Tissue-Specific Dependence of Th1 Cells on the Amino Acid Transporter SLC38A1 in Inflammation

Amino acid (AA) uptake is essential for T cell metabolism and function, but how tissue sites and inflammation affect CD4+ T cell subset requirements for specific AA remains uncertain. Here we tested CD4+ T cell AA demands with in vitro and multiple in vivo CRISPR screens and identify subset- and tissue-specific dependencies on the AA transporter SLC38A1 (SNAT1). While dispensable for T cell persistence and expansion over time in vitro and in vivo lung inflammation, SLC38A1 was critical for Th1 but not Th17 cell-driven Experimental Autoimmune Encephalomyelitis (EAE) and contributed to Th1 cell-driven inflammatory bowel disease. SLC38A1 deficiency reduced mTORC1 signaling and glycolytic activity in Th1 cells, in part by reducing intracellular glutamine and disrupting hexosamine biosynthesis and redox regulation. Similarly, pharmacological inhibition of SLC38 transporters delayed EAE but did not affect lung inflammation. Subset- and tissue-specific dependencies of CD4+ T cells on AA transporters may guide selective immunotherapies. HIGHLIGHTS T cells dynamically regulate glutamine amino acid transporters when activated SLC38A1 supports Th1 cell mTORC1 and proliferation by redox and hexosamine pathways Targeting SLC38A1 does not affect lung inflammation but delays IBD and EAE Nutrient transporter needs of T cell subsets vary based on disease and tissue site

[1]  Emma S. Hathaway,et al.  Differential Effects of Glutamine Inhibition Strategies on Antitumor CD8 T Cells. , 2023, Journal of immunology.

[2]  J. Rathmell,et al.  Microenvironmental influences on T cell immunity in cancer and inflammation , 2022, Cellular & Molecular Immunology.

[3]  D. Cantrell,et al.  Protein synthesis, degradation, and energy metabolism in T cell immunity , 2022, Cellular & molecular immunology.

[4]  S. Bröer,et al.  Quantitative modelling of amino acid transport and homeostasis in mammalian cells , 2021, Nature Communications.

[5]  J. Xia,et al.  MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights , 2021, Nucleic Acids Res..

[6]  Sierra M. Barone,et al.  Targeting In Vivo Metabolic Vulnerabilities of Th2 and Th17 Cells Reduces Airway Inflammation , 2021, The Journal of Immunology.

[7]  J. Rabinowitz,et al.  MTHFD2 is a Metabolic Checkpoint Controlling Effector and Regulatory T Cell Fate and Function , 2021, bioRxiv.

[8]  D. Sabatini,et al.  CRISPR screens in physiologic medium reveal conditionally essential genes in human cells , 2020, bioRxiv.

[9]  M. V. Heiden,et al.  Cell Programmed Nutrient Partitioning in the Tumor Microenvironment , 2020, bioRxiv.

[10]  A. Chinnaiyan,et al.  Cancer SLC43A2 alters T cell methionine metabolism and histone methylation , 2020, Nature.

[11]  D. Kell,et al.  The RESOLUTE consortium: unlocking SLC transporters for drug discovery , 2020, Nature Reviews Drug Discovery.

[12]  J. Gu,et al.  Loss of core fucosylation enhances the anticancer activity of cytotoxic T lymphocytes by increasing PD‐1 degradation , 2020, European journal of immunology.

[13]  Maxim N. Artyomov,et al.  Methionine Metabolism Shapes T Helper Cell Responses through Regulation of Epigenetic Reprogramming. , 2020, Cell metabolism.

[14]  J. Powell,et al.  Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion , 2019, Science.

[15]  F. Chaudhry,et al.  Multifaceted regulation of the system A transporter Slc38a2 suggests nanoscale regulation of amino acid metabolism and cellular signaling , 2019, Neuropharmacology.

[16]  E. Hinoi,et al.  Inhibition of the glutamine transporter SNAT1 confers neuroprotection in mice by modulating the mTOR-autophagy system , 2019, Communications Biology.

[17]  J. Rabinowitz,et al.  T Cell Activation Depends on Extracellular Alanine. , 2019, Cell reports.

[18]  M. Lenardo,et al.  Human Plasma-like Medium Improves T Lymphocyte Activation , 2019, bioRxiv.

[19]  Ho-Joon Lee,et al.  A large-scale analysis of targeted metabolomics data from heterogeneous biological samples provides insights into metabolite dynamics , 2019, Metabolomics.

[20]  J. Asara,et al.  Ex vivo and in vivo stable isotope labelling of central carbon metabolism and related pathways with analysis by LC–MS/MS , 2019, Nature Protocols.

[21]  Ho-Joon Lee,et al.  Meta-analysis of targeted metabolomics data from heterogeneous biological samples provides insights into metabolite dynamics , 2019, bioRxiv.

[22]  Linda V. Sinclair,et al.  Antigen receptor control of methionine metabolism in T cells , 2018, bioRxiv.

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

[24]  S. Pinho,et al.  Glycans as Key Checkpoints of T Cell Activity and Function , 2018, Front. Immunol..

[25]  Ho-Joon Lee,et al.  Macrophage Released Pyrimidines Inhibit Gemcitabine Therapy in Pancreatic Cancer , 2018, bioRxiv.

[26]  Daniel E. Carlin,et al.  A unified GenomeSpace recipe to identify essential genes and associated subnetworks from Genome-Scale CRISPR-Cas9 knockout screens , 2018, F1000Research.

[27]  R. Peebles,et al.  Testosterone Decreases House Dust Mite–Induced Type 2 and IL-17A–Mediated Airway Inflammation , 2018, The Journal of Immunology.

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

[29]  Lorin Crawford,et al.  A Landscape of Therapeutic Cooperativity in KRAS Mutant Cancers Reveals Principles for Controlling Tumor Evolution. , 2017, Cell reports.

[30]  E. Closs,et al.  Reconstitution of T Cell Proliferation under Arginine Limitation: Activated Human T Cells Take Up Citrulline via L-Type Amino Acid Transporter 1 and Use It to Regenerate Arginine after Induction of Argininosuccinate Synthase Expression , 2017, Front. Immunol..

[31]  Xin Gao,et al.  Physiologic Medium Rewires Cellular Metabolism and Reveals Uric Acid as an Endogenous Inhibitor of UMP Synthase , 2017, Cell.

[32]  Guoyao Wu,et al.  Amino-acid transporters in T-cell activation and differentiation , 2017, Cell Death & Disease.

[33]  Takla Griss,et al.  Serine Is an Essential Metabolite for Effector T Cell Expansion. , 2017, Cell metabolism.

[34]  M. Mann,et al.  L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity , 2016, Cell.

[35]  B. Metzler,et al.  Restricting Glutamine or Glutamine-Dependent Purine and Pyrimidine Syntheses Promotes Human T Cells with High FOXP3 Expression and Regulatory Properties , 2016, The Journal of Immunology.

[36]  Linda V. Sinclair,et al.  Glucose and glutamine fuel protein O-GlcNAcylation to control T cell self-renewal and malignancy , 2016, Nature Immunology.

[37]  Kutty Selva Nandakumar,et al.  System A amino acid transporters regulate glutamine uptake and attenuate antibody‐mediated arthritis , 2015, Immunology.

[38]  Meagan E. Sullender,et al.  Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.

[39]  M. Weekes,et al.  Cell Surface Proteomic Map of HIV Infection Reveals Antagonism of Amino Acid Metabolism by Vpu and Nef , 2015, Cell host & microbe.

[40]  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.

[41]  J. Locasale,et al.  Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses , 2015, Cell.

[42]  K. Giacomini,et al.  SLC transporters as therapeutic targets: emerging opportunities , 2015, Nature Reviews Drug Discovery.

[43]  T. Schoeb,et al.  Th17 cells give rise to Th1 cells that are required for the pathogenesis of colitis , 2015, Proceedings of the National Academy of Sciences.

[44]  Jun S. Liu,et al.  MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens , 2014, Genome Biology.

[45]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[46]  Neville E. Sanjana,et al.  Improved vectors and genome-wide libraries for CRISPR screening , 2014, Nature Methods.

[47]  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.

[48]  Neville E. Sanjana,et al.  Genome-Scale CRISPR-Cas9 Knockout Screening in Human Cells , 2014, Science.

[49]  H. Endou,et al.  LAT1 Is a Critical Transporter of Essential Amino Acids for Immune Reactions in Activated Human T Cells , 2013, The Journal of Immunology.

[50]  H. Schiöth,et al.  Evolutionary origin of amino acid transporter families SLC32, SLC36 and SLC38 and physiological, pathological and therapeutic aspects. , 2013, Molecular aspects of medicine.

[51]  Linda V. Sinclair,et al.  Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation , 2013, Nature Immunology.

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

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

[54]  V. Kuchroo,et al.  Th1, Th17, and Th9 Effector Cells Induce Experimental Autoimmune Encephalomyelitis with Different Pathological Phenotypes1 , 2009, The Journal of Immunology.

[55]  L. Michaux,et al.  The fusion proteins TEL-PDGFRβ and FIP1L1-PDGFRα escape ubiquitination and degradation , 2009, Haematologica.

[56]  D. Guerini,et al.  Requirement for O‐linked N‐acetylglucosaminyltransferase in lymphocytes activation , 2007, The EMBO journal.

[57]  J. Stroud,et al.  FOXP3 Controls Regulatory T Cell Function through Cooperation with NFAT , 2006, Cell.

[58]  M. Hediger,et al.  Functional Properties and Cellular Distribution of the System A Glutamine Transporter SNAT1 Support Specialized Roles in Central Neurons* , 2003, Journal of Biological Chemistry.

[59]  V. Govindaraju,et al.  Proton NMR chemical shifts and coupling constants for brain metabolites , 2000, NMR in biomedicine.