Inherent metabolic preferences differentially regulate the sensitivity of Th1 and Th2 cells to ribosome-inhibiting antibiotics
Neha Jawla
Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorRaunak Kar
Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorVeena S. Patil
Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorCorresponding Author
G. Aneeshkumar Arimbasseri
Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
Correspondence
G. Aneeshkumar Arimbasseri, Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg 110067, New Delhi.
Email: [email protected]
Search for more papers by this authorNeha Jawla
Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorRaunak Kar
Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorVeena S. Patil
Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
Search for more papers by this authorCorresponding Author
G. Aneeshkumar Arimbasseri
Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
Correspondence
G. Aneeshkumar Arimbasseri, Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg 110067, New Delhi.
Email: [email protected]
Search for more papers by this authorAbstract
Mitochondrial translation is essential to maintain mitochondrial function and energy production. Mutations in genes associated with mitochondrial translation cause several developmental disorders, and immune dysfunction is observed in many such patients. Besides genetic mutations, several antibiotics targeting bacterial ribosomes are well-established to inhibit mitochondrial translation. However, the effect of such antibiotics on different immune cells is not fully understood. Here, we addressed the differential effect of mitochondrial translation inhibition on different subsets of helper T cells (Th) of mice and humans. Inhibition of mitochondrial translation reduced the levels of mitochondrially encoded electron transport chain subunits without affecting their nuclear-encoded counterparts. As a result, mitochondrial oxygen consumption reduced dramatically, but mitochondrial mass was unaffected. Most importantly, we show that inhibition of mitochondrial translation induced apoptosis, specifically in Th2 cells. This increase in apoptosis was associated with higher expression of Bim and Puma, two activators of the intrinsic pathway of apoptosis. We propose that this difference in the sensitivity of Th1 and Th2 cells to mitochondrial translation inhibition reflects the intrinsic metabolic demands of these subtypes. Though Th1 and Th2 cells exhibit similar levels of oxidative phosphorylation, Th1 cells exhibit higher levels of aerobic glycolysis than Th2 cells. Moreover, Th1 cells are more sensitive to the inhibition of glycolysis, while higher concentrations of glycolysis inhibitor 2-deoxyglucose are required to induce cell death in the Th2 lineage. These observations reveal that selection of metabolic pathways for substrate utilization during differentiation of Th1 and Th2 lineages is a fundamental process conserved across species.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Open Research
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Supporting Information
Filename | Description |
---|---|
imm13860-sup-0001-FigureS1.pdfPDF document, 954.6 KB | Figure S1. Chloramphenicol inhibits mitochondrial ETC. (A) Representative FACS plot showing the expression of CD44 and CD62L surface markers within CD4+T cells to determine the frequency of naïve population in purified CD4+T cells. (B) Representative western blot showing protein levels of MT-CO1 (Complex IV) and SDHA (Complex II) in Th1 and Th2 cells treated with Veh and CHL. (C–E) Plots showing the activity of mitochondrial ETC Complex IV (C), ETC complex II (D) and citrate synthase (E) in Th1 and Th2 cells. (F) ATP levels estimated in the Veh and CHL-treated Th1 and Th2 cells. (G) Relative mitochondrial DNA copy number quantified using qRT-PCR in Veh and CHL-treated Th1 and Th2 cells. (H) Representative western blot showing protein levels of Drp1 and Mfn2 in Veh and CHL-treated CD4+T cells activated under neutral conditions. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (C–G). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0002-FigureS2.pdfPDF document, 1,012 KB | Figure S2. Mitochondrial translation inhibition reduces proliferation and effector functions of T cells. (A & B) Barplots showing the gene ontology terms enriched in CHL-treated Th1 (A) and Th2 (B) cells. The analysis was done using IPA software (Qiagen). (C & D) Representative western blots for p-S6 and S6 protein levels in the Veh and CHL-treated CD4+T cells in Th1 (C) and Th2 (D) conditions. (E) A bar plot showing the gene ontology terms enriched in the PC1 shown in Figure 2d. (F) Representative FACS plots showing the expression of IFN-γ in the Veh and CHL-treated CD4+T cells under neutral and Th1 polarizing conditions. (G & H). Plots showing cell counts of Th1 (G) and Th2 (H) after 48 h of rest (with IL-2, without anti-CD3/CD28 or antibiotic) of these cells that were activated for 72 h in the presence of the Veh or CHL. (I) Schematics of reactivation of Th1 and Th2 cells: Th1 and Th2 cells were reactivated with anti-CD3/CD28 for 24 h after 48 h of rest in IL-2 without CHL. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (G & H). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0003-FigureS3.pdfPDF document, 869.6 KB | Figure S3. CHL induces apoptosis in Th2 cells. (A) Quantification of T-bet and GATA-3 expressing cells in activated T cells in non-polarizing conditions expressed as the percentage of CD4+T-bet+cells and CD4+GATA-3+ cells. (B) Diagram showing the in vitro activation and differentiation of naive CD4+ T cells for 72 h followed by antibiotic treatment during culture with IL-2 for 72 h. (C) Cell count of Th1 and Th2 cells cultured with CHL for 3 days. The experimental schematic shown in Figure S3B was followed. (D & E) Quantification of proliferation based on cell trace violet (CTV) dye dilution staining after antibiotic treatment of Th1 (D) and Th2 cells (E). The experimental schematic shown in Figure S3B was followed. (F) Quantification of the frequency of live and apoptotic cells measured using Annexin and SYTOX staining in Th1 and Th2 cells treated with Veh and CHL. The experimental schematic shown in Figure 1a was followed. (G) Quantification of caspase 3/7 activity in Veh and CHL-treated Th1 and Th2 cells expressed as the percentage of Caspase 3/7+ cells. The experimental schematic shown in Figure 1a was followed. (H & I) Representative flow plots (H) and accompanying quantification (I) of Annexin V versus Propidium Iodide staining of Th1 and Th2 cells following treatment with 50 μM CHL. The numbers within the plots indicate the percentage of cells in each quarter (Q1–Q4). The experimental schematic shown in Figure S3B was followed. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (A, C, D, E, F, G, I). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0004-FigureS4.pdfPDF document, 531.4 KB | Figure S4. Bim and Puma are upregulated in CHL-treated Th2 cells. (A) Plot showing the mean fluorescence intensity (MFI) of CD95 in Th1 vs Th2 cells measured using FACS. (B) Quantification of FLIP mRNA levels in Th1 and Th2 cells using real-time qPCR. (C) Quantification of the frequency of apoptotic cells in Th1 and Th2 conditions measured using Annexin V staining. (D & E) Naïve CD4+T cells were differentiated into Th1 and Th2 for 3 days and then cultured in IL-2 for subsequent 3 days and subjected to flow cytometric analysis to determine the percentage of live (D) and apoptotic cells (E). (F) Volcano plot showing the apoptosis-related differentially expressed genes in Th1 (left panel) and Th2 (right panel) cells treated with CHL, compared with their Veh-treated counterparts. Red colour dots indicate genes upregulated in CHL-treated cells, and the golden colour dots indicate downregulated genes in CHL-treated cells. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (A–E). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0005-FigureS5.pdfPDF document, 879.1 KB | Figure S5. Th2 cells shows higher dependency on oxidative phosphorylation. (A) Heatmap showing expression of genes involved in glycolysis in Th1 and Th2 cells. Values for the heatmap are row normalized. (B & C) Plot showing basal and compensatory glycolysis in Th1 (B) and Th2 (C) cells treated with Veh and CHL as measured using glycolytic rate assay test. (D) Representative FACS plot of Annexin V versus Propidium Iodide staining of Th1 and Th2 cells following treatment with different conc. (0, 0.5, 0.8, 1 and 2 mM) of 2-DG for 72 h. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (B & C). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0006-FigureS6.pdfPDF document, 1.3 MB | Figure S6. Metabolic differences and differential response to CHL are conserved in human Th1 and Th2 cells. (A) Flow cytometric cell sorting strategy for isolation of naïve CD4+T cells from healthy donors. (B) Diagram showing the strategy for in vitro activation and differentiation of primary CD4+T cells followed by antibiotic treatment during reactivation. (C) Representative flow cytometric analysis of IFN-γ expression in Th1 and Th2 cells at day 15 of culture after PMA/ionomycin restimulation. (D) Relative mRNA expression of IL-4 in Th1 and Th2 cells after PMA/ionomycin restimulation. (E) Quantification of 2-NBDG uptake in Th1 and Th2 cells. A representative flow histogram for the same is given in Figure 6b. (F) Relative mRNA expression of the glycolytic enzyme LDHA in Th1 and Th2 cells. (G & H) Analysis of oxygen consumption rate (OCR) in Th1 and Th2 cells (G) and CHL-treated Th1 and Th2 (H) cells using a Seahorse extracellular flux analyser. (I) Cell count in Th1 and Th2 cells cultured with different conc. of CHL (25, 50 and 100 μM) for 5 days using automated cell counter. (J) Representative flow plots showing the cell trace violet (CTV) dye dilution in Th1 and Th2 cells treated with 100 μM CHL. (K) Quantification of MFI of 2-NBDG staining in Th1(left) and Th2 (right) cell treated with 100 μM CHL. (L) Glycolytic proton efflux rate (glycoPER) of Th1 (left) and Th2 (right) cells treated with 100 μM CHL as determined using a Seahorse extracellular flux analyser. Graphs show mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by paired t-test (D–I, K & L). The number of samples is denoted by the dots in the graphs. |
imm13860-sup-0007-TableS1.docxWord 2007 document , 28.3 KB | Table S1. Reagents used. |
imm13860-sup-0008-TableS2.docxWord 2007 document , 10.4 KB | Table S2. Primers used in the study. |
imm13860-sup-0009-DataS1.xlsxExcel 2007 spreadsheet , 5.1 MB | Data S1. Supporting information. |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1Ansel KM, Lee DU, Rao A. An epigenetic view of helper T cell differentiation. Nat Immunol. 2003; 4: 616–623.
- 2Bird JJ, Brown DR, Mullen AC, Moskowitz NH, Mahowald MA, Sider JR, et al. Helper T cell differentiation is controlled by the cell cycle. Immunity. 1998; 9: 229–237.
- 3Boyton R, Altmann D. Is selection for TCR affinity a factor in cytokine polarization? Trends Immunol. 2002; 23: 526–529.
- 4Zhou L, Chong MMW, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity. 2009; 30: 646–655.
- 5Krauss S, Brand MD, Buttgereit F. Signaling takes a breath-new quantitative perspectives on bioenergetics and signal transduction. Immunity. 2001; 15: 497–502.
- 6Fox CJ, Hammerman PS, Thompson CB. Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol. 2005; 5: 844–852.
- 7Jones RG, Thompson CB. Revving the engine: signal transduction fuels T cell activation. Immunity. 2007; 27: 173–178.
- 8Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011; 186: 3299–3303.
- 9Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, et al. HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011; 208: 1367–1376.
- 10Berod L, Friedrich C, Nandan A, Freitag J, Hagemann S, Harmrolfs K, et al. Erratum: De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med. 2015; 21: 414.
- 11Baixauli F, Martín-Cófreces NB, Morlino G, Carrasco YR, Calabia-Linares C, Veiga E, et al. The mitochondrial fission factor dynamin-related protein 1 modulates T-cell receptor signalling at the immune synapse: Drp1 and T-cell activation. EMBO J. 2011; 30: 1238–1250.
- 12Buck MD, O'Sullivan D, Klein Geltink RI, Curtis JD, Chang C-H, Sanin DE, et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 2016; 166: 63–76.
- 13Chao T, Wang H, Ho P-C. Mitochondrial control and guidance of cellular activities of T cells. Front Immunol. 2017; 8: 473.
- 14Corrado M, Samardžić D, Giacomello M, Rana N, Pearce EL, Scorrano L. Deletion of the mitochondria-shaping protein Opa1 during early thymocyte maturation impacts mature memory T cell metabolism. Cell Death Differ. 2021; 28: 2194–2206.
- 15Song J, Yi X, Gao R, Sun L, Wu Z, Zhang S, et al. Impact of Drp1-mediated mitochondrial dynamics on T cell immune modulation. Front Immunol. 2022; 13:873834.
- 16van der Windt GJW, Everts B, Chang C-H, Curtis JD, Freitas TC, Amiel E, et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity. 2012; 36: 68–78.
- 17D'Souza AD, Parikh N, Kaech SM, Shadel GS. Convergence of multiple signaling pathways is required to coordinately up-regulate mtDNA and mitochondrial biogenesis during T cell activation. Mitochondrion. 2007; 7: 374–385.
- 18Chang C-H, Curtis JD, Maggi LB, Faubert B, Villarino AV, O'Sullivan D, et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell. 2013; 153: 1239–1251.
- 19Kamiński MM, Sauer SW, Klemke C-D, Süss D, Okun JG, Krammer PH, et al. Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: mechanism of ciprofloxacin-mediated immunosuppression. J Immunol. 2010; 184: 4827–4841.
- 20Yi JS, Holbrook BC, Michalek RD, Laniewski NG, Grayson JM. Electron transport complex I is required for CD8+ T cell function. J Immunol. 2006; 177: 852–862.
- 21Tarasenko TN, Pacheco SE, Koenig MK, Gomez-Rodriguez J, Kapnick SM, Diaz F, et al. Cytochrome c oxidase activity is a metabolic checkpoint that regulates cell fate decisions during T cell activation and differentiation. Cell Metab. 2017; 25: 1254–1268.e7.
- 22Tabilas C, Wang J, Liu X, Locasale JW, Smith NL, Rudd BD. Cutting edge: elevated glycolytic metabolism limits the formation of memory CD8 + T cells in early life. J Immunol. 2019; 203: 2571–2576.
- 23Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest. 2013; 123: 4479–4488.
- 24Hong HS, Mbah NE, Shan M, Loesel K, Lin L, Sajjakulnukit P, et al. OXPHOS promotes apoptotic resistance and cellular persistence in T H 17 cells in the periphery and tumor microenvironment. Sci Immunol. 2022; 7:eabm8182.
- 25Alston CL, Rocha MC, Lax NZ, Turnbull DM, Taylor RW. The genetics and pathology of mitochondrial disease. J Pathol. 2017; 241: 236–250.
- 26Wallace DC. Mitochondrial diseases in man and mouse. Science. 1999; 283: 1482–1488.
- 27Wang L, Yang Z, He X, Pu S, Yang C, Wu Q, et al. Mitochondrial protein dysfunction in pathogenesis of neurological diseases. Front Mol Neurosci. 2022; 15:974480.
- 28Taylor RW, Turnbull DM. Mitochondrial DNA mutations in human disease. Nat Rev Genet. 2005; 6: 389–402.
- 29Heidari MM, Khatami M, Kamalipour A, Kalantari M, Movahed M, Emmamy MH, et al. Mitochondrial mutations in protein coding genes of respiratory chain including complexes IV, V, and mt-tRNA genes are associated risk factors for congenital heart disease. EXCLI J. 2022; 21:1306–1330 .
- 30Rötig A. Human diseases with impaired mitochondrial protein synthesis. Biochim Biophys Acta. 2011; 1807: 1198–1205.
- 31Smits P, Antonicka H, van Hasselt PM, Weraarpachai W, Haller W, Schreurs M, et al. Mutation in subdomain G' of mitochondrial elongation factor G1 is associated with combined OXPHOS deficiency in fibroblasts but not in muscle. Eur J Hum Genet. 2011; 19: 275–279.
- 32McKee EE, Ferguson M, Bentley AT, Marks TA. Inhibition of mammalian mitochondrial protein synthesis by Oxazolidinones. Antimicrob Agents Chemother. 2006; 50: 2042–2049.
- 33Singh R, Sripada L, Singh R. Side effects of antibiotics during bacterial infection: mitochondria, the main target in host cell. Mitochondrion. 2014; 16: 50–54.
- 34Colaço HG, Barros A, Neves-Costa A, Seixas E, Pedroso D, Velho T, et al. Tetracycline antibiotics induce host-dependent disease tolerance to infection. Immunity. 2021; 54: 53–67.e7.
- 35Naukkarinen J, Heinonen S, Hakkarainen A, Lundbom J, Vuolteenaho K, Saarinen L, et al. Characterising metabolically healthy obesity in weight-discordant monozygotic twins. Diabetologia. 2014; 57: 167–176.
- 36Munjampalli SKJ, Davis DE. Medicinal-induced behavior disorders. Neurol Clin. 2016; 34: 133–169.
- 37Balbi HJ. Chloramphenicol: a review. Pediatr Rev. 2004; 25: 284–288.
- 38Gajewski TF, Joyce J, Fitch FW. Antiproliferative effect of IFN-gamma in immune regulation. III. Differential selection of TH1 and TH2 murine helper T lymphocyte clones using recombinant IL-2 and recombinant IFN-gamma. J Immunol. 1989; 143: 15–22.
- 39Bauer A, Villunger A, Labi V, Fischer SF, Strasser A, Wagner H, et al. The NF-κB regulator Bcl-3 and the BH3-only proteins Bim and Puma control the death of activated T cells. Proc Natl Acad Sci U S A. 2006; 103: 10979–10984.
- 40Bean GR, Ganesan YT, Dong Y, Takeda S, Liu H, Chan PM, et al. PUMA and BIM are required for oncogene inactivation–induced apoptosis. Sci Signal. 2013; 6: ra20-ra20. doi:10.1126/scisignal.2003483
10.1126/scisignal.2003483 Google Scholar
- 41Peng M, Yin N, Chhangawala S, Xu K, Leslie CS, Li MO. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science. 2016; 354: 481–484.
- 42Tibbitt CA, Stark JM, Martens L, Ma J, Mold JE, Deswarte K, et al. Single-cell RNA sequencing of the T helper cell response to house dust mites defines a distinct gene expression signature in airway Th2 cells. Immunity. 2019; 51: 169–184.e5.
- 43Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 2006; 4: 13–24.
- 44Jin Z, Wei W, Yang M, Du Y, Wan Y. Mitochondrial complex I activity suppresses inflammation and enhances bone resorption by shifting macrophage-osteoclast polarization. Cell Metab. 2014; 20: 483–498.
- 45Baixauli F, Acín-Pérez R, Villarroya-Beltrí C, Mazzeo C, Nuñez-Andrade N, Gabandé-Rodriguez E, et al. Mitochondrial respiration controls lysosomal function during inflammatory T cell responses. Cell Metab. 2015; 22: 485–498.
- 46Desdín-Micó G, Soto-Heredero G, Aranda JF, Oller J, Carrasco E, Gabandé-Rodríguez E, et al. T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science. 2020; 368: 1371–1376.
- 47Wilhelm C, Harrison OJ, Schmitt V, Pelletier M, Spencer SP, Urban JF, et al. Critical role of fatty acid metabolism in ILC2-mediated barrier protection during malnutrition and helminth infection. J Exp Med. 2016; 213: 1409–1418.
- 48Wamelink MMC, Struys EA, Jakobs C. The biochemistry, metabolism and inherited defects of the pentose phosphate pathway: a review. J Inherit Metab Dis. 2008; 31: 703–717.
- 49Tong X, Zhao F, Thompson CB. The molecular determinants of de novo nucleotide biosynthesis in cancer cells. Curr Opin Genet Dev. 2009; 19: 32–37.
- 50Stincone A, Prigione A, Cramer T, Wamelink MMC, Campbell K, Cheung E, et al. The return of metabolism: biochemistry and physiology of the pentose phosphate pathway. Biol Rev. 2015; 90: 927–963.
- 51Varadhachary AS, Perdow SN, Hu C, Ramanarayanan M, Salgame P. Differential ability of T cell subsets to undergo activation-induced cell death. Proc Natl Acad Sci U S A. 1997; 94: 5778–5783.
- 52Zhang X, Brunner T, Carter L, Dutton RW, Rogers P, Bradley L, et al. Unequal death in T helper cell (Th)1 and Th2 effectors: Th1, but not Th2, effectors undergo rapid Fas/FasL-mediated apoptosis. J Exp Med. 1997; 185: 1837–1849.
- 53Zhang XR, Zhang LY, Devadas S, Li L, Keegan AD, Shi YF. Reciprocal expression of TRAIL and CD95L in Th1 and Th2 cells: role of apoptosis in T helper subset differentiation. Cell Death Differ. 2003; 10: 203–210.
- 54Erlacher M, Labi V, Manzl C, Böck G, Tzankov A, Häcker G, et al. Puma cooperates with Bim, the rate-limiting BH3-only protein in cell death during lymphocyte development, in apoptosis induction. J Exp Med. 2006; 203: 2939–2951.
- 55Willis SN, Adams JM. Life in the balance: how BH3-only proteins induce apoptosis. Curr Opin Cell Biol. 2005; 17: 617–625.
- 56Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, et al. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol Cell. 2005; 17: 525–535.
- 57Marani M, Tenev T, Hancock D, Downward J, Lemoine NR. Identification of novel isoforms of the BH3 domain protein Bim which directly activate Bax to trigger apoptosis. Mol Cell Biol. 2002; 22: 3577–3589.
- 58Hildeman DA, Zhu Y, Mitchell TC, Bouillet P, Strasser A, Kappler J, et al. Activated T cell death in vivo mediated by proapoptotic bcl-2 family member bim. Immunity. 2002; 16: 759–767.
- 59Pellegrini M, Belz G, Bouillet P, Strasser A. Shutdown of an acute T cell immune response to viral infection is mediated by the proapoptotic Bcl-2 homology 3-only protein Bim. Proc Natl Acad Sci U S A. 2003; 100: 14175–14180.
- 60Hughes PD, Belz GT, Fortner KA, Budd RC, Strasser A, Bouillet P. Apoptosis regulators Fas and Bim cooperate in shutdown of chronic immune responses and prevention of autoimmunity. Immunity. 2008; 28: 197–205.
- 61Jay DC, Mitchell DM, Williams MA. Bim mediates the elimination of functionally unfit Th1 responders from the memory pool. PLoS One. 2013; 8:e67363.
- 62Larsen SE, Bilenkin A, Tarasenko TN, Arjunaraja S, Stinson JR, McGuire PJ, et al. Sensitivity to restimulation-induced cell death is linked to glycolytic metabolism in human T cells. J Immunol. 2017; 198: 147–155.
- 63Secinaro MA, Fortner KA, Dienz O, Logan A, Murphy MP, Anathy V, et al. Glycolysis promotes caspase-3 activation in lipid rafts in T cells. Cell Death Dis. 2018; 9: 62.
- 64Wojciechowski S, Jordan MB, Zhu Y, White J, Zajac AJ, Hildeman DA. Bim mediates apoptosis of CD127(lo) effector T cells and limits T cell memory. Eur J Immunol. 2006; 36: 1694–1706.
- 65Lisci M, Barton PR, Randzavola LO, Ma CY, Marchingo JM, Cantrell DA, et al. Mitochondrial translation is required for sustained killing by cytotoxic T cells. Science. 2021; 374:eabe9977.
- 66Almeida L, Dhillon-LaBrooy A, Castro CN, Adossa N, Carriche GM, Guderian M, et al. Ribosome-targeting antibiotics impair T cell effector function and ameliorate autoimmunity by blocking mitochondrial protein synthesis. Immunity. 2021; 54: 68–83.e6.
- 67Han J, Kim SJ, Ryu MJ, Jang Y, Lee MJ, Ju X, et al. Chloramphenicol mitigates oxidative stress by inhibiting translation of mitochondrial complex I in dopaminergic neurons of toxin-induced Parkinson's disease model. Oxidative Med Cell Longev. 2019; 2019:4174803.
- 68Liu YJ, McIntyre RL, Janssens GE, Williams EG, Lan J, van Weeghel M, et al. Mitochondrial translation and dynamics synergistically extend lifespan in C. Elegans through HLH-30. J Cell Biol. 2020; 219:e201907067.
- 69León B, Ballesteros-Tato A. Modulating Th2 cell immunity for the treatment of asthma. Front Immunol. 2021; 12:637948.