1
|
Fan S, Kang B, Li S, Li W, Chen C, Chen J, Deng L, Chen D, Zhou J. Exploring the multifaceted role of RASGRP1 in disease: immune, neural, metabolic, and oncogenic perspectives. Cell Cycle 2024; 23:722-746. [PMID: 38865342 PMCID: PMC11229727 DOI: 10.1080/15384101.2024.2366009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/25/2023] [Indexed: 06/14/2024] Open
Abstract
RAS guanyl releasing protein 1 (RASGRP1) is a guanine nucleotide exchange factor (GEF) characterized by the presence of a RAS superfamily GEF domain. It functions as a diacylglycerol (DAG)-regulated nucleotide exchange factor, specifically activating RAS through the exchange of bound GDP for GTP. Activation of RAS by RASGRP1 has a wide range of downstream effects at the cellular level. Thus, it is not surprising that many diseases are associated with RASGRP1 disorders. Here, we present an overview of the structure and function of RASGRP1, its crucial role in the development, expression, and regulation of immune cells, and its involvement in various signaling pathways. This review comprehensively explores the relationship between RASGRP1 and various diseases, elucidates the underlying molecular mechanisms of RASGRP1 in each disease, and identifies potential therapeutic targets. This study provides novel insights into the role of RASGRP1 in insulin secretion and highlights its potential as a therapeutic target for diabetes. The limitations and challenges associated with studying RASGRP1 in disease are also discussed.
Collapse
Affiliation(s)
- Shangzhi Fan
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weiyi Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Canyu Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lijing Deng
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danjun Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Qin J, Liu Q, Liu A, Leng S, Wang S, Li C, Ma J, Peng J, Xu M. Empagliflozin modulates CD4 + T-cell differentiation via metabolic reprogramming in immune thrombocytopenia. Br J Haematol 2022; 198:765-775. [PMID: 35675486 DOI: 10.1111/bjh.18293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/03/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
Immune thrombocytopenia (ITP) is an acquired autoimmune disease, in which the imbalance of CD4+ T cell subsets play a key role in the pathogenesis. Since T cells highly depend on metabolism for their function, we hypothesized that T cell dysfunction may be due to intracellular metabolic reprogramming. We found that in ITP, T cell metabolism shifts from oxidative phosphorylation to glycolysis. Empagliflozin, a sodium-glucose cotransporter 2 inhibitor, has shown regulatory metabolic effects on proximal tubular epithelial cells and cardiac cells beyond glucose lowering. However, the effects of empagliflozin on T cells remain unknown. To further investigate the metabolic dysfunction of CD4+ T cells in ITP, we explored the effect of empagliflozin on CD4+ T-cell differentiation in ITP. Our results are the first to show that increased glycolysis in CD4+ T cells resulted in an unbalanced CD4+ T-cell population. Furthermore, empagliflozin can affect the differentiation of CD4+ T-cell subsets by inhibiting Th1 and Th17 cell populations while increasing Tregs. Empagliflozin appears to regulate CD4+ T cells through inhibiting the mTOR signal pathway. Considering these results, we propose that empagliflozin could be used as a potential therapeutic option for ITP by modulating metabolic reprogramming in CD4+ T cells.
Collapse
Affiliation(s)
- Jing Qin
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiang Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Anli Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaoqiu Leng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuwen Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaoyang Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ji Ma
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Miao Xu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
3
|
Mouri K, Guo MH, de Boer CG, Lissner MM, Harten IA, Newby GA, DeBerg HA, Platt WF, Gentili M, Liu DR, Campbell DJ, Hacohen N, Tewhey R, Ray JP. Prioritization of autoimmune disease-associated genetic variants that perturb regulatory element activity in T cells. Nat Genet 2022; 54:603-612. [PMID: 35513721 PMCID: PMC9793778 DOI: 10.1038/s41588-022-01056-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 03/18/2022] [Indexed: 12/30/2022]
Abstract
Genome-wide association studies (GWASs) have uncovered hundreds of autoimmune disease-associated loci; however, the causal genetic variants within each locus are mostly unknown. Here, we perform high-throughput allele-specific reporter assays to prioritize disease-associated variants for five autoimmune diseases. By examining variants that both promote allele-specific reporter expression and are located in accessible chromatin, we identify 60 putatively causal variants that enrich for statistically fine-mapped variants by up to 57.8-fold. We introduced the risk allele of a prioritized variant (rs72928038) into a human T cell line and deleted the orthologous sequence in mice, both resulting in reduced BACH2 expression. Naive CD8 T cells from mice containing the deletion had reduced expression of genes that suppress activation and maintain stemness and, upon acute viral infection, displayed greater propensity to become effector T cells. Our results represent an example of an effective approach for prioritizing variants and studying their physiologically relevant effects.
Collapse
Affiliation(s)
| | - Michael H Guo
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carl G de Boer
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Michelle M Lissner
- Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ingrid A Harten
- Systems Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Gregory A Newby
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Hannah A DeBerg
- Systems Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Winona F Platt
- Systems Immunology, Benaroya Research Institute, Seattle, WA, USA
| | | | - David R Liu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Daniel J Campbell
- Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA.
| | - John P Ray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Systems Immunology, Benaroya Research Institute, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Abnormal RasGRP1 Expression in the Post-Mortem Brain and Blood Serum of Schizophrenia Patients. Biomolecules 2022; 12:biom12020328. [PMID: 35204828 PMCID: PMC8869509 DOI: 10.3390/biom12020328] [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: 12/22/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 11/16/2022] Open
Abstract
Schizophrenia (SCZ) is a polygenic severe mental illness. Genome-wide association studies (GWAS) have detected genomic variants associated with this psychiatric disorder and pathway analyses have indicated immune system and dopamine signaling as core components of risk in dorsolateral-prefrontal cortex (DLPFC) and hippocampus, but the mechanistic links remain unknown. The RasGRP1 gene, encoding for a guanine nucleotide exchange factor, is implicated in dopamine signaling and immune response. RasGRP1 has been identified as a candidate risk gene for SCZ and autoimmune disease, therefore representing a possible point of convergence between mechanisms involving the nervous and the immune system. Here, we investigated RasGRP1 mRNA and protein expression in post-mortem DLPFC and hippocampus of SCZ patients and healthy controls, along with RasGRP1 protein content in the serum of an independent cohort of SCZ patients and control subjects. Differences in RasGRP1 expression between SCZ patients and controls were detected both in DLPFC and peripheral blood of samples analyzed. Our results indicate RasGRP1 may mediate risk for SCZ by involving DLPFC and peripheral blood, thus encouraging further studies to explore its possible role as a biomarker of the disease and/or a target for new medication.
Collapse
|
5
|
Heikkinen T, Bragge T, Kuosmanen J, Parkkari T, Gustafsson S, Kwan M, Beltran J, Ghavami A, Subramaniam S, Shahani N, Ramírez-Jarquín UN, Park L, Muñoz-Sanjuán I, Marchionini DM. Global Rhes knockout in the Q175 Huntington's disease mouse model. PLoS One 2021; 16:e0258486. [PMID: 34648564 PMCID: PMC8516231 DOI: 10.1371/journal.pone.0258486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/28/2021] [Indexed: 12/02/2022] Open
Abstract
Huntington's disease (HD) results from an expansion mutation in the polyglutamine tract in huntingtin. Although huntingtin is ubiquitously expressed in the body, the striatum suffers the most severe pathology. Rhes is a Ras-related small GTP-binding protein highly expressed in the striatum that has been reported to modulate mTOR and sumoylation of mutant huntingtin to alter HD mouse model pathogenesis. Reports have varied on whether Rhes reduction is desirable for HD. Here we characterize multiple behavioral and molecular endpoints in the Q175 HD mouse model with genetic Rhes knockout (KO). Genetic RhesKO in the Q175 female mouse resulted in both subtle attenuation of Q175 phenotypic features, and detrimental effects on other kinematic features. The Q175 females exhibited measurable pathogenic deficits, as measured by MRI, MRS and DARPP32, however, RhesKO had no effect on these readouts. Additionally, RhesKO in Q175 mixed gender mice deficits did not affect mTOR signaling, autophagy or mutant huntingtin levels. We conclude that global RhesKO does not substantially ameliorate or exacerbate HD mouse phenotypes in Q175 mice.
Collapse
Affiliation(s)
| | - Timo Bragge
- Charles River Discovery Services, Kuopio, Finland
| | | | | | | | - Mei Kwan
- Psychogenics, Paramus, New Jersey, United States of America
| | - Jose Beltran
- Psychogenics, Paramus, New Jersey, United States of America
| | - Afshin Ghavami
- Psychogenics, Paramus, New Jersey, United States of America
| | - Srinivasa Subramaniam
- The Scripps Research Institute, Department of Neuroscience, Jupiter, Florida, United States of America
| | - Neelam Shahani
- The Scripps Research Institute, Department of Neuroscience, Jupiter, Florida, United States of America
| | | | - Larry Park
- CHDI Management/CHDI Foundation, New York, New York, United States of America
| | | | | |
Collapse
|
6
|
Regulation of the Small GTPase Ras and Its Relevance to Human Disease. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2262:19-43. [PMID: 33977469 DOI: 10.1007/978-1-0716-1190-6_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ras research has experienced a considerable boost in recent years, not least prompted by the Ras initiative launched by the NCI in 2013 ( https://www.cancer.gov/research/key-initiatives/ras ), accompanied and conditioned by a strongly reinvigorated determination within the Ras community to develop therapeutics attacking directly the Ras oncoproteins. As a member of the small G-protein superfamily, function and transforming activity of Ras all revolve about its GDP/GTP loading status. For one thing, the extent of GTP loading will determine the proportion of active Ras in the cell, with implications for intensity and quality of downstream signaling. But also the rate of nucleotide exchange, i.e., the Ras-GDP/GTP cycling rate, can have a major impact on Ras function, as illustrated perhaps most impressively by newly discovered fast-cycling oncogenic mutants of the Ras-related GTPase Rac1. Thus, while the last years have witnessed memorable new findings and technical developments in the Ras field, leading to an improved insight into many aspects of Ras biology, they have not jolted at the basics, but rather deepened our view of the fundamental regulatory principles of Ras activity control. In this brief review, we revisit the role and mechanisms of Ras nucleotide loading and its implications for cancer in the light of recent findings.
Collapse
|
7
|
Fernández-Medarde A, Santos E. Ras GEF Mouse Models for the Analysis of Ras Biology and Signaling. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2262:361-395. [PMID: 33977490 DOI: 10.1007/978-1-0716-1190-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Animal models have become in recent years a crucial tool to understand the physiological and pathological roles of many cellular proteins. They allow analysis of the functional consequences of [1] complete or partial (time- or organ-limited) removal of specific proteins (knockout animals), [2] the exchange of a wild-type allele for a mutant or truncated version found in human illnesses (knock-in), or [3] the effect of overexpression of a given protein in the whole body or in specific organs (transgenic mice). In this regard, the study of phenotypes in Ras GEF animal models has allowed researchers to find specific functions for otherwise very similar proteins, uncovering their role in physiological contexts such as memory formation, lymphopoiesis, photoreception, or body homeostasis. In addition, mouse models have been used to unveil the functional role of Ras GEFs under pathological conditions, including Noonan syndrome, skin tumorigenesis, inflammatory diseases, diabetes, or ischemia among others. In the following sections, we will describe the methodological approaches employed for Ras GEF animal model analyses, as well as the main discoveries made.
Collapse
Affiliation(s)
- Alberto Fernández-Medarde
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain.
| | - Eugenio Santos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain
| |
Collapse
|
8
|
Baars MJ, Douma T, Simeonov DR, Myers DR, Kulhanek K, Banerjee S, Zwakenberg S, Baltissen MP, Amini M, de Roock S, van Wijk F, Vermeulen M, Marson A, Roose JP, Vercoulen Y. Dysregulated RASGRP1 expression through RUNX1 mediated transcription promotes autoimmunity. Eur J Immunol 2021; 51:471-482. [PMID: 33065764 PMCID: PMC7894479 DOI: 10.1002/eji.201948451] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 08/11/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022]
Abstract
RasGRP1 is a Ras guanine nucleotide exchange factor, and an essential regulator of lymphocyte receptor signaling. In mice, Rasgrp1 deletion results in defective T lymphocyte development. RASGRP1-deficient patients suffer from immune deficiency, and the RASGRP1 gene has been linked to autoimmunity. However, how RasGRP1 levels are regulated, and if RasGRP1 dosage alterations contribute to autoimmunity remains unknown. We demonstrate that diminished Rasgrp1 expression caused defective T lymphocyte selection in C57BL/6 mice, and that the severity of inflammatory disease inversely correlates with Rasgrp1 expression levels. In patients with autoimmunity, active inflammation correlated with decreased RASGRP1 levels in CD4+ T cells. By analyzing H3K27 acetylation profiles in human T cells, we identified a RASGRP1 enhancer that harbors autoimmunity-associated SNPs. CRISPR-Cas9 disruption of this enhancer caused lower RasGRP1 expression, and decreased binding of RUNX1 and CBFB transcription factors. Analyzing patients with autoimmunity, we detected reduced RUNX1 expression in CD4+ T cells. Lastly, we mechanistically link RUNX1 to transcriptional regulation of RASGRP1 to reveal a key circuit regulating RasGRP1 expression, which is vital to prevent inflammatory disease.
Collapse
Affiliation(s)
- Matthijs J.D. Baars
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Thera Douma
- Center of Translational ImmunologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Dimitre R. Simeonov
- Diabetes CenterUniversity of California San FranciscoSan FranciscoCAUSA
- Biomedical Sciences Graduate ProgramUniversity of California San FranciscoSan FranciscoCAUSA
| | - Darienne R. Myers
- Biomedical Sciences Graduate ProgramUniversity of California San FranciscoSan FranciscoCAUSA
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Kayla Kulhanek
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Saikat Banerjee
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Susan Zwakenberg
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Marijke P. Baltissen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode InstituteRadboud University NijmegenNijmegenThe Netherlands
| | - Mojtaba Amini
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Sytze de Roock
- Pediatric Immunology and Rheumatology, Wilhelmina Children's HospitalUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Femke van Wijk
- Center of Translational ImmunologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Pediatric Immunology and Rheumatology, Wilhelmina Children's HospitalUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode InstituteRadboud University NijmegenNijmegenThe Netherlands
| | - Alexander Marson
- Diabetes CenterUniversity of California San FranciscoSan FranciscoCAUSA
- J. David Gladstone InstitutesSan FranciscoCAUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCAUSA
- Department of Microbiology and ImmunologyUniversity of CaliforniaSan FranciscoCAUSA
| | - Jeroen P. Roose
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Yvonne Vercoulen
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
9
|
Nras Q61R/+ and Kras-/- cooperate to downregulate Rasgrp1 and promote lympho-myeloid leukemia in early T-cell precursors. Blood 2021; 137:3259-3271. [PMID: 33512434 PMCID: PMC8351901 DOI: 10.1182/blood.2020009082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
Kras−/−; NrasQ61R/+ mice develop early onset of T-cell malignancy that recapitulates many biological and molecular features of human ETP-ALL. We identify Rasgrp1 as a negative regulator of Ras/ERK signaling in oncogenic Nras-driven ETP-like leukemia.
Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) is an aggressive subtype of T-cell ALL. Although genetic mutations hyperactivating cytokine receptor/Ras signaling are prevalent in ETP-ALL, it remains unknown how activated Ras signaling contributes to ETP-ALL. Here, we find that in addition to the frequent oncogenic RAS mutations, wild-type (WT) KRAS transcript level was significantly downregulated in human ETP-ALL cells. Similarly, loss of WT Kras in NrasQ61R/+ mice promoted hyperactivation of extracellular signal-regulated kinase (ERK) signaling, thymocyte hyperproliferation, and expansion of the ETP compartment. Kras−/−; NrasQ61R/+ mice developed early onset of T-cell malignancy that recapitulates many biological and molecular features of human ETP-ALL. Mechanistically, RNA-sequencing analysis and quantitative proteomics study identified that Rasgrp1, a Ras guanine nucleotide exchange factor, was greatly downregulated in mouse and human ETP-ALL. Unexpectedly, hyperactivated Nras/ERK signaling suppressed Rasgrp1 expression and reduced Rasgrp1 level led to increased ERK signaling, thereby establishing a positive feedback loop to augment Nras/ERK signaling and promote cell proliferation. Corroborating our cell line data, Rasgrp1 haploinsufficiency induced Rasgrp1 downregulation and increased phosphorylated ERK level and ETP expansion in NrasQ61R/+ mice. Our study identifies Rasgrp1 as a negative regulator of Ras/ERK signaling in oncogenic Nras-driven ETP-like leukemia.
Collapse
|
10
|
Strength of tonic T cell receptor signaling instructs T follicular helper cell-fate decisions. Nat Immunol 2020; 21:1384-1396. [PMID: 32989327 PMCID: PMC7578106 DOI: 10.1038/s41590-020-0781-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022]
Abstract
T follicular helper (TFH) cells are critical in adaptive immune responses to pathogens and vaccines; however, what drives the initiation of their developmental program remains unclear. Studies suggest that a T cell antigen receptor (TCR)-dependent mechanism may be responsible for the earliest TFH cell-fate decision, but a critical aspect of the TCR has been overlooked: tonic TCR signaling. We hypothesized that tonic signaling influences early TFH cell development. Here, two murine TCR-transgenic CD4+ T cells, LLO56 and LLO118, which recognize the same antigenic peptide presented on major histocompatibility complex molecules but experience disparate strengths of tonic signaling, revealed low tonic signaling promotes TFH cell differentiation. Polyclonal T cells paralleled these findings, with naive Nur77 expression distinguishing TFH cell potential. Two mouse lines were also generated to both increase and decrease tonic signaling strength, directly establishing an inverse relationship between tonic signaling strength and TFH cell development. Our findings elucidate a central role for tonic TCR signaling in early TFH cell-lineage decisions.
Collapse
|
11
|
Myers DR, Norlin E, Vercoulen Y, Roose JP. Active Tonic mTORC1 Signals Shape Baseline Translation in Naive T Cells. Cell Rep 2020; 27:1858-1874.e6. [PMID: 31067469 PMCID: PMC6593126 DOI: 10.1016/j.celrep.2019.04.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 01/25/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Naive CD4+ T cells are an example of dynamic cell homeostasis: T cells need to avoid autoreactivity while constantly seeing self-peptides, yet they must be primed to react to foreign antigens during infection. The instructive signals that balance this primed yet quiescent state are unknown. Interactions with self-peptides result in membrane-proximal, tonic signals in resting T cells. Here we reveal selective and robust tonic mTORC1 signals in CD4+ T cells that influence T cell fate decisions. We find that the Ras exchange factor Rasgrp1 is necessary to generate tonic mTORC1 signals. Genome-wide ribosome profiling of resting, primary CD4+ T cells uncovers a baseline translational landscape rich in mTOR targets linked to mitochondria, oxidative phosphorylation, and splicing. Aberrantly increased tonic mTORC1 signals from a Rasgrp1Anaef allele result in immunopathology with spontaneous appearance of T peripheral helper cells, follicular helper T cells, and anti-nuclear antibodies that are preceded by subtle alterations in the translational landscape. Myers et al. evaluate a mouse model of autoimmunity, Rasgrp1Anaef. They find that T cells with the Rasgrp1Anaef allele exhibit altered signaling from Rasgrp1 to the mTORC1 pathway in the basal state. They show that increased basal Rasgrp1Anaef-mTORC1 signals lead to an altered translational landscape in T cells and immunopathology.
Collapse
Affiliation(s)
- Darienne R Myers
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emilia Norlin
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yvonne Vercoulen
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
12
|
Hamatani H, Eng DG, Hiromura K, Pippin JW, Shankland SJ. CD44 impacts glomerular parietal epithelial cell changes in the aged mouse kidney. Physiol Rep 2020; 8:e14487. [PMID: 32597007 PMCID: PMC7322268 DOI: 10.14814/phy2.14487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
CD44 contributes to the activation of glomerular parietal epithelial cells (PECs). Although CD44 expression is higher in PECs of healthy aged mice, the biological role of CD44 in PECs in this context remains unclear. Accordingly, young (4 months) and aged (24 months) CD44-/- mice were compared to age-matched CD44+/+ mice, both aged in a nonstressed environment. Parietal epithelial cell densities were similar in both young and aged CD44+/+ and CD44-/- mice. Phosphorylated ERK 1/2 (pERK) was higher in aged CD44+/+ mice. Vimentin and α-SMA, markers of changes to the epithelial cell phenotype, were present in PECs in aged CD44+/+ mice, but absent in aged CD44-/- mice in both outer cortical (OC) and juxtamedullary (JM) glomeruli. Because age-related glomerular hypertrophy was lower in CD44-/- mice, mTOR activation was assessed by phospho-S6 ribosomal protein (pS6RP) staining. Parietal epithelial cells and glomerular tuft staining for pS6RP was lower in aged CD44-/- mice compared to aged CD44+/+ mice. Podocyte density was higher in aged CD44-/- mice in both OC and JM glomeruli. These changes were accompanied by segmental and global glomerulosclerosis in aged CD44+/+ mice, but absent in aged CD44-/- mice. These results show that the increase in CD44 in PECs in aged kidneys contributes to several changes to the glomerulus during healthy aging in mice, and may involve ERK and mTOR activation.
Collapse
Affiliation(s)
- Hiroko Hamatani
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Diana G. Eng
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Jeffrey W. Pippin
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Stuart J. Shankland
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
13
|
Huang H, Long L, Zhou P, Chapman NM, Chi H. mTOR signaling at the crossroads of environmental signals and T-cell fate decisions. Immunol Rev 2020; 295:15-38. [PMID: 32212344 PMCID: PMC8101438 DOI: 10.1111/imr.12845] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/19/2020] [Indexed: 12/28/2022]
Abstract
The evolutionarily conserved serine/threonine kinase mTOR (mechanistic target of rapamycin) forms the distinct protein complexes mTORC1 and mTORC2 and integrates signals from the environment to coordinate downstream signaling events and various cellular processes. T cells rely on mTOR activity for their development and to establish their homeostasis and functional fitness. Here, we review recent progress in our understanding of the upstream signaling and downstream targets of mTOR. We also provide an updated overview of the roles of mTOR in T-cell development, homeostasis, activation, and effector-cell fate decisions, as well as its important impacts on the suppressive activity of regulatory T cells. Moreover, we summarize the emerging roles of mTOR in T-cell exhaustion and transdifferentiation. A better understanding of the contribution of mTOR to T-cell fate decisions will ultimately aid in the therapeutic targeting of mTOR in human disease.
Collapse
Affiliation(s)
- Hongling Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Lingyun Long
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Equal contribution
| | - Peipei Zhou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Equal contribution
| | - Nicole M. Chapman
- 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
|
14
|
Yahaya T, Salisu T. Genes predisposing to type 1 diabetes mellitus and pathophysiology: a narrative review. MEDICAL JOURNAL OF INDONESIA 2020; 29:100-9. [DOI: 10.13181/mji.rev.203732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 01/23/2020] [Indexed: 02/08/2023] Open
Abstract
The possibility of targeting the causal genes along with the mechanisms of pathogenically complex diseases has led to numerous studies on the genetic etiology of some diseases. In particular, studies have added more genes to the list of type 1 diabetes mellitus (T1DM) suspect genes, necessitating an update for the interest of all stakeholders. Therefore this review articulates T1DM suspect genes and their pathophysiology. Notable electronic databases, including Medline, Scopus, PubMed, and Google-Scholar were searched for relevant information. The search identified over 73 genes suspected in the pathogenesis of T1DM, with human leukocyte antigen, insulin gene, and cytotoxic T lymphocyte-associated antigen 4 accounting for most of the cases. Mutations in these genes, along with environmental factors, may produce a defective immune response in the pancreas, resulting in β-cell autoimmunity, insulin deficiency, and hyperglycemia. The mechanisms leading to these cellular reactions are gene-specific and, if targeted in diabetic individuals, may lead to improved treatment. Medical practitioners are advised to formulate treatment procedures that target these genes in patients with T1DM.
Collapse
|
15
|
Jia Y, Yang Q, Wang Y, Li W, Chen X, Xu T, Tian Z, Feng M, Zhang L, Tang W, Tian N, Zhou L, Song W, Zhao X. Hyperactive PI3Kδ predisposes naive T cells to activation via aerobic glycolysis programs. Cell Mol Immunol 2020; 18:1783-1797. [PMID: 32099075 DOI: 10.1038/s41423-020-0379-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 01/15/2023] Open
Abstract
Activated phosphoinositide 3-kinase δ syndrome (APDS) is an autosomal-dominant combined immunodeficiency disorder resulting from pathogenic gain-of-function (GOF) mutations in the PIK3CD gene. Patients with APDS display abnormal T cell homeostasis. However, the mechanisms by which PIK3CD GOF contributes to this feature remain unknown. Here, with a cohort of children with PIK3CD GOF mutations from multiple regions of China and a corresponding CRISPR/Cas9 gene-edited mouse model, we reported that hyperactive PI3Kδ disrupted TNaive cell homeostasis in the periphery by intrinsically promoting the growth, proliferation, and activation of TNaive cells. Our results showed that PIK3CD GOF resulted in loss of the quiescence-associated gene expression profile in naive T cells and promoted naive T cells to overgrow, hyperproliferate and acquire an activated functional status. Naive PIK3CD GOF T cells exhibited an enhanced glycolytic capacity and reduced mitochondrial respiration in the resting or activated state. Blocking glycolysis abrogated the abnormal splenic T cell pool and reversed the overactivated phenotype induced by PIK3CD GOF in vivo and in vitro. These results suggest that enhanced aerobic glycolysis is required for PIK3CD GOF-induced overactivation of naive T cells and provide a potential therapeutic approach for targeting glycolysis to treat patients with APDS as well as other immune disorders.
Collapse
Affiliation(s)
- Yanjun Jia
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qiuyun Yang
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanping Wang
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenyan Li
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Xu
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhirui Tian
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Minxuan Feng
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Zhang
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenjing Tang
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Na Tian
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Zhou
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Xiaodong Zhao
- National Clinical Research for Child Health and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Myers DR, Wheeler B, Roose JP. mTOR and other effector kinase signals that impact T cell function and activity. Immunol Rev 2020; 291:134-153. [PMID: 31402496 DOI: 10.1111/imr.12796] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/11/2019] [Indexed: 12/27/2022]
Abstract
T cells play important roles in autoimmune diseases and cancer. Following the cloning of the T cell receptor (TCR), the race was on to map signaling proteins that contributed to T cell activation downstream of the TCR as well as co-stimulatory molecules such as CD28. We term this "canonical TCR signaling" here. More recently, it has been appreciated that T cells need to accommodate increased metabolic needs that stem from T cell activation in order to function properly. A central role herein has emerged for mechanistic/mammalian target of rapamycin (mTOR). In this review we briefly cover canonical TCR signaling to set the stage for discussion on mTOR signaling, mRNA translation, and metabolic adaptation in T cells. We also discuss the role of mTOR in follicular helper T cells, regulatory T cells, and other T cell subsets. Our lab recently uncovered that "tonic signals", which pass through proximal TCR signaling components, are robustly and selectively transduced to mTOR to promote baseline translation of various mRNA targets. We discuss insights on (tonic) mTOR signaling in the context of T cell function in autoimmune diseases such as lupus as well as in cancer immunotherapy through CAR-T cell or checkpoint blockade approaches.
Collapse
Affiliation(s)
- Darienne R Myers
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin Wheeler
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
17
|
Klemm P, Rajendiran A, Fragoulis A, Wruck C, Schippers A, Wagner N, Bopp T, Tenbrock K, Ohl K. Nrf2 expression driven by Foxp3 specific deletion of Keap1 results in loss of immune tolerance in mice. Eur J Immunol 2020; 50:515-524. [PMID: 31840803 DOI: 10.1002/eji.201948285] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/26/2019] [Accepted: 12/13/2019] [Indexed: 12/23/2022]
Abstract
The transcription factor Nrf2 regulates oxidative stress responses. However, the specific function of Nrf2 in Tregs, the central regulators of immune homeostasis, is unclear. Here, we report an unexpected but important role of Nrf2 in Tregs. Nrf2 expression driven by Foxp3 specific deletion of Keap1 resulted in an autoinflammatory phenotype with enhanced effector T cell activation and immune cell infiltrates in the lung. While early postnatal death of mice with Foxp3 specific deletion of Keap1 was most probably due to ectopic Foxp3cre expression and subsequent Keap1 deletion in epithelial cells, bone marrow chimeras suggest that Nrf2 activation intrinsically in Tregs contributes to a loss of Treg cells and diminished peripheral tolerance. Moreover, Nrf2 activation was associated with a loss of Foxp3 expression, but an enhanced glucose uptake and mTOR activity in Tregs, thus mimicking a metabolic phenotype that is associated with impaired lineage stability and cell functioning.
Collapse
Affiliation(s)
- Patricia Klemm
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | | | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Christoph Wruck
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Angela Schippers
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Norbert Wagner
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Tobias Bopp
- Institute of Immunology, University Medical Center, The Johannes Gutenberg University Mainz, Mainz, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Kim Ohl
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Aachen, Germany
| |
Collapse
|
18
|
Gbenedio OM, Bonnans C, Grun D, Wang CY, Hatch AJ, Mahoney MR, Barras D, Matli M, Miao Y, Garcia KC, Tejpar S, Delorenzi M, Venook AP, Nixon AB, Warren RS, Roose JP, Depeille P. RasGRP1 is a potential biomarker to stratify anti-EGFR therapy response in colorectal cancer. JCI Insight 2019; 5:127552. [PMID: 31237864 DOI: 10.1172/jci.insight.127552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is the third most frequent neoplastic disorder and is a main cause of tumor-related mortality as many patients progress to stage IV metastatic CRC. Standard care consists of combination chemotherapy (FOLFIRI or FOLFOX). Patients with WT KRAS typing are eligible to receive anti-EGFR therapy combined with chemotherapy. Unfortunately, predicting efficacy of CRC anti-EGFR therapy has remained challenging. Here we uncover that the EGFR-pathway component RasGRP1 acts as CRC tumor suppressor in the context of aberrant Wnt signaling. We find that RasGRP1 suppresses EGF-driven proliferation of colonic epithelial organoids. Having established that RasGRP1 dosage levels impacts biology, we focused on CRC patients next. Mining five different data platforms, we establish that RasGRP1 expression levels decrease with CRC progression and predict poor clinical outcome of patients. Lastly, deletion of one or two Rasgrp1 alleles makes CRC spheroids more susceptible to EGFR inhibition. Retrospective analysis of the CALGB80203 clinical trial shows that addition of anti-EGFR therapy to chemotherapy significantly improves outcome for CRC patients when tumors express low RasGRP1 suppressor levels. In sum, RasGRP1 is a unique biomarker positioned in the EGFR pathway and of potential relevance to anti-EGFR therapy for CRC patients.
Collapse
Affiliation(s)
| | - Caroline Bonnans
- Department of Anatomy, UCSF, San Francisco, California, USA.,Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Delphine Grun
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Chih-Yang Wang
- Department of Anatomy, UCSF, San Francisco, California, USA
| | - Ace J Hatch
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Michelle R Mahoney
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | - David Barras
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mary Matli
- Department of Surgery, UCSF, San Francisco, California, USA
| | - Yi Miao
- Department of Molecular and Cellular Physiology, Department of Structural Biology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Department of Structural Biology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Sabine Tejpar
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Mauro Delorenzi
- Bioinformatics Core Facility (BCF) at SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Alan P Venook
- Hematology/Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Andrew B Nixon
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jeroen P Roose
- Department of Anatomy, UCSF, San Francisco, California, USA
| | | |
Collapse
|
19
|
Gruenbacher G, Thurnher M. Mevalonate Metabolism in Cancer Stemness and Trained Immunity. Front Oncol 2018; 8:394. [PMID: 30298120 PMCID: PMC6160868 DOI: 10.3389/fonc.2018.00394] [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: 07/09/2018] [Accepted: 08/31/2018] [Indexed: 12/28/2022] Open
Abstract
Mevalonate metabolism provides cancer and immune cells with diverse products to ensure cell functionality. Similar metabolic reprogramming that raises mevalonate metabolism to higher levels appears to drive both, epithelial mesenchymal transition (EMT) of cancer cells, a reverse differentiation program that generates cancer cells with stem cell properties, and immune cell training for increased responsiveness to secondary stimulation. In this review, we address how mevalonate metabolism supports cancer development and stemness on the one hand, and on the other promotes immune responsiveness. In view of this dual nature of mevalonate metabolism, strategies to manipulate this metabolic pathway as part of anti-cancer therapies require careful analysis of risks versus benefits.
Collapse
Affiliation(s)
- Georg Gruenbacher
- Immunotherapy Research Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Research Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
RASGRP1 mutation in autoimmune lymphoproliferative syndrome-like disease. J Allergy Clin Immunol 2018; 142:595-604.e16. [DOI: 10.1016/j.jaci.2017.10.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 12/18/2022]
|
21
|
Latour S, Winter S. Inherited Immunodeficiencies With High Predisposition to Epstein-Barr Virus-Driven Lymphoproliferative Diseases. Front Immunol 2018; 9:1103. [PMID: 29942301 PMCID: PMC6004768 DOI: 10.3389/fimmu.2018.01103] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/02/2018] [Indexed: 01/16/2023] Open
Abstract
Epstein–Barr Virus (EBV) is a gamma-herpes virus that infects 90% of humans without any symptoms in most cases, but has an oncogenic potential, especially in immunocompromised individuals. In the past 30 years, several primary immunodeficiencies (PIDs) associated with a high risk to develop EBV-associated lymphoproliferative disorders (LPDs), essentially consisting of virus-associated hemophagocytic syndrome, non-malignant and malignant B-cell LPDs including non-Hodgkin and Hodgkin’s types of B lymphomas have been characterized. Among them are SH2D1A (SAP), XIAP, ITK, MAGT1, CD27, CD70, CTPS1, RASGRP1, and CORO1A deficiencies. Penetrance of EBV infection ranges from 50 to 100% in those PIDs. Description of large cohorts and case reports has refined the specific phenotypes associated with these PIDs helping to the diagnosis. Specific pathways required for protective immunity to EBV have emerged from studies of these PIDs. SLAM-associated protein-dependent SLAM receptors and MAGT1-dependent NKG2D pathways are important for T and NK-cell cytotoxicity toward EBV-infected B-cells, while CD27–CD70 interactions are critical to drive the expansion of EBV-specific T-cells. CTPS1 and RASGRP1 deficiencies further strengthen that T-lymphocyte expansion is a key step in the immune response to EBV. These pathways appear to be also important for the anti-tumoral immune surveillance of abnormal B cells. Monogenic PIDs should be thus considered in case of any EBV-associated LPDs.
Collapse
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| | - Sarah Winter
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| |
Collapse
|
22
|
Kielbinski M, Setkowicz Z, Gzielo K, Janeczko K. Profiles of gene expression in the hippocampal formation of rats with experimentally-induced brain dysplasia. Dev Neurobiol 2018; 78:718-735. [DOI: 10.1002/dneu.22595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/28/2018] [Accepted: 04/06/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Michal Kielbinski
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Kinga Gzielo
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Krzysztof Janeczko
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| |
Collapse
|
23
|
Liu YH, Corbett C, Klaska IP, Makinen K, Nickerson JM, Cornall RJ, Kuffova L, Forrester JV. Partial retinal photoreceptor loss in a transgenic mouse model associated with reduced levels of interphotoreceptor retinol binding protein (IRBP, RBP3). Exp Eye Res 2018; 172:54-65. [PMID: 29571629 DOI: 10.1016/j.exer.2018.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Organ-specific transgenic membrane expression of hen egg lysozyme (HEL) as a "neo-self antigen" has been used in several models to study immunological tolerance. In this study we report the changes which occur in the B10.BR mouse retina when membrane-bound HEL is expressed in photoreceptors under the control of the promoter for interphotoreceptor retinoid binding protein (IRBP, RBP3). On direct clinical examination of the single transgenic (sTg-IRBP:HEL) mouse fundus, a low-level increase in retinal degeneration compared to non-transgenic controls was observed, presenting as drusenoid deposits and occasional small patches of atrophy. On histological examination, there was an overall shortening of outer segments and loss of photoreceptor nuclei in sTg-IRBP:HEL mice, which was more pronounced in the retinal periphery, particularly inferiorly. The fundoscopically observed lesions did not correlate with the photoreceptor shortening/loss but appeared to be located at the level of the retinal pigment epithelium/choriocapillaris layer and were an exaggeration in size and number of similar age-related changes found in wild type (WT) mice. In addition, neither the atrophic lesions nor the photoreceptor shortening were associated with common retinal degeneration genes, nor were they caused by exposure to light damage since mice housed at both high and low ambient light levels had similar degrees of retinal degeneration. Instead, sTg-IRBP:HEL mice expressed reduced levels of soluble retinal IRBP compared to WT mice which were present from postnatal day16 (P16) and preceded development of photoreceptor shortening (onset P21). We propose that insertion of the HEL transgene in the photoreceptor membrane disrupted normal photoreceptor function and led to reduced levels of soluble IRBP and retinal thinning. A similar phenotype has been observed in IRBP deficient mice. Despite the retinal thinning, the amount of HEL expressed in the retina was sufficient to act as an autoantigenic target when the mice were crossed to the HEL T cell receptor Tg mouse, since double transgenic (dTg-IRBP:HEL) mice spontaneously developed a severe uveoretinitis with onset at weaning. We suggest that, although membrane expression of foreign transgene products is likely to modify the structure and function of tissues and cells, the technology provides useful models to investigate mechanisms of antigen-specific immunological tolerance.
Collapse
Affiliation(s)
- Yi-Hsia Liu
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Clare Corbett
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Izabela P Klaska
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Institute of Ophthalmology, University College London, London, UK
| | - Kimmo Makinen
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Human Health, Novozymes A/S, Bagsvaerd, Denmark
| | | | | | - Lucia Kuffova
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Department of Ophthalmology, NHS Grampian, Aberdeen, UK
| | - John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; University of Western Australia, Lions Eye Institute, Perth, Western Australia, Australia.
| |
Collapse
|
24
|
SNX27 links DGKζ to the control of transcriptional and metabolic programs in T lymphocytes. Sci Rep 2017; 7:16361. [PMID: 29180720 PMCID: PMC5703713 DOI: 10.1038/s41598-017-16370-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/10/2017] [Indexed: 01/10/2023] Open
Abstract
Sorting nexin 27 (SNX27) recycles PSD-95, Dlg1, ZO-1 (PDZ) domain-interacting membrane proteins and is essential to sustain adequate brain functions. Here we define a fundamental SNX27 function in T lymphocytes controlling antigen-induced transcriptional activation and metabolic reprogramming. SNX27 limits the activation of diacylglycerol (DAG)-based signals through its high affinity PDZ-interacting cargo DAG kinase ζ (DGKζ). SNX27 silencing in human T cells enhanced T cell receptor (TCR)-stimulated activator protein 1 (AP-1)- and nuclear factor κB (NF-κB)-mediated transcription. Transcription did not increase upon DGKζ silencing, suggesting that DGKζ function is dependent on SNX27. The enhanced transcriptional activation in SNX27-silenced cells contrasted with defective activation of the mammalian target of rapamycin (mTOR) pathway. The analysis of Snx27−/− mice supported a role for SNX27 in the control of T cell growth. This study broadens our understanding of SNX27 as an integrator of lipid-based signals with the control of transcription and metabolic pathways.
Collapse
|
25
|
Metabolic pressure and the breach of immunological self-tolerance. Nat Immunol 2017; 18:1190-1196. [DOI: 10.1038/ni.3851] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022]
|
26
|
Vercoulen Y, Kondo Y, Iwig JS, Janssen AB, White KA, Amini M, Barber DL, Kuriyan J, Roose JP. A Histidine pH sensor regulates activation of the Ras-specific guanine nucleotide exchange factor RasGRP1. eLife 2017; 6:29002. [PMID: 28952923 PMCID: PMC5643099 DOI: 10.7554/elife.29002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/05/2017] [Indexed: 02/04/2023] Open
Abstract
RasGRPs are guanine nucleotide exchange factors that are specific for Ras or Rap, and are important regulators of cellular signaling. Aberrant expression or mutation of RasGRPs results in disease. An analysis of RasGRP1 SNP variants led to the conclusion that the charge of His 212 in RasGRP1 alters signaling activity and plasma membrane recruitment, indicating that His 212 is a pH sensor that alters the balance between the inactive and active forms of RasGRP1. To understand the structural basis for this effect we compared the structure of autoinhibited RasGRP1, determined previously, to those of active RasGRP4:H-Ras and RasGRP2:Rap1b complexes. The transition from the autoinhibited to the active form of RasGRP1 involves the rearrangement of an inter-domain linker that displaces inhibitory inter-domain interactions. His 212 is located at the fulcrum of these conformational changes, and structural features in its vicinity are consistent with its function as a pH-dependent switch.
Collapse
Affiliation(s)
- Yvonne Vercoulen
- Department of Anatomy, University of California, San Francisco, San Francisco, United States.,Molecular Cancer Research, Center for Molecular Medicine, UMC Utrecht, Utrecht University, Utrecht, Netherlands
| | - Yasushi Kondo
- Department of Molecular and Cell Biology and Chemistry, University of California, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, United States
| | - Jeffrey S Iwig
- Department of Molecular and Cell Biology and Chemistry, University of California, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, United States
| | - Axel B Janssen
- Department of Anatomy, University of California, San Francisco, San Francisco, United States
| | - Katharine A White
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Mojtaba Amini
- Molecular Cancer Research, Center for Molecular Medicine, UMC Utrecht, Utrecht University, Utrecht, Netherlands
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - John Kuriyan
- Department of Molecular and Cell Biology and Chemistry, University of California, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, United States.,Department of Chemistry, University of California, Berkeley, United States.,Divisions of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, United States
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
27
|
Myers DR, Zikherman J, Roose JP. Tonic Signals: Why Do Lymphocytes Bother? Trends Immunol 2017; 38:844-857. [PMID: 28754596 DOI: 10.1016/j.it.2017.06.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 01/09/2023]
Abstract
Since the 1990s it has been known that B and T lymphocytes exhibit low-level, constitutive signaling in the basal state (tonic signaling). These lymphocytes display a range of affinity for self, which in turn generates a range of tonic signaling. Surprisingly, what signaling pathways are active in the basal state and the functional relevance of the observed tonic signaling heterogeneity remain open questions today. Here we summarize what is known about the mechanistic and functional details of tonic signaling. We highlight recent advances that have increased our understanding of how the amount of tonic signal impacts immune function, describing novel tools that have moved the field forward and toward a molecular understanding of tonic signaling.
Collapse
Affiliation(s)
- Darienne R Myers
- Department of Anatomy, School of Medicine, University of California, San Francisco, CA 94143, USA
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Jeroen P Roose
- Department of Anatomy, School of Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
28
|
Zeng H, Chi H. mTOR signaling in the differentiation and function of regulatory and effector T cells. Curr Opin Immunol 2017; 46:103-111. [PMID: 28535458 DOI: 10.1016/j.coi.2017.04.005] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 11/17/2022]
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway integrates environmental signals and cellular metabolism to regulate T cell development, activation and differentiation. Recent studies reveal the importance of exquisite control of mTOR activity for proper T cell function, and detailed molecular mechanisms that regulate mTOR signaling in different T cell subsets. Here, we review the latest advances in our understanding of the mTOR pathway and its regulation in the differentiation and function of regulatory T cells and effector T cells.
Collapse
Affiliation(s)
- Hu Zeng
- 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
|
29
|
Andrada E, Liébana R, Merida I. Diacylglycerol Kinase ζ Limits Cytokine-dependent Expansion of CD8 + T Cells with Broad Antitumor Capacity. EBioMedicine 2017; 19:39-48. [PMID: 28438506 PMCID: PMC5440620 DOI: 10.1016/j.ebiom.2017.04.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/03/2017] [Accepted: 04/12/2017] [Indexed: 11/18/2022] Open
Abstract
Interleukin-2 and -15 drive expansion/differentiation of cytotoxic CD8+ T cells that eliminate targets via antigen-independent killing. This property is clinically relevant for the improvement of T cell-based antitumor therapies. Diacylglycerol kinase α and ζ (DGKα/ζ) metabolize the diacylglycerol generated following antigen recognition by T lymphocytes. Enhanced expression of these two lipid kinases in tumor-infiltrating CD8+ T cells promotes a hyporesponsive state that contributes to tumor immune escape. Inhibition of these two enzymes might thus be of interest for potentiating conventional antigen-directed tumor elimination. In this study, we sought to characterize the contribution of DGKα and ζ to antigen-independent cytotoxic functions of CD8+ T cells. Analysis of DGKζ-deficient mice showed an increase in bystander memory-like CD8+ T cell populations not observed in DGKα-deficient mice. We demonstrate that DGKζ limits cytokine responses in an antigen-independent manner. Cytokine-specific expansion of DGKζ-deficient CD8+ T cells promoted enhanced differentiation of innate-like cytotoxic cells in vitro, and correlated with the more potent in vivo anti-tumor responses of DGKζ-deficient mice engrafted with the murine A20 lymphoma. Our studies reveal a isoform-specific function for DGKζ downstream of IL-2/IL-15-mediated expansion of innate-like cytotoxic T cells, Pharmacological manipulation of DGKζ activity is of therapeutic interest for cytokine-directed anti-tumor treatments. DGKζ, a well-characterized negative regulator of TCR signals, also limits IL-2/15 function. DGKζ impairs cytokine-induced differentiation of cytotoxic T cell populations with innate-like ability to kill targets. As a result, DGKζ-deficient mice demonstrate enhanced rejection of implanted B cell lymphoma compared to wild type mice. Targeting DGKζ activity might be of interest to enhance cytokine-mediated antitumor therapies.
The immune system defends the body from foreign invaders. In cancer, tumors disguise as self-body cells and evade immune attack. For this reason it is important to identify the mechanism that stop T lymphocytes from recognize and destroy tumors. In this study we investigate the role of Diacylglycerol kinase zeta (DGKζ) as an inhibitor of antitumor T cell functions. We demonstrate that lymphoma cells injected in mice genetically modified to lack DGKζ expression develop smaller tumors that resolve more rapidly than those grown in normal mice. Our studies suggest that inhibition of DGKζ could help to reinforce the antitumor capacity of immune T lymphocytes.
Collapse
Affiliation(s)
- Elena Andrada
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), E-28049 Madrid, Spain
| | - Rosa Liébana
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), E-28049 Madrid, Spain
| | - Isabel Merida
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), E-28049 Madrid, Spain.
| |
Collapse
|
30
|
Predominant contribution of DGKζ over DGKα in the control of PKC/PDK‐1‐regulated functions in T cells. Immunol Cell Biol 2017; 95:549-563. [DOI: 10.1038/icb.2017.7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/16/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
|
31
|
Affiliation(s)
- Jeroen P Roose
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
32
|
Christensen SM, Tu HL, Jun JE, Alvarez S, Triplet MG, Iwig JS, Yadav KK, Bar-Sagi D, Roose JP, Groves JT. One-way membrane trafficking of SOS in receptor-triggered Ras activation. Nat Struct Mol Biol 2016; 23:838-46. [PMID: 27501536 PMCID: PMC5016256 DOI: 10.1038/nsmb.3275] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023]
Abstract
SOS is a key activator of the small GTPase Ras. In cells, SOS-Ras signaling is thought to be initiated predominantly by membrane recruitment of SOS via the adaptor Grb2 and balanced by rapidly reversible Grb2-SOS binding kinetics. However, SOS has multiple protein and lipid interactions that provide linkage to the membrane. In reconstituted-membrane experiments, these Grb2-independent interactions were sufficient to retain human SOS on the membrane for many minutes, during which a single SOS molecule could processively activate thousands of Ras molecules. These observations raised questions concerning how receptors maintain control of SOS in cells and how membrane-recruited SOS is ultimately released. We addressed these questions in quantitative assays of reconstituted SOS-deficient chicken B-cell signaling systems combined with single-molecule measurements in supported membranes. These studies revealed an essentially one-way trafficking process in which membrane-recruited SOS remains trapped on the membrane and continuously activates Ras until being actively removed via endocytosis.
Collapse
Affiliation(s)
- Sune M. Christensen
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Hsiung-Lin Tu
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Jesse E. Jun
- Department of Anatomy, University of California, San Francisco, California, USA
| | - Steven Alvarez
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Meredith G. Triplet
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Jeffrey S. Iwig
- Howard Hughes Medical Institute, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Kamlesh K. Yadav
- Department of Biochemistry, New York University School of Medicine, New York, USA
| | - Dafna Bar-Sagi
- Department of Biochemistry, New York University School of Medicine, New York, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, California, USA
| | - Jay T. Groves
- Department of Chemistry, University of California, Berkeley, California, USA
| |
Collapse
|
33
|
Ksionda O, Melton AA, Bache J, Tenhagen M, Bakker J, Harvey R, Winter SS, Rubio I, Roose JP. RasGRP1 overexpression in T-ALL increases basal nucleotide exchange on Ras rendering the Ras/PI3K/Akt pathway responsive to protumorigenic cytokines. Oncogene 2016; 35:3658-68. [PMID: 26549032 PMCID: PMC4868787 DOI: 10.1038/onc.2015.431] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/31/2015] [Accepted: 10/05/2015] [Indexed: 12/25/2022]
Abstract
Ras GTPases are activated by RasGEFs and inactivated by RasGAPs, which stimulate the hydrolysis of RasGTP to inactive RasGDP. GTPase-impairing somatic mutations in RAS genes, such as KRAS(G12D), are among the most common oncogenic events in metastatic cancer. A different type of cancer Ras signal, driven by overexpression of the RasGEF RasGRP1 (Ras guanine nucleotide-releasing protein 1), was recently implicated in pediatric T-cell acute lymphoblastic leukemia (T-ALL) patients and murine models, in which RasGRP1 T-ALLs expand in response to treatment with interleukins (ILs) 2, 7 and 9. Here, we demonstrate that IL-2/7/9 stimulation activates Erk and Akt pathways downstream of Ras in RasGRP1 T-ALL but not in normal thymocytes. In normal lymphocytes, RasGRP1 is recruited to the membrane by diacylglycerol (DAG) in a phospholipase C-γ (PLCγ)-dependent manner. Surprisingly, we find that leukemic RasGRP1-triggered Ras-Akt signals do not depend on acute activation of PLCγ to generate DAG but rely on baseline DAG levels instead. In agreement, using three distinct assays that measure different aspects of the RasGTP/GDP cycle, we established that overexpression of RasGRP1 in T-ALLs results in a constitutively high GTP-loading rate of Ras, which is constantly counterbalanced by hydrolysis of RasGTP. KRAS(G12D) T-ALLs do not show constitutive GTP loading of Ras. Thus, we reveal an entirely novel type of leukemogenic Ras signals that is based on a RasGRP1-driven increased in flux through the RasGTP/GDP cycle, which is mechanistically very different from KRAS(G12D) signals. Our studies highlight the dynamic balance between RasGEF and RasGAP in these T-ALLs and put forth a new model in which IL-2/7/9 decrease RasGAP activity.
Collapse
Affiliation(s)
- O Ksionda
- Department of Anatomy, Roose University of California, San Francisco, San Francisco, CA, USA
| | - AA Melton
- Department of Anatomy, Roose University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - J Bache
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - M Tenhagen
- Department of Anatomy, Roose University of California, San Francisco, San Francisco, CA, USA
| | - J Bakker
- Department of Anatomy, Roose University of California, San Francisco, San Francisco, CA, USA
| | - R Harvey
- Department of Pediatrics, University of New Mexico School of Medicine Albuquerque, NM, USA
| | - SS Winter
- Department of Pediatrics, University of New Mexico School of Medicine Albuquerque, NM, USA
| | - I Rubio
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Friedrich-Schiller-University, Jena, Germany
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - JP Roose
- Department of Anatomy, Roose University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
34
|
Smith C, Buhlmann JE, Wang X, Bartlett A, Lim B, Barrington RA. CD275-Independent IL-17-Producing T Follicular Helper-like Cells in Lymphopenic Autoimmune-Prone Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:4935-46. [PMID: 27183569 DOI: 10.4049/jimmunol.1402193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 04/15/2016] [Indexed: 12/27/2022]
Abstract
T cells undergo homeostatic expansion and acquire an activated phenotype in lymphopenic microenvironments. Restoration of normal lymphocyte numbers typically re-establishes normal homeostasis, and proinflammatory cytokine production returns to baseline. Mice deficient in guanine nucleotide exchange factor RasGRP1 exhibit dysregulated homeostatic expansion, which manifests as lymphoproliferative disease with autoantibody production. Our previous work revealed that autoreactive B cells lacking RasGRP1 break tolerance early during development, as well as during germinal center responses, suggesting that T cell-independent and T cell-dependent mechanisms are responsible. Examination of whether a particular T cell subset is involved in the breach of B cell tolerance revealed increased Th17 cells in Rasgrp1-deficient mice relative to control mice. Rasgrp1-deficient mice lacking IL-17R had fewer germinal centers, and germinal centers that formed contained fewer autoreactive B cells, suggesting that IL-17 signaling is required for a break in B cell tolerance in germinal centers. Interestingly, a fraction of Th17 cells from Rasgrp1-deficient mice were CXCR5(+) and upregulated levels of CD278 coordinate with their appearance in germinal centers, all attributes of T follicular helper cells (Tfh17). To determine whether CD278-CD275 interactions were required for the development of Tfh17 cells and for autoantibody, Rasgrp1-deficient mice were crossed with CD275-deficient mice. Surprisingly, mice deficient in RasGRP1 and CD275 formed Tfh17 cells and germinal centers and produced similar titers of autoantibodies as mice deficient in only RasGRP1. Therefore, these studies suggest that requirements for Tfh cell development change in lymphopenia-associated autoimmune settings.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Janet E Buhlmann
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Xiaogan Wang
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Amber Bartlett
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| | - Bing Lim
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115
| | - Robert A Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688; and
| |
Collapse
|
35
|
Guo B, Rothstein TL. RasGRP1 Is an Essential Signaling Molecule for Development of B1a Cells with Autoantigen Receptors. THE JOURNAL OF IMMUNOLOGY 2016; 196:2583-90. [PMID: 26851222 DOI: 10.4049/jimmunol.1502132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/04/2016] [Indexed: 01/08/2023]
Abstract
B1a cells, particularly the PD-L2(+) B1a cell subset, are enriched with autoantigen-specific receptors. However, the underlying molecular mechanism responsible for the skewed selection of autoreactive B1a cells remains unclear. In this study, we find that B1 cells express only Ras guanyl nucleotide-releasing protein (RasGRP) 1, whereas B2 cells express mostly RasGRP3 and little RasGRP1. RasGRP1 is indispensable for transduction of weak signals. RasGRP1 deficiency markedly impairs B1a cell development and reduces serum natural IgM production; in particular, B1a cells that express autoantigen receptors, such as anti-phosphatidylcholine B1a cells, are virtually eliminated. Thus, unlike Btk and other signalosome components, RasGRP1 deficiency selectively affects only the B1a cell population with autoantigen receptors rather than the entire pool of B1a cells.
Collapse
Affiliation(s)
- Benchang Guo
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Manhasset, NY 11030;
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Manhasset, NY 11030; Department of Medicine, Hofstra North Shore-Long Island Jewish School of Medicine, Manhasset, NY 11030; and Department of Molecular Medicine, Hofstra North Shore-Long Island Jewish School of Medicine, Manhasset, NY 11030
| |
Collapse
|
36
|
Ramiscal RR, Parish IA, Lee-Young RS, Babon JJ, Blagih J, Pratama A, Martin J, Hawley N, Cappello JY, Nieto PF, Ellyard JI, Kershaw NJ, Sweet RA, Goodnow CC, Jones RG, Febbraio MA, Vinuesa CG, Athanasopoulos V. Attenuation of AMPK signaling by ROQUIN promotes T follicular helper cell formation. eLife 2015; 4. [PMID: 26496200 PMCID: PMC4716841 DOI: 10.7554/elife.08698] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022] Open
Abstract
T follicular helper cells (Tfh) are critical for the longevity and quality of antibody-mediated protection against infection. Yet few signaling pathways have been identified to be unique solely to Tfh development. ROQUIN is a post-transcriptional repressor of T cells, acting through its ROQ domain to destabilize mRNA targets important for Th1, Th17, and Tfh biology. Here, we report that ROQUIN has a paradoxical function on Tfh differentiation mediated by its RING domain: mice with a T cell-specific deletion of the ROQUIN RING domain have unchanged Th1, Th2, Th17, and Tregs during a T-dependent response but show a profoundly defective antigen-specific Tfh compartment. ROQUIN RING signaling directly antagonized the catalytic α1 subunit of adenosine monophosphate-activated protein kinase (AMPK), a central stress-responsive regulator of cellular metabolism and mTOR signaling, which is known to facilitate T-dependent humoral immunity. We therefore unexpectedly uncover a ROQUIN–AMPK metabolic signaling nexus essential for selectively promoting Tfh responses. DOI:http://dx.doi.org/10.7554/eLife.08698.001 The immune system protects the body from invading microbes like bacteria and viruses. Upon recognizing the presence of these microbes, cells in the immune system are activated to destroy the foreign threat and clear it from the body. A type of immune cell called T follicular helper cells (or Tfh for short) are formed during an infection and are essential for coordinating other immune cells to produce high-quality antibody proteins that attack the microbes. Without Tfh cells, life-long production of these protective antibodies is severely crippled, which can cause common variable immune deficiency and other serious immunodeficiency diseases. On the other hand, the body must also avoid generating excessive numbers of Tfh cells, which can lead to the production of antibodies that attack healthy cells of the body. ROQUIN is a protein that inhibits the formation of Tfh cells and other types of active T cells. A region on the protein called the ROQ domain destabilizes particular molecules of ribonucleic acid (RNA) that are required for these specialist T cells to form and work properly. ROQUIN belongs to a large family of enzymes that have a so-called RING domain, which is a feature that enables these enzymes to attach tags onto specific target proteins to modify their activity or stability. However, it was not known whether the RING domain of ROQUIN was active. Ramiscal et al. now address this question in mice. Unexpectedly, the experiments show that the RING domain is required to promote the formation of Tfh cells, but not other types of active T cells. This domain allows ROQUIN to repress an enzyme called AMPK, which normally blocks cell growth by regulating cell metabolism. The findings suggest that the different roles of the ROQ and RING domains allow ROQUIN to fine-tune the numbers of Tfh cells so that they remain within a safe range. In the future, these findings may aid the development of vaccines that are more efficient at generating protective Tfh cells to prevent infectious diseases. DOI:http://dx.doi.org/10.7554/eLife.08698.002
Collapse
Affiliation(s)
- Roybel R Ramiscal
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Ian A Parish
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Robert S Lee-Young
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Jeffrey J Babon
- Division of Structural Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Julianna Blagih
- Department of Physiology, Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Alvin Pratama
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Jaime Martin
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Naomi Hawley
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Jean Y Cappello
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Pablo F Nieto
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Julia I Ellyard
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Nadia J Kershaw
- Division of Structural Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Rebecca A Sweet
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Christopher C Goodnow
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia.,Immunology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Russell G Jones
- Department of Physiology, Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Vicki Athanasopoulos
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|
37
|
Abstract
Foxp3(+) Tregs are central regulators of immune tolerance. As dysregulated Treg responses contribute to disease pathogenesis, novel approaches to target the immunomodulatory functions of Tregs are currently under investigation. mTORC1 and mTORC2 are therapeutic targets of interest. Recent studies revealed that mTOR signaling impacts conventional T-cell homeostasis, activation and differentiation. Moreover, mTOR controls the differentiation and functions of Tregs, suggesting that its activity could be targeted to modulate Treg responses. Here, we summarize how Tregs suppress immune responses, their roles in disease development and methods used to alter their functions therapeutically. We also discuss the diverse effects exerted by mTOR inhibition on the development, homeostasis, and functions of conventional T cells and Tregs. We conclude with a discussion of how modulation of mTOR activity in Tregs may be therapeutically beneficial or detrimental in different disease settings.
Collapse
Affiliation(s)
- Nicole M Chapman
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
38
|
Depeille P, Henricks LM, van de Ven RAH, Lemmens E, Wang CY, Matli M, Werb Z, Haigis KM, Donner D, Warren R, Roose JP. RasGRP1 opposes proliferative EGFR-SOS1-Ras signals and restricts intestinal epithelial cell growth. Nat Cell Biol 2015; 17:804-15. [PMID: 26005835 PMCID: PMC4652934 DOI: 10.1038/ncb3175] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/08/2015] [Indexed: 02/08/2023]
Abstract
The character of EGFR signals can influence cell fate but mechanistic insights into intestinal EGFR-Ras signalling are limited. Here we show that two distinct Ras nucleotide exchange factors, RasGRP1 and SOS1, lie downstream of EGFR but act in functional opposition. RasGRP1 is expressed in intestinal crypts where it restricts epithelial growth. High RasGRP1 expression in colorectal cancer (CRC) patient samples correlates with a better clinical outcome. Biochemically, we find that RasGRP1 creates a negative feedback loop that limits proliferative EGFR-SOS1-Ras signals in CRC cells. Genetic Rasgrp1 depletion from mice with either an activating mutation in KRas or with aberrant Wnt signalling due to a mutation in Apc resulted in both cases in exacerbated Ras-ERK signalling and cell proliferation. The unexpected opposing cell biological effects of EGFR-RasGRP1 and EGFR-SOS1 signals in the same cell shed light on the intricacy of EGFR-Ras signalling in normal epithelium and carcinoma.
Collapse
Affiliation(s)
- Philippe Depeille
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Linda M. Henricks
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Robert A. H. van de Ven
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Ed Lemmens
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Chih-Yang Wang
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Mary Matli
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kevin M. Haigis
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - David Donner
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Robert Warren
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
39
|
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]
|
40
|
Chapman NM, Chi H. mTOR Links Environmental Signals to T Cell Fate Decisions. Front Immunol 2015; 5:686. [PMID: 25653651 PMCID: PMC4299512 DOI: 10.3389/fimmu.2014.00686] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/20/2014] [Indexed: 12/18/2022] Open
Abstract
T cell fate decisions play an integral role in maintaining the health of organisms under homeostatic and inflammatory conditions. The localized microenvironment in which developing and mature T cells reside provides signals that serve essential functions in shaping these fate decisions. These signals are derived from the immune compartment, including antigens, co-stimulation, and cytokines, and other factors, including growth factors and nutrients. The mechanistic target of rapamycin (mTOR), a vital sensor of signals within the immune microenvironment, is a central regulator of T cell biology. In this review, we discuss how various environmental cues tune mTOR activity in T cells, and summarize how mTOR integrates these signals to influence multiple aspects of T cell biology.
Collapse
Affiliation(s)
- 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
|