1
|
Xiao Z, Wang S, Luo L, Lv W, Feng P, Sun Y, Yang Q, He J, Cao G, Yin Z, Yang M. Lkb1 orchestrates γδ T-cell metabolic and functional fitness to control IL-17-mediated autoimmune hepatitis. Cell Mol Immunol 2024; 21:546-560. [PMID: 38641698 PMCID: PMC11143210 DOI: 10.1038/s41423-024-01163-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/02/2024] [Indexed: 04/21/2024] Open
Abstract
γδ T cells play a crucial role in immune surveillance and serve as a bridge between innate and adaptive immunity. However, the metabolic requirements and regulation of γδ T-cell development and function remain poorly understood. In this study, we investigated the role of liver kinase B1 (Lkb1), a serine/threonine kinase that links cellular metabolism with cell growth and proliferation, in γδ T-cell biology. Our findings demonstrate that Lkb1 is not only involved in regulating γδ T lineage commitment but also plays a critical role in γδ T-cell effector function. Specifically, T-cell-specific deletion of Lkb1 resulted in impaired thymocyte development and distinct alterations in γδ T-cell subsets in both the thymus and peripheral lymphoid tissues. Notably, loss of Lkb1 inhibited the commitment of Vγ1 and Vγ4 γδ T cells, promoted the maturation of IL-17-producing Vγ6 γδ T cells, and led to the occurrence of fatal autoimmune hepatitis (AIH). Notably, clearance of γδ T cells or blockade of IL-17 significantly attenuated AIH. Mechanistically, Lkb1 deficiency disrupted metabolic homeostasis and AMPK activity, accompanied by increased mTORC1 activation, thereby causing overactivation of γδ T cells and enhanced apoptosis. Interestingly, activation of AMPK or suppression of mTORC1 signaling effectively inhibited IL-17 levels and attenuated AIH in Lkb1-deficient mice. Our findings highlight the pivotal role of Lkb1 in maintaining the homeostasis of γδ T cells and preventing IL-17-mediated autoimmune diseases, providing new insights into the metabolic programs governing the subset determination and functional differentiation of thymic γδ T cells.
Collapse
Affiliation(s)
- Zhiqiang Xiao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shanshan Wang
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Liang Luo
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Wenkai Lv
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Peiran Feng
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Yadong Sun
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
| | - Jun He
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. Institute of Laboratory Animal Science, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Guangchao Cao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China.
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China.
- The Biomedical Translational Research Institute, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China.
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. Institute of Laboratory Animal Science, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
2
|
Hu L, Liu M, Tang B, Li Q, Pan BS, Xu C, Lin HK. Posttranslational regulation of liver kinase B1 (LKB1) in human cancer. J Biol Chem 2023; 299:104570. [PMID: 36870679 PMCID: PMC10068580 DOI: 10.1016/j.jbc.2023.104570] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome (PJS) and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies has uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Lanlin Hu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingxin Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Tang
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiang Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
3
|
Werlen G, Jain R, Jacinto E. MTOR Signaling and Metabolism in Early T Cell Development. Genes (Basel) 2021; 12:genes12050728. [PMID: 34068092 PMCID: PMC8152735 DOI: 10.3390/genes12050728] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) controls cell fate and responses via its functions in regulating metabolism. Its role in controlling immunity was unraveled by early studies on the immunosuppressive properties of rapamycin. Recent studies have provided insights on how metabolic reprogramming and mTOR signaling impact peripheral T cell activation and fate. The contribution of mTOR and metabolism during early T-cell development in the thymus is also emerging and is the subject of this review. Two major T lineages with distinct immune functions and peripheral homing organs diverge during early thymic development; the αβ- and γδ-T cells, which are defined by their respective TCR subunits. Thymic T-regulatory cells, which have immunosuppressive functions, also develop in the thymus from positively selected αβ-T cells. Here, we review recent findings on how the two mTOR protein complexes, mTORC1 and mTORC2, and the signaling molecules involved in the mTOR pathway are involved in thymocyte differentiation. We discuss emerging views on how metabolic remodeling impacts early T cell development and how this can be mediated via mTOR signaling.
Collapse
|
4
|
Abstract
During thymocyte development at the double positive stage, thymocytes are subjected to a TCR quality check process termed "thymocyte selection." TCRs with proper binding capabilities to MHC molecules (with self-peptide) are able to transduce cell survival signals and allow the continuing of development to single positive T cells. It has been known that TCRs in DP cells can transduce signals with higher efficiency than peripheral mature T cells, even though they share most of the signaling components. Recent studies have revealed some thymocyte-specific signaling modulators including Themis and Tespa1. The activation of TCR signaling during positive selection results in the activation of several key transcription factors and extensive gene expression change, which has been revealed by newly developed systemic transcriptome analysis tools, and could be used for the evaluation of positive selection process. The fate determination postpositive selection is also governed on the epigenetic level including both DNA methylation and histone modifications.
Collapse
Affiliation(s)
- Jun Lyu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lie Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Linrong Lu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China; Department of Dermatology and Rheumatology in Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Fan D, Li J, Li Y, Guo Y, Zhang X, Wang W, Liu X, Liu J, Dai L, Zhang L, Kang Q, Ji Z. Protein 4.1R negatively regulates CD8 + T-cell activation by modulating phosphorylation of linker for activation of T cells. Immunology 2019; 157:312-321. [PMID: 31135971 DOI: 10.1111/imm.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 11/29/2022] Open
Abstract
Protein 4.1R, an 80 000 MW membrane skeleton protein, is a vital component of the red blood cell membrane cytoskeleton that stabilizes the spectrin-actin network and regulates membrane properties of deformability and mechanical stability. It has been shown that 4.1R is expressed in T cells, including CD8+ T cells, but its role in CD8+ T cells remains unclear. Here, we have explored the role of 4.1R in CD8+ T cells using 4.1R-/- mice. Our results showed that cell activation, proliferation and secretion levels of interleukin-2 and interferon-γ were significantly increased in 4.1R-/- CD8+ T cells. Furthermore, the phosphorylation levels of linker for activation of T cells (LAT) and its downstream signaling molecule extracellular signal-regulated kinase were enhanced in the absence of 4.1R. In vitro co-immunoprecipitation experiments showed a direct interaction between 4.1R and LAT. Moreover, 4.1R-/- CD8+ T cells and mice exhibited an enhanced T-cell-dependent immune response. These data enabled the identification of a negative regulation function for 4.1R in CD8+ T cells by a direct association between 4.1R and LAT, possibly through inhibiting phosphorylation of LAT and then modulating intracellular signal transduction.
Collapse
Affiliation(s)
- Dandan Fan
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianhui Li
- Department of Pathology, Xuchang Central Hospital Affiliated To Henan University of Science and Technology, Xuchang, China
| | - Yi Li
- Henan Key Laboratory of Medical Pathogen Biology, Center for Disease Control and Prevention of Henan Province, Zhengzhou, China
| | - Yaxin Guo
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaolin Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Wen Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaojie Liu
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liguo Zhang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhenyu Ji
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
LKB1 expressed in dendritic cells governs the development and expansion of thymus-derived regulatory T cells. Cell Res 2019; 29:406-419. [PMID: 30940876 DOI: 10.1038/s41422-019-0161-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/08/2019] [Indexed: 01/06/2023] Open
Abstract
Liver Kinase B1 (LKB1) plays a key role in cellular metabolism by controlling AMPK activation. However, its function in dendritic cell (DC) biology has not been addressed. Here, we find that LKB1 functions as a critical brake on DC immunogenicity, and when lost, leads to reduced mitochondrial fitness and increased maturation, migration, and T cell priming of peripheral DCs. Concurrently, loss of LKB1 in DCs enhances their capacity to promote output of regulatory T cells (Tregs) from the thymus, which dominates the outcome of peripheral immune responses, as suggested by increased resistance to asthma and higher susceptibility to cancer in CD11cΔLKB1 mice. Mechanistically, we find that loss of LKB1 specifically primes thymic CD11b+ DCs to facilitate thymic Treg development and expansion, which is independent from AMPK signalling, but dependent on mTOR and enhanced phospholipase C β1-driven CD86 expression. Together, our results identify LKB1 as a critical regulator of DC-driven effector T cell and Treg responses both in the periphery and the thymus.
Collapse
|
7
|
Li Y, Hu S, Wang J, Chen S, Jia X, Lai S. Molecular cloning, polymorphism, and expression analysis of the LKB1/STK11 gene and its association with non-specific digestive disorder in rabbits. Mol Cell Biochem 2018; 449:127-136. [PMID: 29637416 DOI: 10.1007/s11010-018-3349-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/05/2018] [Indexed: 01/14/2023]
Abstract
Liver kinase B1 (LKB1, also called STK11) encodes a serine/threonine kinase mutated in Peutz-Jeghers cancer syndrome characterized by gastrointestinal polyposis. Although LKB1 plays an important role in regulating energy homeostasis, cell growth, and metabolism via activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), nothing is known about its molecular characteristics and possible involvement in non-specific digestive disorder (NSDD) of rabbits. In the present study, we first cloned the coding sequence (CDS) of rabbit LKB1, which consisted of 1317 bp encoding 438 amino acids (AAs) and contained a highly conserved S_TKc kinase domain. Its deduced AA sequence showed 87.93-91.10% similarities with that of other species. In order to determine its involvement in NSDD, a NSDD rabbit model was built by a dietary fiber deficiency. The polymorphic site of LKB1 was then investigated in both healthy and NSDD groups using directing sequencing. Our results suggested that a synonymous variant site (840 c. G > C, CCC→CCG) existed in its S_TKc domain, which was associated with susceptibility to NSDD. Furthermore, qPCR was utilized to examine the mRNA levels of LKB1 and its downstream targets (i.e., PRKAA2, mTOR and NF-kβ) in several intestinal-related tissues from both healthy and NSDD groups. Significant changes in their expression levels between two groups indicated that impaired LKB1 signaling contributed to the intestinal abnormality in NSDD rabbits. Taken together, it could be concluded that LKB1 might be a potential candidate gene affecting the occurrence of rabbit NSDD. This information may serve as a basis for further investigations on rabbit digestive diseases.
Collapse
Affiliation(s)
- Yanhong Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China
| | - Shenqiang Hu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China
| | - Jie Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China
| | - Shiyi Chen
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China
| | - Xianbo Jia
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China
| | - Songjia Lai
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu Campus, Huimin Road #211, Wenjiang, 611130, Sichuan, China.
| |
Collapse
|
8
|
Zhang M, Deng YN, Zhang JY, Liu J, Li YB, Su H, Qu QM. SIRT3 Protects Rotenone-induced Injury in SH-SY5Y Cells by Promoting Autophagy through the LKB1-AMPK-mTOR Pathway. Aging Dis 2018; 9:273-286. [PMID: 29896416 PMCID: PMC5963348 DOI: 10.14336/ad.2017.0517] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 05/17/2017] [Indexed: 12/27/2022] Open
Abstract
SIRT3 is a class III histone deacetylase that modulates energy metabolism, genomic stability and stress resistance. It has been implicated as a potential therapeutic target in a variety of neurodegenerative diseases, including Parkinson’s disease (PD). Our previous study demonstrates that SIRT3 had a neuroprotective effect on a rotenone-induced PD cell model, however, the exact mechanism is unknown. In this study, we investigated the underlying mechanism. We established a SIRT3 stable overexpression cell line using lentivirus infection in SH-SY5Y cells. Then, a PD cell model was established using rotenone. Our data demonstrate that overexpression of SIRT3 increased the level of the autophagy markers LC3 II and Beclin 1. After addition of the autophagy inhibitor 3-MA, the protective effect of SIRT3 diminished: the cell viability decreased, while the apoptosis rate increased; α-synuclein accumulation enhanced; ROS production increased; antioxidants levels, including SOD and GSH, decreased; and MMP collapsed. These results reveal that SIRT3 has neuroprotective effects on a PD cell model by up-regulating autophagy. Furthermore, SIRT3 overexpression also promoted LKB1 phosphorylation, followed by activation of AMPK and decreased phosphorylation of mTOR. These results suggest that the LKB1-AMPK-mTOR pathway has a role in induction of autophagy. Together, our findings indicate a novel mechanism by which SIRT3 protects a rotenone-induced PD cell model through the regulation of autophagy, which, in part, is mediated by activation of the LKB1-AMPK-mTOR pathway.
Collapse
Affiliation(s)
- Meng Zhang
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong-Ning Deng
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing-Yi Zhang
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Liu
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan-Bo Li
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hua Su
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, USA
| | - Qiu-Min Qu
- 1Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Arp2/3 complex controls T cell homeostasis by maintaining surface TCR levels via regulating TCR + endosome trafficking. Sci Rep 2017; 7:8952. [PMID: 28827576 PMCID: PMC5566485 DOI: 10.1038/s41598-017-08357-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
T cell receptor (TCR) signaling is important for T cell homeostasis and function. However, how surface TCR levels are regulated and its biological significance on T cells remains largely unknown. Here, we show that the T cell-specific deletion of Arpc2, a component of Arp2/3 complex, results in compromised peripheral T cell homeostasis. Arp2/3 complex-nucleated actin filaments are essential for maintaining surface TCR levels by regulating TCR+ endosome trafficking in resting state and controlling polarization of TCR+ endosomes during immune synapse formation in T cells. Additionally, Arpc2-TKO T cells are unable to form immune synapse. Interestingly, defected T cell homeostasis is caused by reduced surface TCR levels but not impaired immune synapse formation. Collectively, our findings suggest that Arp2/3 complex-nucleated actin filaments are required for maintaining surface TCR levels via regulating TCR+ endosome trafficking which is essential for T cell homeostasis.
Collapse
|
10
|
Ross FA, Hawley SA, Auciello FR, Gowans GJ, Atrih A, Lamont DJ, Hardie DG. Mechanisms of Paradoxical Activation of AMPK by the Kinase Inhibitors SU6656 and Sorafenib. Cell Chem Biol 2017; 24:813-824.e4. [PMID: 28625738 PMCID: PMC5522529 DOI: 10.1016/j.chembiol.2017.05.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/28/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023]
Abstract
SU6656, a Src kinase inhibitor, was reported to increase fat oxidation and reduce body weight in mice, with proposed mechanisms involving AMP-activated protein kinase (AMPK) activation via inhibition of phosphorylation of either LKB1 or AMPK by the Src kinase, Fyn. However, we report that AMPK activation by SU6656 is independent of Src kinases or tyrosine phosphorylation of LKB1 or AMPK and is not due to decreased cellular energy status or binding at the ADaM site on AMPK. SU6656 is a potent AMPK inhibitor, yet binding at the catalytic site paradoxically promotes phosphorylation of Thr172 by LKB1. This would enhance phosphorylation of downstream targets provided the lifetime of Thr172 phosphorylation was sufficient to allow dissociation of the inhibitor and subsequent catalysis prior to its dephosphorylation. By contrast, sorafenib, a kinase inhibitor in clinical use, activates AMPK indirectly by inhibiting mitochondrial metabolism and increasing cellular AMP:ADP and/or ADP:ATP ratios.
Collapse
Affiliation(s)
- Fiona A Ross
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Simon A Hawley
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - F Romana Auciello
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Graeme J Gowans
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Abdelmadjid Atrih
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Douglas J Lamont
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| |
Collapse
|
11
|
Protein kinase D regulates positive selection of CD4 + thymocytes through phosphorylation of SHP-1. Nat Commun 2016; 7:12756. [PMID: 27670070 PMCID: PMC5052653 DOI: 10.1038/ncomms12756] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 07/29/2016] [Indexed: 02/06/2023] Open
Abstract
Thymic selection shapes an appropriate T cell antigen receptor (TCR) repertoire during T cell development. Here, we show that a serine/threonine kinase, protein kinase D (PKD), is crucial for thymocyte positive selection. In T cell-specific PKD-deficient (PKD2/PKD3 double-deficient) mice, the generation of CD4 single positive thymocytes is abrogated. This defect is likely caused by attenuated TCR signalling during positive selection and incomplete CD4 lineage specification in PKD-deficient thymocytes; however, TCR-proximal tyrosine phosphorylation is not affected. PKD is activated in CD4+CD8+ double positive (DP) thymocytes on stimulation with positively selecting peptides. By phosphoproteomic analysis, we identify SH2-containing protein tyrosine phosphatase-1 (SHP-1) as a direct substrate of PKD. Substitution of wild-type SHP-1 by phosphorylation-defective mutant (SHP-1S557A) impairs generation of CD4+ thymocytes. These results suggest that the PKD–SHP-1 axis positively regulates TCR signalling to promote CD4+ T cell development. The three isoforms of protein kinase D (PKD) have important but often redundant roles in cell signalling. Here the authors show, by generating PKD2/3 double-deficient mice, that PKD is essential for TCR signalling in thymocytes, and identify SHP-1 as a PKD target critical for development of CD4+ T cells.
Collapse
|
12
|
Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism. Nature 2016; 531:651-5. [PMID: 26982734 DOI: 10.1038/nature17412] [Citation(s) in RCA: 646] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 02/10/2016] [Indexed: 12/16/2022]
Abstract
CD8(+) T cells have a central role in antitumour immunity, but their activity is suppressed in the tumour microenvironment. Reactivating the cytotoxicity of CD8(+) T cells is of great clinical interest in cancer immunotherapy. Here we report a new mechanism by which the antitumour response of mouse CD8(+) T cells can be potentiated by modulating cholesterol metabolism. Inhibiting cholesterol esterification in T cells by genetic ablation or pharmacological inhibition of ACAT1, a key cholesterol esterification enzyme, led to potentiated effector function and enhanced proliferation of CD8(+) but not CD4(+) T cells. This is due to the increase in the plasma membrane cholesterol level of CD8(+) T cells, which causes enhanced T-cell receptor clustering and signalling as well as more efficient formation of the immunological synapse. ACAT1-deficient CD8(+) T cells were better than wild-type CD8(+) T cells at controlling melanoma growth and metastasis in mice. We used the ACAT inhibitor avasimibe, which was previously tested in clinical trials for treating atherosclerosis and showed a good human safety profile, to treat melanoma in mice and observed a good antitumour effect. A combined therapy of avasimibe plus an anti-PD-1 antibody showed better efficacy than monotherapies in controlling tumour progression. ACAT1, an established target for atherosclerosis, is therefore also a potential target for cancer immunotherapy.
Collapse
|
13
|
Nelson RK, Gould KA. An Lck-cre transgene accelerates autoantibody production and lupus development in (NZB × NZW)F1 mice. Lupus 2015; 25:137-54. [PMID: 26385218 DOI: 10.1177/0961203315603139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 07/29/2015] [Indexed: 11/16/2022]
Abstract
Lupus is an autoimmune disease characterized by the development of antinuclear autoantibodies and immune complex-mediated tissue damage. T cells in lupus patients appear to undergo apoptosis at an increased rate, and this enhanced T cell apoptosis has been postulated to contribute to lupus pathogenesis by increasing autoantigen load. However, there is no direct evidence to support this hypothesis. In this study, we show that an Lck-cre transgene, which increases T cell apoptosis as a result of T cell-specific expression of cre recombinase, accelerates the development of autoantibodies and nephritis in lupus-prone (NZB × NZW)F1 mice. Although the enhanced T cell apoptosis in Lck-cre transgenic mice resulted in an overall decrease in the relative abundance of splenic CD4(+) and CD8(+) T cells, the proportion of activated CD4(+) T cells was increased and no significant change was observed in the relative abundance of suppressive T cells. We postulate that the Lck-cre transgene promoted lupus by enhancing T cell apoptosis, which, in conjunction with the impaired clearance of apoptotic cells in lupus-prone mice, increased the nuclear antigen load and accelerated the development of anti-nuclear autoantibodies. Furthermore, our results also underscore the importance of including cre-only controls in studies using the cre-lox system.
Collapse
Affiliation(s)
- R K Nelson
- Department of Genetics, Cell Biology & Anatomy, Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - K A Gould
- Department of Genetics, Cell Biology & Anatomy, Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
14
|
Walsh NC, Waters LR, Fowler JA, Lin M, Cunningham CR, Brooks DG, Rehg JE, Morse HC, Teitell MA. LKB1 inhibition of NF-κB in B cells prevents T follicular helper cell differentiation and germinal center formation. EMBO Rep 2015; 16:753-68. [PMID: 25916856 PMCID: PMC4467859 DOI: 10.15252/embr.201439505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 03/06/2015] [Accepted: 03/16/2015] [Indexed: 11/09/2022] Open
Abstract
T-cell-dependent antigenic stimulation drives the differentiation of B cells into antibody-secreting plasma cells and memory B cells, but how B cells regulate this process is unclear. We show that LKB1 expression in B cells maintains B-cell quiescence and prevents the premature formation of germinal centers (GCs). Lkb1-deficient B cells (BKO) undergo spontaneous B-cell activation and secretion of multiple inflammatory cytokines, which leads to splenomegaly caused by an unexpected expansion of T cells. Within this cytokine response, increased IL-6 production results from heightened activation of NF-κB, which is suppressed by active LKB1. Secreted IL-6 drives T-cell activation and IL-21 production, promoting T follicular helper (TFH ) cell differentiation and expansion to support a ~100-fold increase in steady-state GC B cells. Blockade of IL-6 secretion by BKO B cells inhibits IL-21 expression, a known inducer of TFH -cell differentiation and expansion. Together, these data reveal cell intrinsic and surprising cell extrinsic roles for LKB1 in B cells that control TFH -cell differentiation and GC formation, and place LKB1 as a central regulator of T-cell-dependent humoral immunity.
Collapse
Affiliation(s)
- Nicole C Walsh
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Lynnea R Waters
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Jessica A Fowler
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Mark Lin
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Cameron R Cunningham
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute University of California, Los Angeles, CA, USA
| | - David G Brooks
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute University of California, Los Angeles, CA, USA
| | - Jerold E Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases National Institutes of Health, Rockville, MD, USA
| | - Michael A Teitell
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA, USA Molecular Biology Institute, University of California, Los Angeles, CA, USA Broad Stem Cell Research Center, Departments of Pediatrics and Bioengineering, California NanoSystems Institute, and Jonsson Comprehensive Cancer Center University of California, Los Angeles, CA, USA
| |
Collapse
|
15
|
Liu T, Qin W, Hou L, Huang Y. MicroRNA-17 promotes normal ovarian cancer cells to cancer stem cells development via suppression of the LKB1-p53-p21/WAF1 pathway. Tumour Biol 2014; 36:1881-93. [PMID: 25510663 DOI: 10.1007/s13277-014-2790-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/29/2014] [Indexed: 01/06/2023] Open
Abstract
The mechanism underlying the development of human ovarian cancer is poorly understood. The liver kinase protein, LKB1, is hypothesized to play a pivotal role in tumor cell proliferation and invasion capacity through regulation of p53 and p21/WAF1 expression. Previous studies suggest LKB1 may, in turn, be regulated by microRNA-17. Here, we examined the role of miR-17 in the expression of LKB1 and the downstream effects on proliferation and invasion capacity of normal ovarian cancer cells (OCCs) and ovarian stem cells. In this study, both the mRNA and protein expression levels of LKB1, p53, and p21 decreased in OCCs following transfection with a miR-17 expression plasmid. MiR-17 expression affected cell cycle regulation and stimulated the proliferation and invasion capacity of OCCs in vitro. ChIP assays indicated that the binding efficiency of p53 to the p21/WAF1 gene promoter was much lower in miR-17 transfected OCCs than in OCCs transfected with a mutated miR-17. Co-immunoprecipitation and western blotting showed significantly lower levels of p53 and p53 Ser15-pho in the miR-17 transfected OCCs as compared to the mutant miR-17 transfected OCCs. Xenograft experiments confirmed that suppression of tumor growth in vivo occurred in the absence of functional miR-17. These findings suggest that mature miR-17 expression may have an important role in the pathogenesis of human ovarian tumors through its interference with the LKB1-p53-p21/WAF1 pathway expression by epigenetic modification. These findings are of potential importance in the identification of novel therapeutic targets in human ovarian cancer.
Collapse
Affiliation(s)
- Te Liu
- Shanghai Tenth People's Hospital, Medical School, Tongji University, Shanghai, 200072, China,
| | | | | | | |
Collapse
|
16
|
CBAP promotes thymocyte negative selection by facilitating T-cell receptor proximal signaling. Cell Death Dis 2014; 5:e1518. [PMID: 25393474 PMCID: PMC4260732 DOI: 10.1038/cddis.2014.474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/29/2014] [Accepted: 10/06/2014] [Indexed: 12/11/2022]
Abstract
T-cell receptor (TCR)-transduced signaling is critical to thymocyte development at the CD4/CD8 double-positive stage, but the molecules involved in this process are not yet fully characterized. We previously demonstrated that GM-CSF/IL-3/IL-5 receptor common β-chain-associated protein (CBAP) modulates ZAP70-mediated T-cell migration and adhesion. On the basis of the high expression of CBAP during thymocyte development, we investigated the function of CBAP in thymocyte development using a CBAP knockout mouse. CBAP-deficient mice showed normal early thymocyte development and positive selection. In contrast, several negative selection models (including TCR transgene, superantigen staphylococcal enterotoxin B, and anti-CD3 antibody treatment) revealed an attenuation of TCR-induced thymocyte deletion in CBAP knockout mice. This phenotype correlated with a reduced accumulation of BIM upon TCR crosslinking in CBAP-deficient thymocytes. Loss of CBAP led to reduced TCR-induced phosphorylation of proteins involved in both proximal and distal signaling events, including ZAP70, LAT, PLCγ1, and JNK1/2. Moreover, TCR-induced association of LAT signalosome components was reduced in CBAP-deficient thymocytes. Our data demonstrate that CBAP is a novel component in the TCR signaling pathway and modulates thymocyte apoptosis during negative selection.
Collapse
|
17
|
Sun Y, Zhu X, Chen X, Liu H, Xu Y, Chu Y, Wang G, Liu X. The mediator subunit Med23 contributes to controlling T-cell activation and prevents autoimmunity. Nat Commun 2014; 5:5225. [PMID: 25301163 DOI: 10.1038/ncomms6225] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/10/2014] [Indexed: 12/11/2022] Open
Abstract
T-cell activation is critical for successful immune responses and is controlled at multiple levels. Although many changes of T-cell receptor-associated signalling molecules affect T-cell activation, the transcriptional mechanisms that control this process remain largely unknown. Here we find that T cell-specific deletion of the mediator subunit Med23 leads to hyperactivation of T cells and aged Med23-deficient mice exhibit an autoimmune syndrome. Med23 specifically and consistently promotes the transcription of multiple negative regulators of T-cell activation. In the absence of Med23, the T-cell activation threshold is lower, which results in enhanced antitumour T-cell function. Cumulatively, our data suggest that Med23 contributes to controlling T-cell activation at the transcriptional level and prevents the development of autoimmunity.
Collapse
Affiliation(s)
- Yang Sun
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoyan Zhu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xufeng Chen
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Xu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yajing Chu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
18
|
Lyn-mediated procaspase 8 dimerization blocks apoptotic signaling in B-cell chronic lymphocytic leukemia. Blood 2014; 123:875-83. [DOI: 10.1182/blood-2013-02-485540] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Key Points
Lyn’s overexpression mediates resistance to apoptosis by promoting phosphorylation and dimerization of procaspase 8 in B-CLL cells.
Collapse
|
19
|
Abstract
Phospholipases are enzymes that use phospholipids as substrate and are classified in three major classes A, C and D based on the reaction they catalyse. Phosphatidylinositol-specific Phospholipase C enzymes utilize phosphatidylinositol 4,5-bisphosphate as substrate and cleave the bond between the glycerol and the phosphate to produce important second messenger such as inositol trisphosphate and diacylglycerol. The Phospholipase C members are the most well-known phospholipases for their role in lipid signalling and cell proliferation and comprise 13 isoforms classified in 6 distinct sub-families. In particular, signalling activated by Phospholipase C γ, mostly activated by receptor and non-receptor tyrosine kinases, is well characterized in different cell systems. Increasing evidence suggest that Phospholipase C γ plays a key role in cell migration and invasion. Because of its role in cell growth and invasion, aberrant Phospholipase C γ signalling can contribute to carcinogenesis. A major challenge facing investigators who seek to target Phospholipase C γ directly is the fact that it is considered an "undruggable" protein. Indeed, isoform specificity and toxicity represents a big hurdle in the development of Phospholipase C γ small molecule inhibitors. Therefore, a future development in the field could be the identification of interacting partners as therapeutic targets that could be more druggable than Phospholipase C γ.
Collapse
Affiliation(s)
- Rossano Lattanzio
- Aging Research Centre, G. d'Annunzio University Foundation, 66013 Chieti, Italy.
| | | | | |
Collapse
|
20
|
Zarrouk M, Rolf J, Cantrell DA. LKB1 mediates the development of conventional and innate T cells via AMP-dependent kinase autonomous pathways. PLoS One 2013; 8:e60217. [PMID: 23533675 PMCID: PMC3606301 DOI: 10.1371/journal.pone.0060217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 02/23/2013] [Indexed: 11/18/2022] Open
Abstract
The present study has examined the role of the serine/threonine kinase LKB1 in the survival and differentiation of CD4/8 double positive thymocytes. LKB1-null DPs can respond to signals from the mature α/β T-cell-antigen receptor and initiate positive selection. However, in the absence of LKB1, thymocytes fail to mature to conventional single positive cells causing severe lymphopenia in the peripheral lymphoid tissues. LKB1 thus appears to be dispensable for positive selection but important for the maturation of positively selected thymocytes. LKB1 also strikingly prevented the development of invariant Vα14 NKT cells and innate TCR αβ gut lymphocytes. Previous studies with gain of function mutants have suggested that the role of LKB1 in T cell development is mediated by its substrate the AMP-activated protein kinase (AMPK). The present study now analyses the impact of AMPK deletion in DP thymocytes and shows that the role of LKB1 during the development of both conventional and innate T cells is mediated by AMPK-independent pathways.
Collapse
Affiliation(s)
- Marouan Zarrouk
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | | | | |
Collapse
|
21
|
Yang K, Chi H. mTOR and metabolic pathways in T cell quiescence and functional activation. Semin Immunol 2013; 24:421-8. [PMID: 23375549 DOI: 10.1016/j.smim.2012.12.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 12/11/2012] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR), an evolutionally conserved serine and threonine kinase, plays a critical role in the promotion of cell growth and proliferation via integration of cellular and environmental cues. In adaptive immunity, the mTOR pathway orchestrates multiple physiological processes including the development and homeostasis of T cells under steady state, and their subsequent activation and differentiation upon antigen recognition. Associated with such fate decisions is the dynamic reprogramming of T cell metabolic pathways, as naïve, activated and memory cells are defined by distinct bioenergetic and biosynthetic activities. Emerging evidence indicates that mTOR signaling intersects with T cell metabolism at two major levels to constitute a critical control mechanism of T cell fate decisions. First, as a central environmental sensor, mTOR links immune signaling and the availability of nutrients, especially amino acids. Second, mTOR activates specific metabolic pathways in T cells such as aerobic glycolysis (also known as the "Warburg effect") in a process dependent upon the induction of transcription factors MYC and HIF1α. Understanding how mTOR interplays with T cell metabolism to dictate T cell fates and functions will provide fundamental insights into the mechanism of immune responses and the development of novel therapeutics against immune-mediated diseases. In this review, we summarize the current advances on mTOR signaling and T cell metabolism in the control of development, homeostasis, activation and differentiation of T cells.
Collapse
Affiliation(s)
- Kai Yang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
22
|
Abstract
Germinal center kinases (GCKs) participate in a variety of signaling pathways needed to regulate cellular functions including apoptosis, cell proliferation, polarity and migration. Recent studies have shown that GCKs are participants in both adaptive and innate immune regulation. However, the differential activation and regulatory mechanisms of GCKs, as well as upstream and downstream signaling molecules, remain to be fully defined. It remains unresolved whether and how GCKs may cross-talk with existing signaling pathways. This review stresses the progresses in research of GCKs relevant to the immune system.
Collapse
|
23
|
Matarese G, Procaccini C, Rosa V. At the crossroad of T cells, adipose tissue, and diabetes. Immunol Rev 2012; 249:116-34. [DOI: 10.1111/j.1600-065x.2012.01154.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
24
|
Blagih J, Krawczyk CM, Jones RG. LKB1 and AMPK: central regulators of lymphocyte metabolism and function. Immunol Rev 2012; 249:59-71. [DOI: 10.1111/j.1600-065x.2012.01157.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | - Connie M. Krawczyk
- Department of Microbiology and Immunology; McGill University; Montreal; Quebec
| | | |
Collapse
|
25
|
Wang D, Zheng M, Lei L, Ji J, Yao Y, Qiu Y, Ma L, Lou J, Ouyang C, Zhang X, He Y, Chi J, Wang L, Kuang Y, Wang J, Cao X, Lu L. Tespa1 is involved in late thymocyte development through the regulation of TCR-mediated signaling. Nat Immunol 2012; 13:560-8. [PMID: 22561606 DOI: 10.1038/ni.2301] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/28/2012] [Indexed: 12/11/2022]
Abstract
Signaling via the T cell antigen receptor (TCR) during the CD4(+)CD8(+) double-positive developmental stage determines thymocyte selection and lineage commitment. Here we describe a previously uncharacterized T cell-expressed protein, Tespa1, with critical functions during the positive selection of thymocytes. Tespa1(-/-) mice had fewer mature thymic CD4(+) and CD8(+) T cells, which reflected impaired thymocyte development. Tespa1 associated with the TCR signaling components PLC-γ1 and Grb2, and Tespa1 deficiency resulted in attenuated TCR signaling, as reflected by defective activation of the Erk-AP-1 and Ca(2+)-NFAT pathways. Our findings demonstrate that Tespa1 is a component of the TCR signalosome and is essential for T cell selection and maturation through the regulation of TCR signaling during T cell development.
Collapse
Affiliation(s)
- Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chi H. Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev Immunol 2012; 12:325-38. [PMID: 22517423 DOI: 10.1038/nri3198] [Citation(s) in RCA: 728] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The evolutionarily conserved kinase mTOR (mammalian target of rapamycin) couples cell growth and metabolism to environmental inputs in eukaryotes. T cells depend on mTOR signalling to integrate immune signals and metabolic cues for their proper maintenance and activation. Under steady-state conditions, mTOR is actively controlled by multiple inhibitory mechanisms, and this enforces normal T cell homeostasis. Antigen recognition by naive CD4(+) and CD8(+) T cells triggers mTOR activation, which in turn programmes the differentiation of these cells into functionally distinct lineages. This Review focuses on the signalling mechanisms of mTOR in T cell homeostatic and functional fates, and discusses the therapeutic implications of targeting mTOR in T cells.
Collapse
Affiliation(s)
- Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| |
Collapse
|
27
|
Yang YR, Choi JH, Chang JS, Kwon HM, Jang HJ, Ryu SH, Suh PG. Diverse cellular and physiological roles of phospholipase C-γ1. Adv Biol Regul 2012; 52:138-151. [PMID: 21964416 DOI: 10.1016/j.advenzreg.2011.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 05/31/2023]
Affiliation(s)
- Yong Ryoul Yang
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
28
|
Sinclair C, Saini M, Schim van der Loeff I, Sakaguchi S, Seddon B. The Long-Term Survival Potential of Mature T Lymphocytes Is Programmed During Development in the Thymus. Sci Signal 2011; 4:ra77. [DOI: 10.1126/scisignal.2002246] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
MacIver NJ, Blagih J, Saucillo DC, Tonelli L, Griss T, Rathmell JC, Jones RG. The liver kinase B1 is a central regulator of T cell development, activation, and metabolism. THE JOURNAL OF IMMUNOLOGY 2011; 187:4187-98. [PMID: 21930968 DOI: 10.4049/jimmunol.1100367] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
T cell activation leads to engagement of cellular metabolic pathways necessary to support cell proliferation and function. However, our understanding of the signal transduction pathways that regulate metabolism and their impact on T cell function remains limited. The liver kinase B1 (LKB1) is a serine/threonine kinase that links cellular metabolism with cell growth and proliferation. In this study, we demonstrate that LKB1 is a critical regulator of T cell development, viability, activation, and metabolism. T cell-specific ablation of the gene that encodes LKB1 resulted in blocked thymocyte development and a reduction in peripheral T cells. LKB1-deficient T cells exhibited defects in cell proliferation and viability and altered glycolytic and lipid metabolism. Interestingly, loss of LKB1 promoted increased T cell activation and inflammatory cytokine production by both CD4(+) and CD8(+) T cells. Activation of the AMP-activated protein kinase (AMPK) was decreased in LKB1-deficient T cells. AMPK was found to mediate a subset of LKB1 functions in T lymphocytes, as mice lacking the α1 subunit of AMPK displayed similar defects in T cell activation, metabolism, and inflammatory cytokine production, but normal T cell development and peripheral T cell homeostasis. LKB1- and AMPKα1-deficient T cells each displayed elevated mammalian target of rapamycin complex 1 signaling and IFN-γ production that could be reversed by rapamycin treatment. Our data highlight a central role for LKB1 in T cell activation, viability, and metabolism and suggest that LKB1-AMPK signaling negatively regulates T cell effector function through regulation of mammalian target of rapamycin activity.
Collapse
Affiliation(s)
- Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|