Raynor JL, Chapman NM, Lim SA, Chi H. Signaling networks in immunometabolism. Cell Res 2020; 30:328-342. [PMID: 32203134 PMCID: PMC7118125 DOI: 10.1038/s41422-020-0301-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Adaptive immunity is essential for pathogen and tumor eradication, but may also trigger uncontrolled or pathological inflammation. T cell receptor, co-stimulatory and cytokine signals coordinately dictate specific signaling networks that trigger the activation and functional programming of T cells. In addition, cellular metabolism promotes T cell responses and is dynamically regulated through the interplay of serine/threonine kinases, immunological cues and nutrient signaling networks. In this review, we summarize the upstream regulators and signaling effectors of key serine/threonine kinase-mediated signaling networks, including PI3K–AGC kinases, mTOR and LKB1–AMPK pathways that regulate metabolism, especially in T cells. We also provide our perspectives about the pending questions and clinical applicability of immunometabolic signaling. Understanding the regulators and effectors of immunometabolic signaling networks may uncover therapeutic targets to modulate metabolic programming and T cell responses in human disease.
Collapse
Affiliation(s)
- Jordy Saravia
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jana L Raynor
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Seon Ah Lim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
15
|
Yang J, Wang HX, Xie J, Li L, Wang J, Wan ECK, Zhong XP. DGK α and ζ Activities Control T H1 and T H17 Cell Differentiation. Front Immunol 2020; 10:3048. [PMID: 32010133 PMCID: PMC6974463 DOI: 10.3389/fimmu.2019.03048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 12/12/2019] [Indexed: 01/09/2023] Open
Abstract
CD4+ T helper (TH) cells are critical for protective adaptive immunity against pathogens, and they also contribute to the pathogenesis of autoimmune diseases. How TH differentiation is regulated by the TCR's downstream signaling is still poorly understood. We describe here that diacylglycerol kinases (DGKs), which are enzymes that convert diacylglycerol (DAG) to phosphatidic acid, exert differential effects on TH cell differentiation in a DGK dosage-dependent manner. A deficiency of either DGKα or ζ selectively impaired TH1 differentiation without obviously affecting TH2 and TH17 differentiation. However, simultaneous ablation of both DGKα and ζ promoted TH1 and TH17 differentiation in vitro and in vivo, leading to exacerbated airway inflammation. Furthermore, we demonstrate that dysregulation of TH17 differentiation of DGKα and ζ double-deficient CD4+ T cells was, at least in part, caused by increased mTOR complex 1/S6K1 signaling.
Collapse
Affiliation(s)
- Jialong Yang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Hong-Xia Wang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Jinhai Xie
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Lei Li
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Jinli Wang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Edwin C K Wan
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Xiao-Ping Zhong
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Hematologic Malignancies and Cellular Therapies Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
16
|
Pan Y, Deng W, Xie J, Zhang S, Wan ECK, Li L, Tao H, Hu Z, Chen Y, Ma L, Gao J, Zhong XP. Graded diacylglycerol kinases α and ζ activities ensure mucosal-associated invariant T-cell development in mice. Eur J Immunol 2019; 50:192-204. [PMID: 31710099 DOI: 10.1002/eji.201948289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 01/22/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells participate in both protective immunity and pathogenesis of diseases. Most murine MAIT cells express an invariant TCRVα19-Jα33 (iVα19) TCR, which triggers signals crucial for their development. However, signal pathways downstream of the iVα19TCR and their regulation in MAIT cells are unknown. Diacylglycerol (DAG) is a critical second messenger that relays the TCR signal to multiple downstream signaling cascades. DAG is terminated by DAG kinase (DGK)-mediated phosphorylation and conversion to phosphatidic acid. We have demonstrated here that downregulation of DAG caused by enhanced DGK activity impairs late-stage MAIT cell maturation in both thymus and spleen. Moreover, deficiency of DGKζ but not DGKα by itself causes modest decreases in MAIT cells, and deficiency of both DGKα and ζ results in severe reductions of MAIT cells in an autonomous manner. Our studies have revealed that DAG signaling is not only critical but also must be tightly regulated by DGKs for MAIT cell development and that both DGKα and, more prominently, DGKζ contribute to the overall DGK activity for MAIT cell development.
Collapse
Affiliation(s)
- Yun Pan
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wenhai Deng
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jinhai Xie
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shimeng Zhang
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Edwin C K Wan
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Department of Microbiology, Immunology, & Cell Biology and Department of Neuroscience, West Virginia University, Morgantown, WV
| | - Lei Li
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Department of Breast and Thyroid Surgery and Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huishan Tao
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiming Hu
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yongping Chen
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University and Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jimin Gao
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiao-Ping Zhong
- Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, NC.,Department of Immunology and Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Duke Cancer Institute, Duke University Medical Center, Durham, NC
| |
Collapse
|
17
|
DGKα in Neutrophil Biology and Its Implications for Respiratory Diseases. Int J Mol Sci 2019; 20:ijms20225673. [PMID: 31766109 PMCID: PMC6887790 DOI: 10.3390/ijms20225673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022] Open
Abstract
Diacylglycerol kinases (DGKs) play a key role in phosphoinositide signaling by removing diacylglycerol and generating phosphatidic acid. Besides the well-documented role of DGKα and DGKζ as negative regulators of lymphocyte responses, a robust body of literature points to those enzymes, and specifically DGKα, as crucial regulators of leukocyte function. Upon neutrophil stimulation, DGKα activation is necessary for migration and a productive response. The role of DGKα in neutrophils is evidenced by its aberrant behavior in juvenile periodontitis patients, which express an inactive DGKα transcript. Together with in vitro experiments, this suggests that DGKs may represent potential therapeutic targets for disorders where inflammation, and neutrophils in particular, plays a major role. In this paper we focus on obstructive respiratory diseases, including asthma and chronic obstructive pulmonary disease (COPD), but also rare genetic diseases such as alpha-1-antitrypsin deficiency. Indeed, the biological role of DGKα is understudied outside the T lymphocyte field. The recent wave of research aiming to develop novel and specific inhibitors as well as KO mice will allow a better understanding of DGK's role in neutrophilic inflammation. Better knowledge and pharmacologic tools may also allow DGK to move from the laboratory bench to clinical trials.
Collapse
|
18
|
Nair A, Chauhan P, Saha B, Kubatzky KF. Conceptual Evolution of Cell Signaling. Int J Mol Sci 2019; 20:E3292. [PMID: 31277491 PMCID: PMC6651758 DOI: 10.3390/ijms20133292] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
During the last 100 years, cell signaling has evolved into a common mechanism for most physiological processes across systems. Although the majority of cell signaling principles were initially derived from hormonal studies, its exponential growth has been supported by interdisciplinary inputs, e.g., from physics, chemistry, mathematics, statistics, and computational fields. As a result, cell signaling has grown out of scope for any general review. Here, we review how the messages are transferred from the first messenger (the ligand) to the receptor, and then decoded with the help of cascades of second messengers (kinases, phosphatases, GTPases, ions, and small molecules such as cAMP, cGMP, diacylglycerol, etc.). The message is thus relayed from the membrane to the nucleus where gene expression ns, subsequent translations, and protein targeting to the cell membrane and other organelles are triggered. Although there are limited numbers of intracellular messengers, the specificity of the response profiles to the ligands is generated by the involvement of a combination of selected intracellular signaling intermediates. Other crucial parameters in cell signaling are its directionality and distribution of signaling strengths in different pathways that may crosstalk to adjust the amplitude and quality of the final effector output. Finally, we have reflected upon its possible developments during the coming years.
Collapse
Affiliation(s)
- Arathi Nair
- National Center for Cell Science (NCCS), Ganeshkhind, Pune 411007, India
| | - Prashant Chauhan
- National Center for Cell Science (NCCS), Ganeshkhind, Pune 411007, India
| | - Bhaskar Saha
- National Center for Cell Science (NCCS), Ganeshkhind, Pune 411007, India.
| | - Katharina F Kubatzky
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Jang SY, Hwang J, Kim BS, Lee EY, Oh BH, Kim MH. Structural basis of inactivation of Ras and Rap1 small GTPases by Ras/Rap1-specific endopeptidase from the sepsis-causing pathogen Vibrio vulnificus. J Biol Chem 2018; 293:18110-18122. [PMID: 30282804 PMCID: PMC6254334 DOI: 10.1074/jbc.ra118.004857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/25/2018] [Indexed: 12/15/2022] Open
Abstract
Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are secreted by Gram-negative bacteria and function as primary virulence-promoting macromolecules that deliver multiple cytopathic and cytotoxic effector domains into the host cytoplasm. Among these effectors, Ras/Rap1-specific endopeptidase (RRSP) catalyzes the sequence-specific cleavage of the Switch I region of the cellular substrates Ras and Rap1 that are crucial for host innate immune defenses during infection. To dissect the molecular basis underpinning RRSP-mediated substrate inactivation, we determined the crystal structure of an RRSP from the sepsis-causing bacterial pathogen Vibrio vulnificus (VvRRSP). Structural and biochemical analyses revealed that VvRRSP is a metal-independent TIKI family endopeptidase composed of an N-terminal membrane-localization and substrate-recruitment domain (N lobe) connected via an inter-lobe linker to the C-terminal active site-coordinating core β-sheet-containing domain (C lobe). Structure-based mutagenesis identified the 2His/2Glu catalytic residues in the core catalytic domain that are shared with other TIKI family enzymes and that are essential for Ras processing. In vitro KRas cleavage assays disclosed that deleting the N lobe in VvRRSP causes complete loss of enzymatic activity. Endogenous Ras cleavage assays combined with confocal microscopy analysis of HEK293T cells indicated that the N lobe functions both in membrane localization via the first α-helix and in substrate assimilation by altering the functional conformation of the C lobe to facilitate recruitment of cellular substrates. Collectively, these results indicate that RRSP is a critical virulence factor that robustly inactivates Ras and Rap1 and augments the pathogenicity of invading bacteria via the combined effects of its N and C lobes.
Collapse
Affiliation(s)
- Song Yee Jang
- From the Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141,; the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and
| | - Jungwon Hwang
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and.
| | - Byoung Sik Kim
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and; the Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Eun-Young Lee
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and
| | - Byung-Ha Oh
- From the Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141,.
| | - Myung Hee Kim
- the Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, and.
| |
Collapse
|
20
|
CRISPR/Cas9-Mediated Knockout of DGK Improves Antitumor Activities of Human T Cells. Cancer Res 2018; 78:4692-4703. [DOI: 10.1158/0008-5472.can-18-0030] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/29/2018] [Accepted: 06/19/2018] [Indexed: 11/16/2022]
|
21
|
Wesley EM, Xin G, McAllister D, Malarkannan S, Newman DK, Dwinell MB, Cui W, Johnson BD, Riese MJ. Diacylglycerol kinase ζ (DGKζ) and Casitas b-lineage proto-oncogene b-deficient mice have similar functional outcomes in T cells but DGKζ-deficient mice have increased T cell activation and tumor clearance. Immunohorizons 2018; 2:107-118. [PMID: 30027154 DOI: 10.4049/immunohorizons.1700055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeting negative regulators downstream of the T cell receptor (TCR) represents a novel strategy to improve cancer immunotherapy. Two proteins that serve as critical inhibitory regulators downstream of the TCR are diacylglycerol kinase ζ (DGKζ), a regulator of Ras and PKC-θ signaling, and Casitas b-lineage proto-oncogene b (Cbl-b), an E3 ubiquitin ligase that predominantly regulates PI(3)K signaling. We sought to compare the signaling and functional effects that result from deletion of DGKζ, Cbl-b, or both (double knockout, DKO) in T cells, and to evaluate tumor responses generated in a clinically relevant orthotopic pancreatic tumor model. We found that whereas deletion of Cbl-b primarily served to enhance NF-κB signaling, deletion of DGKζ enhanced TCR-mediated signal transduction downstream of Ras/Erk and NF-κB. Deletion of DGKζ or Cbl-b comparably enhanced CD8+ T cell functional responses, such as proliferation, production of IFNγ, and generation of granzyme B when compared with WT T cells. DKO T cells demonstrated enhanced function above that observed with single knockout T cells after weak, but not strong, stimulation. Deletion of DGKζ, but not Cbl-b, however, resulted in significant increases in numbers of activated (CD44hi) CD8+ T cells in both non-treated and tumor-bearing mice. DGKζ-deficient mice also had enhanced control of pancreatic tumor cell growth compared to Cbl-b-deficient mice. This represents the first direct comparison between mice of these genotypes and suggests that T cell immunotherapies may be better improved by targeting TCR signaling molecules that are regulated by DGKζ as opposed to molecules regulated by Cbl-b.
Collapse
Affiliation(s)
- Erin M Wesley
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Gang Xin
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI
| | - Donna McAllister
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Subramaniam Malarkannan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Michael B Dwinell
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI
| | - Bryon D Johnson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Matthew J Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
22
|
Unexpected positive control of NFκB and miR-155 by DGKα and ζ ensures effector and memory CD8+ T cell differentiation. Oncotarget 2018; 7:33744-64. [PMID: 27014906 PMCID: PMC5085116 DOI: 10.18632/oncotarget.8164] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/04/2016] [Indexed: 11/25/2022] Open
Abstract
Signals from the T-cell receptor (TCR) and γ-chain cytokine receptors play crucial roles in initiating activation and effector/memory differentiation of CD8 T-cells. We report here that simultaneous deletion of both diacylglycerol kinase (DGK) α and ζ (DKO) severely impaired expansion of CD8 effector T cells and formation of memory CD8 T-cells after Listeria monocytogenes infection. Moreover, ablation of both DGKα and ζ in preformed memory CD8 T-cells triggered death and impaired homeostatic proliferation of these cells. DKO CD8 T-cells were impaired in priming due to decreased expression of chemokine receptors and migration to the draining lymph nodes. Moreover, DKO CD8 T-cells were unexpectedly defective in NFκB-mediated miR-155 transcript, leading to excessive SOCS1 expression and impaired γ-chain cytokine signaling. Our data identified a DGK-NFκB-miR-155-SOCS1 axis that bridges TCR and γ-chain cytokine signaling for robust CD8 T-cell primary and memory responses to bacterial infection.
Collapse
|
23
|
Jing W, Gershan JA, Holzhauer S, Weber J, Palen K, McOlash L, Pulakanti K, Wesley E, Rao S, Johnson BD, Riese MJ. T Cells Deficient in Diacylglycerol Kinase ζ Are Resistant to PD-1 Inhibition and Help Create Persistent Host Immunity to Leukemia. Cancer Res 2017; 77:5676-5686. [DOI: 10.1158/0008-5472.can-17-1309] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/24/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
|
24
|
Abstract
There has been dramatic success in treating patients with adoptive transfer of autologous T cells genetically modified to express a chimeric antigen receptor redirecting them to the antigen CD19. Despite this success, the application of chimeric antigen receptor T-cell therapy in solid malignancies has encountered many challenges that need to be overcome if similar success across other cancers is to become a reality. These challenges can be classified into 6 categories: the heterogeneity of tumor cell clones and tumor-associated antigen expression; poor T-cell trafficking into the tumor site; poor T-cell survival and persistence; the presence of suppressive immune cells; the secretion of suppressive soluble factors in the tumor microenvironment; and the upregulation of T-cell intrinsic inhibitory pathways. We outline specific representative hurdles in each of these categories and summarize the progress made in understanding them and developing strategies to overcome them.
Collapse
|
25
|
Chen SS, Hu Z, Zhong XP. Diacylglycerol Kinases in T Cell Tolerance and Effector Function. Front Cell Dev Biol 2016; 4:130. [PMID: 27891502 PMCID: PMC5103287 DOI: 10.3389/fcell.2016.00130] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022] Open
Abstract
Diacylglycerol kinases (DGKs) are a family of enzymes that regulate the relative levels of diacylglycerol (DAG) and phosphatidic acid (PA) in cells by phosphorylating DAG to produce PA. Both DAG and PA are important second messengers cascading T cell receptor (TCR) signal by recruiting multiple effector molecules, such as RasGRP1, PKCθ, and mTOR. Studies have revealed important physiological functions of DGKs in the regulation of receptor signaling and the development and activation of immune cells. In this review, we will focus on recent progresses in our understanding of two DGK isoforms, α and ζ, in CD8 T effector and memory cell differentiation, regulatory T cell development and function, and invariant NKT cell development and effector lineage differentiation.
Collapse
Affiliation(s)
- Shelley S Chen
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center Durham, NC, USA
| | - Zhiming Hu
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical CenterDurham, NC, USA; Institute of Biotherapy, School of Biotechnology, Southern Medical UniversityGuangzhou, China
| | - Xiao-Ping Zhong
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical CenterDurham, NC, USA; Department of Immunology, Duke University Medical CenterDurham, NC, USA; Hematologic Malignancies and Cellular Therapies Program, Duke Cancer Institute, Duke University Medical CenterDurham, NC, USA
| |
Collapse
|
26
|
Merida I, Andrada E, Gharbi SI, Avila-Flores A. Redundant and specialized roles for diacylglycerol kinases and in the control of T cell functions. Sci Signal 2015; 8:re6. [DOI: 10.1126/scisignal.aaa0974] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
27
|
Joshi RP, Schmidt AM, Das J, Pytel D, Riese MJ, Lester M, Diehl JA, Behrens EM, Kambayashi T, Koretzky GA. The ζ isoform of diacylglycerol kinase plays a predominant role in regulatory T cell development and TCR-mediated ras signaling. Sci Signal 2013; 6:ra102. [PMID: 24280043 DOI: 10.1126/scisignal.2004373] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diacylglycerol (DAG) is a critical second messenger that mediates T cell receptor (TCR)-stimulated signaling. The abundance of DAG is reduced by the diacylglycerol kinases (DGKs), which catalyze the conversion of DAG to phosphatidic acid (PA) and thus inhibit DAG-mediated signaling. In T cells, the predominant DGK isoforms are DGKα and DGKζ, and deletion of the genes encoding either isoform enhances DAG-mediated signaling. We found that DGKζ, but not DGKα, suppressed the development of natural regulatory T (T(reg)) cells and predominantly mediated Ras and Akt signaling downstream of the TCR. The differential functions of DGKα and DGKζ were not attributable to differences in protein abundance in T cells or in their localization to the contact sites between T cells and antigen-presenting cells. RasGRP1, a key DAG-mediated activator of Ras signaling, associated to a greater extent with DGKζ than with DGKα; however, in silico modeling of TCR-stimulated Ras activation suggested that a difference in RasGRP1 binding affinity was not sufficient to cause differences in the functions of each DGK isoform. Rather, the model suggested that a greater catalytic rate for DGKζ than for DGKα might lead to DGKζ exhibiting increased suppression of Ras-mediated signals compared to DGKα. Consistent with this notion, experimental studies demonstrated that DGKζ was more effective than DGKα at catalyzing the metabolism of DAG to PA after TCR stimulation. The enhanced effective enzymatic production of PA by DGKζ is therefore one possible mechanism underlying the dominant functions of DGKζ in modulating T(reg) cell development.
Collapse
Affiliation(s)
- Rohan P Joshi
- 1Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Diacylglycerol (DAG), a second messenger generated by phospholipase Cγ1 activity upon engagement of a T-cell receptor, triggers several signaling cascades that play important roles in T cell development and function. A family of enzymes called DAG kinases (DGKs) catalyzes the phosphorylation of DAG to phosphatidic acid, acting as a braking mechanism that terminates DAG-mediated signals. Two DGK isoforms, α and ζ, are expressed predominantly in T cells and synergistically regulate the development of both conventional αβ T cells and invariant natural killer T cells in the thymus. In mature T cells, the activity of these DGK isoforms aids in the maintenance of self-tolerance by preventing T-cell hyperactivation upon T cell receptor stimulation and by promoting T-cell anergy. In CD8 cells, reduced DGK activity is associated with enhanced primary responses against viruses and tumors. Recent work also has established an important role for DGK activity at the immune synapse and identified partners that modulate DGK function. In addition, emerging evidence points to previously unappreciated roles for DGK function in directional secretion and T-cell adhesion. This review describes the multitude of roles played by DGKs in T cell development and function and emphasizes recent advances in the field.
Collapse
Affiliation(s)
- Sruti Krishna
- Department of Pediatrics, Division of Allergy and Immunology and Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
29
|
Shin J, Xie D, Zhong XP. MicroRNA-34a enhances T cell activation by targeting diacylglycerol kinase ζ. PLoS One 2013; 8:e77983. [PMID: 24147106 PMCID: PMC3798301 DOI: 10.1371/journal.pone.0077983] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/06/2013] [Indexed: 11/18/2022] Open
Abstract
The engagement of the T cell receptor (TCR) induces the generation of diacylglycerol (DAG), an important second messenger activating both the Ras/Erk and PKCθ/NFκB pathways. DAG kinases (DGKs) participate in the metabolism of DAG by converting it to phosphatidic acid. DGKζ has been demonstrated to be able to inhibit DAG signaling following TCR engagement. Deficiency of DGKζ increases the sensitivity of T cells to TCR stimulation, resulting in enhanced T cell activation ex vivo and in vivo. However, the mechanisms that control DGKζ expression are poorly understood. Here we demonstrate that DGKζ mRNA is a direct target of a cellular microRNA miR-34a. The DGKζ transcript is decreased, whereas the primary miR-34a is upregulated upon TCR stimulation. Ectopic miR-34a expression suppresses DGKζ protein expression through the seed match binding to both the 3' untranslated region and coding region of DGKζ mRNA, leading to increased ERK1/2 phosphorylation and surface expression of the T cell activation marker CD69 following TCR cross-linking. In contrast, overexpression of a miR-34a competitive inhibitor increases DGKζ expression and suppresses TCR-mediated T cell activation. Together, our data demonstrate that miR-34a is a negative regulator for DGKζ and may play an important role in regulating T cell activation.
Collapse
Affiliation(s)
- Jinwook Shin
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Danli Xie
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Ping Zhong
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
30
|
Wheeler ML, Dong MB, Brink R, Zhong XP, DeFranco AL. Diacylglycerol kinase ζ limits B cell antigen receptor-dependent activation of ERK signaling to inhibit early antibody responses. Sci Signal 2013; 6:ra91. [PMID: 24129701 DOI: 10.1126/scisignal.2004189] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Signaling downstream of the B cell antigen receptor (BCR) is tightly regulated to enable cells to gauge the strength and duration of antigen-receptor interactions and to respond appropriately. We investigated whether metabolism of the second messenger diacylglycerol (DAG) by members of the family of DAG kinases (DGKs) played a role in modulating the magnitude of signaling by DAG downstream of the BCR. In the absence of DGKζ, the threshold for BCR signaling, measured as activation of the Ras-extracellular signal-regulated kinase (ERK) pathway, was markedly reduced in mature follicular B cells, which resulted in enhanced responses to antigen in vitro and in vivo. Inhibition of DAG signaling by DGKζ limited the number of antibody-secreting cells that were generated early in response to T cell-independent type 2 antigens, as well as to T cell-dependent antigens. Furthermore, the effect of loss of DGKζ closely resembled the effect of increasing the affinity of the BCR for antigen during the T cell-dependent antibody response. These results suggest that the magnitude of DAG signaling is important for translating the affinity of the BCR for antigen into the amount of antibody produced during the early stages of an immune response.
Collapse
Affiliation(s)
- Matthew L Wheeler
- 1Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
31
|
Diacylglycerol kinase zeta positively controls the development of iNKT-17 cells. PLoS One 2013; 8:e75202. [PMID: 24073253 PMCID: PMC3779165 DOI: 10.1371/journal.pone.0075202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/13/2013] [Indexed: 11/22/2022] Open
Abstract
Invariant natural killer T (iNKT) cells play important roles in bridging innate and adaptive immunity via rapidly producing a variety of cytokines. A small subset of iNKT cells produces IL-17 and is generated in the thymus during iNKT-cell ontogeny. The mechanisms that control the development of these IL-17-producing iNKT-17 cells (iNKT-17) are still not well defined. Diacylglycerol kinase ζ (DGKζ) belongs to a family of enzymes that catalyze the phosphorylation and conversion of diacylglycerol to phosphatidic acid, two important second messengers involved in signaling from numerous receptors. We report here that DGKζ plays an important role in iNKT-17 development. A deficiency of DGKζ in mice causes a significant reduction of iNKT-17 cells, which is correlated with decreased RORγt and IL-23 receptor expression. Interestingly, iNKT-17 defects caused by DGKζ deficiency can be corrected in chimeric mice reconstituted with mixed wild-type and DGKζ-deficient bone marrow cells. Taken together, our data identify DGKζ as an important regulator of iNKT-17 development through iNKT-cell extrinsic mechanisms.
Collapse
|
32
|
Krishna S, Zhong XP. Regulation of Lipid Signaling by Diacylglycerol Kinases during T Cell Development and Function. Front Immunol 2013; 4:178. [PMID: 23847619 PMCID: PMC3701226 DOI: 10.3389/fimmu.2013.00178] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 06/19/2013] [Indexed: 01/14/2023] Open
Abstract
Diacylglycerol (DAG) and phosphatidic acid (PA) are bioactive lipids synthesized when the T cell receptor binds to a cognate peptide-MHC complex. DAG triggers signaling by recruiting Ras guanyl-releasing protein 1, PKCθ, and other effectors, whereas PA binds to effector molecules that include mechanistic target of rapamycin, Src homology region 2 domain-containing phosphatase 1, and Raf1. While DAG-mediated pathways have been shown to play vital roles in T cell development and function, the importance of PA-mediated signals remains less clear. The diacylglycerol kinase (DGK) family of enzymes phosphorylates DAG to produce PA, serving as a molecular switch that regulates the relative levels of these critical second messengers. Two DGK isoforms, α and ζ, are predominantly expressed in T lineage cells and play an important role in conventional αβ T cell development. In mature T cells, the activity of these DGK isoforms aids in the maintenance of self-tolerance by preventing T cell hyper-activation and promoting T cell anergy. In this review, we discuss the roles of DAG-mediated pathways, PA-effectors, and DGKs in T cell development and function. We also highlight recent work that has uncovered previously unappreciated roles for DGK activity, for instance in invariant NKT cell development, anti-tumor and anti-viral CD8 responses, and the directional secretion of soluble effectors.
Collapse
Affiliation(s)
- Sruti Krishna
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center , Durham, NC , USA ; Department of Immunology, Duke University Medical Center , Durham, NC , USA
| | | |
Collapse
|
33
|
Joshi RP, Koretzky GA. Diacylglycerol kinases: regulated controllers of T cell activation, function, and development. Int J Mol Sci 2013; 14:6649-73. [PMID: 23531532 PMCID: PMC3645659 DOI: 10.3390/ijms14046649] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/07/2013] [Accepted: 03/14/2013] [Indexed: 01/22/2023] Open
Abstract
Diacylglycerol kinases (DGKs) are a diverse family of enzymes that catalyze the conversion of diacylglycerol (DAG), a crucial second messenger of receptor-mediated signaling, to phosphatidic acid (PA). Both DAG and PA are bioactive molecules that regulate a wide set of intracellular signaling proteins involved in innate and adaptive immunity. Clear evidence points to a critical role for DGKs in modulating T cell activation, function, and development. More recently, studies have elucidated factors that control DGK function, suggesting an added complexity to how DGKs act during signaling. This review summarizes the available knowledge of the function and regulation of DGK isoforms in signal transduction with a particular focus on T lymphocytes.
Collapse
Affiliation(s)
- Rohan P. Joshi
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; E-Mail:
| | - Gary A. Koretzky
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; E-Mail:
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-215-746-5522; Fax: +1-215-746-5525
| |
Collapse
|
34
|
Gorentla BK, Krishna S, Shin J, Inoue M, Shinohara ML, Grayson JM, Fukunaga R, Zhong XP. Mnk1 and 2 are dispensable for T cell development and activation but important for the pathogenesis of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2012; 190:1026-37. [PMID: 23269249 DOI: 10.4049/jimmunol.1200026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
T cell development and activation are usually accompanied by expansion and production of numerous proteins that require active translation. The eukaryotic translation initiation factor 4E (eIF4E) binds to the 5' cap structure of mRNA and is critical for cap-dependent translational initiation. It has been hypothesized that MAPK-interacting kinase 1 and 2 (Mnk1/2) promote cap-dependent translation by phosphorylating eIF4E at serine 209 (S209). Pharmacologic studies using inhibitors have suggested that Mnk1/2 have important roles in T cells. However, genetic evidence supporting such conclusions is lacking. Moreover, the signaling pathways that regulate Mnk1/2 in T cells remain unclear. We demonstrate that TCR engagement activates Mnk1/2 in primary T cells. Such activation is dependent on Ras-Erk1/2 signaling and is inhibited by diacylglycerol kinases α and ζ. Mnk1/2 double deficiency in mice abolishes TCR-induced eIF4E S209 phosphorylation, indicating their absolute requirement for eIF4E S209 phosphorylation. However, Mnk1/2 double deficiency does not affect the development of conventional αβ T cells, regulatory T cells, or NKT cells. Furthermore, T cell activation, in vivo primary and memory CD8 T cell responses to microbial infection, and NKT cell cytokine production were not obviously altered by Mnk1/2 deficiency. Although Mnk1/2 deficiency causes decreased IL-17 and IFN-γ production by CD4 T cells following immunization of mice with myelin oligodendrocyte glycoprotein peptide in complete Freund's adjuvant, correlating with milder experimental autoimmune encephalomyelitis scores, it does not affect Th cell differentiation in vitro. Together, these data suggest that Mnk1/2 has a minimal role in T cell development and activation but may regulate non-T cell lineages to control Th1 and Th17 differentiation in vivo.
Collapse
Affiliation(s)
- Balachandra K Gorentla
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Gorentla BK, Zhong XP. T cell Receptor Signal Transduction in T lymphocytes. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2012; 2012:5. [PMID: 23946894 PMCID: PMC3740441 DOI: 10.4172/2155-9899.s12-005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The T cell receptor (TCR) recognizes self or foreign antigens presented by major histocompatibility complex (MHC) molecules. Engagement of the TCR triggers the formation of multi-molecular signalosomes that lead to the generation of second messengers and subsequent activation of multiple distal signaling cascades, such as the Ca+2-calcineurin-NFAT, RasGRP1-Ras-Erk1/2, PKCθ-IKK-NFκB, and TSC1/2-mTOR pathways. These signaling cascades control many aspects of T cell biology. Mechanisms have been evolved to fine-tune TCR signaling to maintain T cell homeostasis and self-tolerance, and to properly mount effective responses to microbial infection. Defects or deregulation of TCR signaling has been implicated in the pathogenesis of multiple human diseases.
Collapse
Affiliation(s)
- Balachandra K Gorentla
- Pediatric Biology Center, Translational Health Science and Technology Institute, Gurgaon, 122016, India
| | - Xiao-Ping Zhong
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
36
|
Abstract
Diacylglycerol kinase α (DGKα) regulates diacylglycerol levels, catalyzing its conversion into phosphatidic acid. The α isoform is central to immune response regulation; it downmodulates Ras-dependent pathways and is necessary for establishment of the unresponsive state termed anergy. DGKα functions are regulated in part at the transcriptional level although the mechanisms involved remain poorly understood. Here, we analyzed the 5' end structure of the mouse DGKα gene and detected three binding sites for forkhead box O (FoxO) transcription factors, whose function was confirmed using luciferase reporter constructs. FoxO1 and FoxO3 bound to the 5' regulatory region of DGKα in quiescent T cells, as well as after interleukin-2 (IL-2) withdrawal in activated T cells. FoxO binding to this region was lost after complete T cell activation or IL-2 addition, events that correlated with FoxO phosphorylation and a sustained decrease in DGKα gene expression. These data strongly support a role for FoxO proteins in promoting high DGKα levels and indicate a mechanism by which DGKα function is downregulated during productive T cell responses. Our study establishes a basis for a causal relationship between DGKα downregulation, IL-2, and anergy avoidance.
Collapse
|
37
|
Johnson DS, Chen YH. Ras family of small GTPases in immunity and inflammation. Curr Opin Pharmacol 2012; 12:458-63. [PMID: 22401931 DOI: 10.1016/j.coph.2012.02.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 02/02/2012] [Accepted: 02/03/2012] [Indexed: 11/29/2022]
Abstract
The Ras superfamily of small GTPases is a group of more than 150 small G proteins, all of which share some degree of homology to the founding member Ras. These small GTPases function as molecular switches within cells, impacting nearly all cellular processes. The Ras superfamily can be further divided into several smaller subfamilies, with those proteins that most closely resemble Ras belonging to the Ras subfamily. While heavily studied within the field of cancer biology, the Ras family of proteins also plays cardinal roles in immunity and inflammation. Here we review the roles of these molecular switches in regulating immune cell homeostasis and functions.
Collapse
Affiliation(s)
- Derek S Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | |
Collapse
|