Glutaminolysis and peripheral CD4+ T cell differentiation: from mechanism to intervention strategy

To maintain the body’s regular immune system, CD4+ T cell homeostasis is crucial, particularly T helper (Th1, Th17) cells and T regulatory (Treg) cells. Abnormally differentiated peripheral CD4+ T cells are responsible for the occurrence and development of numerous diseases, including autoimmune diseases, transplantation rejection, and irritability. Searching for an effective interventional approach to control this abnormal differentiation is therefore especially important. As immunometabolism progressed, the inherent metabolic factors underlying the immune cell differentiation have gradually come to light. Mounting number of studies have revealed that glutaminolysis plays an indelible role in the differentiation of CD4+ T cells. Besides, alterations in the glutaminolysis can also lead to changes in the fate of peripheral CD4+ T cells. All of this indicate that the glutaminolysis pathway has excellent potential for interventional regulation of CD4+ T cells differentiation. Here, we summarized the process by which glutaminolysis regulates the fate of CD4+ T cells during differentiation and further investigated how to reshape abnormal CD4+ T cell differentiation by targeting glutaminolysis.

[1]  K. Park,et al.  Targeting glutamine metabolism as a therapeutic strategy for cancer , 2023, Experimental & molecular medicine.

[2]  S. Abrignani,et al.  Human T lymphocytes at tumor sites , 2022, Seminars in Immunopathology.

[3]  Ying Shen,et al.  Metabolic and Nonmetabolic Functions of PSAT1 Coordinate Signaling Cascades to Confer EGFR Inhibitor Resistance and Drive Progression in Lung Adenocarcinoma. , 2022, Cancer research.

[4]  G. Wang,et al.  Inhibition of glutaminolysis ameliorates lupus by regulating T and B cell subsets and downregulating the mTOR/P70S6K/4EBP1 and NLRP3/caspase-1/IL-1β pathways in MRL/lpr mice. , 2022, International immunopharmacology.

[5]  G. K. Khurana Hershey,et al.  The role of the CBM complex in allergic inflammation and disease. , 2022, The Journal of allergy and clinical immunology.

[6]  R. Cianciolo,et al.  Murine cytomegalovirus promotes renal allograft inflammation via Th1/17 cells and IL‐17A , 2022, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[7]  V. Kuchroo,et al.  B Cell IL-4 Drives Th2 Responses In Vivo, Ameliorates Allograft Rejection, and Promotes Allergic Airway Disease , 2022, Frontiers in Immunology.

[8]  L. Fong,et al.  Cytotoxic CD4+ T cells in cancer: Expanding the immune effector toolbox , 2021, Immunity.

[9]  Ping-Chih Ho,et al.  Fueling T-cell Antitumor Immunity: Amino Acid Metabolism Revisited , 2021, Cancer Immunology Research.

[10]  B. Skålhegg,et al.  Identification and characterization of a novel glutaminase inhibitor , 2021, FEBS open bio.

[11]  S. Im,et al.  Glutamine deficiency shifts the asthmatic state toward neutrophilic airway inflammation , 2021, Allergy.

[12]  Yana Geng,et al.  Bergenin, a PPARγ agonist, inhibits Th17 differentiation and subsequent neutrophilic asthma by preventing GLS1-dependent glutaminolysis , 2021, Acta Pharmacologica Sinica.

[13]  D. Krappmann,et al.  A patent review of MALT1 inhibitors (2013-present) , 2021, Expert opinion on therapeutic patents.

[14]  Yun Zheng,et al.  The role of GLS1-mediated glutaminolysis/2-HG/H3K4me3 and GSH/ROS signals in Th17 responses counteracted by PPARγ agonists , 2021, Theranostics.

[15]  M. Kopf,et al.  Redox regulation of immunometabolism , 2020, Nature Reviews Immunology.

[16]  Wei-Chen Lee,et al.  Targeting glutamine metabolism as an effective means to promote allograft acceptance while inhibit tumor growth. , 2020, Transplant immunology.

[17]  Ya Chun Yu,et al.  Glutamine reliance in cell metabolism , 2020, Experimental & Molecular Medicine.

[18]  S. Bröer Amino Acid Transporters as Targets for Cancer Therapy: Why, Where, When, and How , 2020, International journal of molecular sciences.

[19]  W. Zhou,et al.  GLS1-mediated glutaminolysis unbridled by MALT1 protease promotes psoriasis pathogenesis. , 2020, The Journal of clinical investigation.

[20]  Zhuo-wei Hu,et al.  PML-RARα interaction with TRIB3 impedes PPARγ/RXR function and triggers dyslipidemia in acute promyelocytic leukemia , 2020, Theranostics.

[21]  W. Ren,et al.  Glutamine metabolism in Th17/Treg cell fate: applications in Th17 cell-associated diseases , 2020, Science China Life Sciences.

[22]  Hong Jin,et al.  T Cell Metabolism: A New Perspective on Th17/Treg Cell Imbalance in Systemic Lupus Erythematosus , 2020, Frontiers in Immunology.

[23]  Peng Wang,et al.  The Regulatory Effects of mTOR Complexes in the Differentiation and Function of CD4+ T Cell Subsets , 2020, Journal of immunology research.

[24]  Nicole M. Chapman,et al.  mTOR signaling at the crossroads of environmental signals and T‐cell fate decisions , 2020, Immunological reviews.

[25]  Delong Meng,et al.  Glutamine and asparagine activate mTORC1 independently of Rag GTPases , 2020, The Journal of Biological Chemistry.

[26]  Ligong Chen,et al.  Solute carrier transporters: the metabolic gatekeepers of immune cells , 2019, Acta pharmaceutica Sinica. B.

[27]  H. Lee,et al.  A Variant of SLC1A5 Is a Mitochondrial Glutamine Transporter for Metabolic Reprogramming in Cancer Cells. , 2019, Cell metabolism.

[28]  Linda V. Sinclair,et al.  Quantitative analysis of how Myc controls T cell proteomes and metabolic pathways during T cell activation , 2019, bioRxiv.

[29]  T. Masuko,et al.  Anti‐tumor effects of an antagonistic mAb against the ASCT2 amino acid transporter on KRAS‐mutated human colorectal cancer cells , 2019, Cancer medicine.

[30]  G. Tsokos,et al.  Glutaminase 1 Inhibition Reduces Glycolysis and Ameliorates Lupus‐like Disease in MRL/lpr Mice and Experimental Autoimmune Encephalomyelitis , 2019, Arthritis & rheumatology.

[31]  Graham M. West,et al.  Cryo-EM structures of the human glutamine transporter SLC1A5 (ASCT2) in the outward-facing conformation , 2019, eLife.

[32]  R. Beyaert,et al.  Ubiquitination and phosphorylation of the CARD11-BCL10-MALT1 signalosome in T cells. , 2019, Cellular immunology.

[33]  Jingwei Jiang,et al.  Topotecan induces apoptosis via ASCT2 mediated oxidative stress in gastric cancer. , 2019, Phytomedicine : international journal of phytotherapy and phytopharmacology.

[34]  S. Gupta,et al.  Delta-Tocotrienol Modulates Glutamine Dependence by Inhibiting ASCT2 and LAT1 Transporters in Non-Small Cell Lung Cancer (NSCLC) Cells: A Metabolomic Approach , 2019, Metabolites.

[35]  Yuquan Wei,et al.  Glutaminase 1 expression in colorectal cancer cells is induced by hypoxia and required for tumor growth, invasion, and metastatic colonization , 2019, Cell Death & Disease.

[36]  Shaohe Wang,et al.  Targeting glutaminase 1 attenuates stemness properties in hepatocellular carcinoma by increasing reactive oxygen species and suppressing Wnt/beta-catenin pathway , 2018, EBioMedicine.

[37]  J. Locasale,et al.  Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism , 2018, Cell.

[38]  J. Mellor,et al.  IDH1: Linking Metabolism and Epigenetics , 2018, Front. Genet..

[39]  Lei Gao,et al.  c-Myc-driven glycolysis via TXNIP suppression is dependent on glutaminase-MondoA axis in prostate cancer. , 2018, Biochemical and biophysical research communications.

[40]  Tingting Wang,et al.  Glutathione de novo synthesis but not recycling process coordinates with glutamine catabolism to control redox homeostasis and directs murine T cell differentiation , 2018, eLife.

[41]  Y. Li,et al.  Resveratrol enhances cisplatin-induced apoptosis in human hepatoma cells via glutamine metabolism inhibition , 2018, BMB reports.

[42]  A. Bröer,et al.  Disruption of Amino Acid Homeostasis by Novel ASCT2 Inhibitors Involves Multiple Targets , 2018, Front. Pharmacol..

[43]  L. Quek,et al.  Benzylserine inhibits breast cancer cell growth by disrupting intracellular amino acid homeostasis and triggering amino acid response pathways , 2018, BMC Cancer.

[44]  E. Pearce,et al.  Unraveling the Complex Interplay Between T Cell Metabolism and Function. , 2018, Annual review of immunology.

[45]  K. Maloy,et al.  Metabolic Adaptations of CD4+ T Cells in Inflammatory Disease , 2018, Front. Immunol..

[46]  G. Tsokos,et al.  Transcriptional factor ICER promotes glutaminolysis and the generation of Th17 cells , 2018, Proceedings of the National Academy of Sciences.

[47]  D. Farber,et al.  Human T Cell Development, Localization, and Function throughout Life. , 2018, Immunity.

[48]  Wenyan Ren,et al.  miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma , 2018, Cell Death & Differentiation.

[49]  William P. Katt,et al.  Characterization of the interactions of potent allosteric inhibitors with glutaminase C, a key enzyme in cancer cell glutamine metabolism , 2018, The Journal of Biological Chemistry.

[50]  S. Gygi,et al.  SAMTOR is an S-adenosylmethionine sensor for the mTORC1 pathway , 2017, Science.

[51]  U. Staubli,et al.  Phenylglycine analogs are inhibitors of the neutral amino acid transporters ASCT1 and ASCT2 and enhance NMDA receptor-mediated LTP in rat visual cortex slices , 2017, Neuropharmacology.

[52]  Frédérick A. Mallette,et al.  Oncogenic Activities of IDH1/2 Mutations: From Epigenetics to Cellular Signaling. , 2017, Trends in cell biology.

[53]  Y. Zhang,et al.  Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism , 2017, Nature.

[54]  G. Qing,et al.  Epigenetic silencing of microRNA-137 enhances ASCT2 expression and tumor glutamine metabolism , 2017, Oncogenesis.

[55]  S. Venneti,et al.  Glutaminolysis: A Hallmark of Cancer Metabolism. , 2017, Annual review of biomedical engineering.

[56]  Jiahuai Han,et al.  2-HG Inhibits Necroptosis by Stimulating DNMT1-Dependent Hypermethylation of the RIP3 Promoter. , 2017, Cell reports.

[57]  M. Bianchi,et al.  GPNA inhibits the sodium-independent transport system l for neutral amino acids , 2017, Amino Acids.

[58]  J. Powell,et al.  mTORC1 Promotes T-bet Phosphorylation To Regulate Th1 Differentiation , 2017, The Journal of Immunology.

[59]  T. Mak,et al.  Glutathione Primes T Cell Metabolism for Inflammation , 2017, Immunity.

[60]  Ming Zhou,et al.  Interplay between Immune Checkpoint Proteins and Cellular Metabolism. , 2017, Cancer research.

[61]  H. Ando,et al.  Establishment of monoclonal antibodies against cell surface domains of ASCT2/SLC1A5 and their inhibition of glutamine-dependent tumor cell growth. , 2017, Biochemical and biophysical research communications.

[62]  M. Magnani,et al.  Glutathione Depletion Is Linked with Th2 Polarization in Mice with a Retrovirus-Induced Immunodeficiency Syndrome, Murine AIDS: Role of Proglutathione Molecules as Immunotherapeutics , 2016, Journal of Virology.

[63]  S. Bröer,et al.  Deletion of Amino Acid Transporter ASCT2 (SLC1A5) Reveals an Essential Role for Transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2) to Sustain Glutaminolysis in Cancer Cells* , 2016, The Journal of Biological Chemistry.

[64]  William P. Katt,et al.  Design and evaluation of novel glutaminase inhibitors. , 2016, Bioorganic & medicinal chemistry.

[65]  M. Boothby Faculty Opinions recommendation of Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. , 2016 .

[66]  S. Kaech,et al.  The multifaceted role of CD4+ T cells in CD8+ T cell memory , 2016, Nature Reviews Immunology.

[67]  Liqing Zhou,et al.  fMiRNA-192 and miRNA-204 Directly Suppress lncRNA HOTTIP and Interrupt GLS1-Mediated Glutaminolysis in Hepatocellular Carcinoma , 2015, PLoS genetics.

[68]  Shao-Cong Sun,et al.  TCR signaling to NF-κB and mTORC1: Expanding roles of the CARMA1 complex. , 2015, Molecular immunology.

[69]  Philippe A. Robert,et al.  Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation , 2015, Science Signaling.

[70]  Jeffrey W. Smith,et al.  Regulation of glutamine carrier proteins by RNF5 determines breast cancer response to ER stress-inducing chemotherapies. , 2015, Cancer cell.

[71]  K. Guan,et al.  Differential regulation of mTORC1 by leucine and glutamine , 2015, Science.

[72]  R. Deberardinis,et al.  Mechanism by which a recently discovered allosteric inhibitor blocks glutamine metabolism in transformed cells , 2014, Proceedings of the National Academy of Sciences.

[73]  Jae-Hoon Chang,et al.  Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. , 2014, Immunity.

[74]  K. Thangavelu,et al.  Structural Basis for the Active Site Inhibition Mechanism of Human Kidney-Type Glutaminase (KGA) , 2014, Scientific Reports.

[75]  B. Ross,et al.  Electrographic seizures are significantly reduced by in vivo inhibition of neuronal uptake of extracellular glutamine in rat hippocampus , 2013, Epilepsy Research.

[76]  A. Simeonov,et al.  Kinetic characterization of ebselen, chelerythrine and apomorphine as glutaminase inhibitors. , 2013, Biochemical and biophysical research communications.

[77]  M. Leuenberger,et al.  The SLC1 high-affinity glutamate and neutral amino acid transporter family. , 2013, Molecular aspects of medicine.

[78]  A. Lane,et al.  Control of Glutamine Metabolism By the Tumor Suppressor Rb , 2013, Oncogene.

[79]  P. Koutentis,et al.  Inactivation of the glutamine/amino acid transporter ASCT2 by 1,2,3-dithiazoles: proteoliposomes as a tool to gain insights in the molecular mechanism of action and of antitumor activity. , 2012, Toxicology and applied pharmacology.

[80]  R. Gill,et al.  CD4 T cells mediate cardiac xenograft rejection via host MHC Class II. , 2012, The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation.

[81]  C. Akdis,et al.  TH17 and TH22 cells: a confusion of antimicrobial response with tissue inflammation versus protection. , 2012, The Journal of allergy and clinical immunology.

[82]  A. Hirao,et al.  PI3K-Akt-mTORC1-S6K1/2 axis controls Th17 differentiation by regulating Gfi1 expression and nuclear translocation of RORγ. , 2021, Cell reports.

[83]  J. Sprent,et al.  The role of interleukin-2 during homeostasis and activation of the immune system , 2012, Nature Reviews Immunology.

[84]  D. Green,et al.  The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. , 2011, Immunity.

[85]  A. Palamara,et al.  Modulation of Th1/Th2 immune responses to HIV-1 Tat by new pro-GSH molecules. , 2011, Vaccine.

[86]  G. Semenza,et al.  Control of TH17/Treg Balance by Hypoxia-Inducible Factor 1 , 2011, Cell.

[87]  D. Green,et al.  HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells , 2011, The Journal of experimental medicine.

[88]  P. Worley,et al.  The mammalian Target of Rapamycin (mTOR) regulates T helper cell differentiation through the selective activation of mTORC1 and mTORC2 signaling , 2011, Nature Immunology.

[89]  S. Crotty,et al.  Follicular helper CD4 T cells (TFH). , 2011, Annual review of immunology.

[90]  M. Magnani,et al.  The increase in intra-macrophage thiols induced by new pro-GSH molecules directs the Th1 skewing in ovalbumin immunized mice. , 2010, Vaccine.

[91]  W. Filipowicz,et al.  The widespread regulation of microRNA biogenesis, function and decay , 2010, Nature Reviews Genetics.

[92]  K. Frauwirth,et al.  Glutamine Uptake and Metabolism Are Coordinately Regulated by ERK/MAPK during T Lymphocyte Activation , 2010, The Journal of Immunology.

[93]  A. Bado,et al.  Luminal leptin inhibits L-glutamine transport in rat small intestine: involvement of ASCT2 and B0AT1. , 2010, American journal of physiology. Gastrointestinal and liver physiology.

[94]  W. Langdon,et al.  Mechanisms of NKT cell anergy induction involve Cbl-b-promoted monoubiquitination of CARMA1 , 2009, Proceedings of the National Academy of Sciences.

[95]  Jeffrey P. MacKeigan,et al.  Bidirectional Transport of Amino Acids Regulates mTOR and Autophagy , 2009, Cell.

[96]  Shelly C. Lu Regulation of glutathione synthesis. , 2009, Molecular aspects of medicine.

[97]  N. Curthoys,et al.  Novel mechanism of inhibition of rat kidney-type glutaminase by bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES). , 2007, The Biochemical journal.

[98]  A. Israël,et al.  Negative feedback loop in T cell activation through IκB kinase-induced phosphorylation and degradation of Bcl10 , 2007, Proceedings of the National Academy of Sciences.

[99]  C. Esslinger,et al.  Ngamma-aryl glutamine analogues as probes of the ASCT2 neutral amino acid transporter binding site. , 2005, Bioorganic & medicinal chemistry.

[100]  D. Schoetz Who, Why, What, and How? , 2004, Diseases of the colon and rectum.

[101]  C. Grewer,et al.  New inhibitors for the neutral amino acid transporter ASCT2 reveal its Na+‐dependent anion leak , 2004, The Journal of physiology.

[102]  J. Hamuro,et al.  The skewing to Th1 induced by lentinan is directed through the distinctive cytokine production by macrophages with elevated intracellular glutathione content. , 2002, International immunopharmacology.

[103]  S. Low,et al.  Mechanisms of Glutamine Transport in Rat Adipocytes and Acute Regulation by Cell Swelling , 2001, Cellular Physiology and Biochemistry.

[104]  T. Ishii,et al.  An anti-ASCT2 monoclonal antibody suppresses gastric cancer growth by inducing oxidative stress and antibody dependent cellular toxicity in preclinical models. , 2018, American journal of cancer research.

[105]  U. Khan,et al.  T Lymphocytes and Autoimmunity. , 2018, International review of cell and molecular biology.

[106]  B. Medoff,et al.  The role of CARMA1 in T cells. , 2013, Critical reviews in immunology.

[107]  A. Khoruts,et al.  In vivo activation of antigen-specific CD4 T cells. , 2001, Annual review of immunology.