Therapeutic opportunities to modulate immune tolerance through the metabolism-chromatin axis

Trends in Endocrinology & Metabolism - Tập 33 - Trang 507-521 - 2022
Anne Lise Ferrara1,2, Antonietta Liotti2, Antonio Pezone3, Veronica De Rosa2
1Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy
2Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy
3Dipartimento di Biologia, Università di Napoli “Federico II”, 80131 Napoli, Italy

Tài liệu tham khảo

Fuchs, 2018, Does the Danger model shed any light on central tolerance?: A response to Al-Yassin, Scand. J. Immunol., 88, 10.1111/sji.12660 Klein, 2014, Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see), Nat. Rev. Immunol., 14, 377, 10.1038/nri3667 Sakaguchi, 2008, Regulatory T cells and immune tolerance, Cell, 133, 775, 10.1016/j.cell.2008.05.009 Sogkas, 2021, Cellular and molecular mechanisms breaking immune tolerance in inborn errors of immunity, Cell. Mol. Immunol., 18, 1122, 10.1038/s41423-020-00626-z Wei, 2017, Reciprocal expression of IL-35 and IL-10 defines two distinct effector Treg subsets that are required for maintenance of immune tolerance, Cell Rep., 21, 1853, 10.1016/j.celrep.2017.10.090 Clement, 2019, Follicular regulatory T cells control humoral and allergic immunity by restraining early B cell responses, Nat. Immunol., 20, 1360, 10.1038/s41590-019-0472-4 Grover, 2021, Regulatory T cells: regulation of identity and function, Front. Immunol., 12, 10.3389/fimmu.2021.750542 Zeng, 2015, Metabolic control of regulatory T cell development and function, Trends Immunol., 36, 3, 10.1016/j.it.2014.08.003 Blagih, 2021, Tissue nutrient environments and their effect on regulatory T cell biology, Front. Immunol., 12, 10.3389/fimmu.2021.637960 He, 2017, Metabolic control of regulatory T cell (Treg) survival and function by Lkb1, Proc. Natl. Acad. Sci. U. S. A., 114, 12542, 10.1073/pnas.1715363114 Shevach, 2014, tTregs, pTregs, and iTregs: similarities and differences, Immunol. Rev., 259, 88, 10.1111/imr.12160 Joseph, 2018, Metabolic regulation of innate and adaptive lymphocyte effector responses, Immunol. Rev., 286, 137, 10.1111/imr.12703 Tian, 2011, Foxp3(+) regulatory T cells exert asymmetric control over murine helper responses by inducing Th2 cell apoptosis, Blood, 118, 1845, 10.1182/blood-2011-04-346056 De Rosa, 2015, Glycolysis controls the induction of human regulatory T cells by modulating the expression of FOXP3 exon 2 splicing variants, Nat. Immunol., 16, 1174, 10.1038/ni.3269 Carbone, 2014, Regulatory T cell proliferative potential is impaired in human autoimmune disease, Nat. Med., 20, 69, 10.1038/nm.3411 Li, 2015, Dissecting the role of the FOXP3 gene in the joint genetic susceptibility to autoimmune thyroiditis and diabetes: a genetic and functional analysis, Gene, 556, 142, 10.1016/j.gene.2014.11.064 Gounaris, 2009, T-regulatory cells shift from a protective anti-inflammatory to a cancer-promoting proinflammatory phenotype in polyposis, Cancer Res., 69, 5490, 10.1158/0008-5472.CAN-09-0304 Lu, 2017, The regulation of immune tolerance by FOXP3, Nat. Rev. Immunol., 17, 703, 10.1038/nri.2017.75 Barzaghi, 2012, Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity, Front. Immunol., 3, 211, 10.3389/fimmu.2012.00211 Lahl, 2007, Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease, J. Exp. Med., 204, 57, 10.1084/jem.20061852 Zheng, 2010, Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate, Nature, 463, 808, 10.1038/nature08750 Nair, 2016, DNA Demethylation of the Foxp3 enhancer is maintained through modulation of ten-eleven-translocation and DNA methyltransferases, Mol. Cells, 39, 888, 10.14348/molcells.2016.0276 Mikami, 2020, Epigenetic conversion of conventional T cells into regulatory T cells by CD28 signal deprivation, Proc. Natl. Acad. Sci. U. S. A., 117, 12258, 10.1073/pnas.1922600117 Jang, 2019, Homeobox protein Hhex negatively regulates Treg cells by inhibiting Foxp3 expression and function, Proc. Natl. Acad. Sci. U. S. A., 116, 25790, 10.1073/pnas.1907224116 Chatila, 2000, JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome, J. Clin. Invest., 106, R75, 10.1172/JCI11679 Mailer, 2018, Alternative splicing of FOXP3-virtue and vice, Front. Immunol., 9, 530, 10.3389/fimmu.2018.00530 Kaur, 2010, Characterisation of Foxp3 splice variants in human CD4+ and CD8+ T cells--identification of Foxp3Delta7 in human regulatory T cells, Mol. Immunol., 48, 321, 10.1016/j.molimm.2010.07.008 Fontenot, 2003, Foxp3 programs the development and function of CD4+CD25+ regulatory T cells, Nat. Immunol., 4, 330, 10.1038/ni904 Kwon, 2017, Different molecular complexes that mediate transcriptional induction and repression by FoxP3, Nat. Immunol., 18, 1238, 10.1038/ni.3835 Feng, 2014, Control of the inheritance of regulatory T cell identity by a cis element in the Foxp3 locus, Cell, 158, 749, 10.1016/j.cell.2014.07.031 Colamatteo, 2019, Molecular mechanisms controlling foxp3 expression in health and autoimmunity: from epigenetic to post-translational regulation, Front. Immunol., 10, 3136, 10.3389/fimmu.2019.03136 Tsun, 2011, Romance of the three kingdoms: RORgammat allies with HIF1alpha against FoxP3 in regulating T cell metabolism and differentiation, Protein Cell, 2, 778, 10.1007/s13238-011-1114-2 Ichiyama, 2008, Foxp3 inhibits RORgammat-mediated IL-17A mRNA transcription through direct interaction with RORgammat, J. Biol. Chem., 283, 17003, 10.1074/jbc.M801286200 Lopes, 2006, Analysis of FOXP3 reveals multiple domains required for its function as a transcriptional repressor, J. Immunol., 177, 3133, 10.4049/jimmunol.177.5.3133 Deng, 2012, Molecular and biological role of the FOXP3 N-terminal domain in immune regulation by T regulatory/suppressor cells, Exp. Mol. Pathol., 93, 334, 10.1016/j.yexmp.2012.09.013 Deng, 2020, FoxP3 in Treg cell biology: a molecular and structural perspective, Clin. Exp. Immunol., 199, 255, 10.1111/cei.13357 Voss, 2021, A guide to interrogating immunometabolism, Nat. Rev. Immunol., 21, 637, 10.1038/s41577-021-00529-8 Pearce, 2013, Metabolic pathways in immune cell activation and quiescence, Immunity, 38, 633, 10.1016/j.immuni.2013.04.005 Kempkes, 2019, Metabolic pathways involved in regulatory T cell functionality, Front. Immunol., 10, 2839, 10.3389/fimmu.2019.02839 Macintyre, 2014, The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function, Cell Metab., 20, 61, 10.1016/j.cmet.2014.05.004 Kishore, 2017, Regulatory T cell migration is dependent on glucokinase-mediated glycolysis, Immunity, 47, 875, 10.1016/j.immuni.2017.10.017 Chi, 2012, Regulation and function of mTOR signalling in T cell fate decisions, Nat. Rev. Immunol., 12, 325, 10.1038/nri3198 Blagih, 2015, The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo, Immunity, 42, 41, 10.1016/j.immuni.2014.12.030 Shyer, 2020, Metabolic signaling in T cells, Cell Res., 30, 649, 10.1038/s41422-020-0379-5 Sun, 2018, mTOR complex 1 signaling regulates the generation and function of central and effector Foxp3(+) regulatory T cells, J. Immunol., 201, 481, 10.4049/jimmunol.1701477 Saeed, 2014, Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, 345, 10.1126/science.1251086 Sun, 2009, Adaptive immune features of natural killer cells, Nature, 457, 557, 10.1038/nature07665 McCommis, 2015, Mitochondrial pyruvate transport: a historical perspective and future research directions, Biochem. J., 466, 443, 10.1042/BJ20141171 Phan, 2017, Metabolic and epigenetic coordination of T cell and macrophage immunity, Immunity, 46, 714, 10.1016/j.immuni.2017.04.016 Covarrubias, 2021, NAD(+) metabolism and its roles in cellular processes during ageing, Nat. Rev. Mol. Cell Biol., 22, 119, 10.1038/s41580-020-00313-x Avvedimento, 2021, Linking NAD metabolism and DNA repair to inflammation in SSc, Nat. Rev. Rheumatol., 17, 381, 10.1038/s41584-021-00629-8 Yang, 2016, NAD(+) metabolism: bioenergetics, signaling and manipulation for therapy, Biochim. Biophys. Acta, 1864, 1787, 10.1016/j.bbapap.2016.06.014 Luo, 2015, Roles of pyruvate, NADH, and mitochondrial complex I in redox balance and imbalance in beta cell function and dysfunction, J. Diabetes Res., 2015, 10.1155/2015/512618 Nieborak, 2018, Metabolic intermediates - cellular messengers talking to chromatin modifiers, Mol. Metab., 14, 39, 10.1016/j.molmet.2018.01.007 Zhang, 2019, PKM2, function and expression and regulation, Cell Biosci., 9, 52, 10.1186/s13578-019-0317-8 Israelsen, 2015, Pyruvate kinase: function, regulation and role in cancer, Semin. Cell Dev. Biol., 43, 43, 10.1016/j.semcdb.2015.08.004 Lee, 2022, Multiple functions of pyruvate kinase M2 in various cell types, J. Cell. Physiol., 237, 128, 10.1002/jcp.30536 Angiari, 2020, Pharmacological activation of pyruvate kinase M2 inhibits CD4(+) T cell pathogenicity and suppresses autoimmunity, Cell Metab., 31, 391, 10.1016/j.cmet.2019.10.015 Saggese, 2020, Metabolic regulation of epigenetic modifications and cell differentiation in cancer, Cancers, 12, 10.3390/cancers12123788 Kaelin, 2013, Influence of metabolism on epigenetics and disease, Cell, 153, 56, 10.1016/j.cell.2013.03.004 Losman, 2013, What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer, Genes Dev., 27, 836, 10.1101/gad.217406.113 Wellen, 2009, ATP-citrate lyase links cellular metabolism to histone acetylation, Science, 324, 1076, 10.1126/science.1164097 Hirschey, 2011, SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome, Mol. Cell, 44, 177, 10.1016/j.molcel.2011.07.019 Xu, 2017, Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism, Nature, 548, 228, 10.1038/nature23475 Frauwirth, 2002, The CD28 signaling pathway regulates glucose metabolism, Immunity, 16, 769, 10.1016/S1074-7613(02)00323-0 Merkenschlager, 2010, PI3 kinase signalling blocks Foxp3 expression by sequestering Foxo factors, J. Exp. Med., 207, 1347, 10.1084/jem.20101156 Sauer, 2008, T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR, Proc. Natl. Acad. Sci. U. S. A., 105, 7797, 10.1073/pnas.0800928105 Shin, 2020, Mitochondrial oxidative phosphorylation regulates the fate decision between pathogenic Th17 and regulatory T cells, Cell Rep., 30, 1898, 10.1016/j.celrep.2020.01.022 Basu, 2015, Foxp3-mediated inhibition of Akt inhibits Glut1 (glucose transporter 1) expression in human T regulatory cells, J. Leukoc. Biol., 97, 279, 10.1189/jlb.2AB0514-273RR Angelin, 2017, Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments, Cell Metab., 25, 1282, 10.1016/j.cmet.2016.12.018 Niculite, 2019, CD 36: focus on epigenetic and post-transcriptional regulation, Front. Genet., 10, 680, 10.3389/fgene.2019.00680 Howie, 2019, A novel role for triglyceride metabolism in Foxp3 expression, Front. Immunol., 10, 1860, 10.3389/fimmu.2019.01860 Brown, 2020, Molecular insights into regulatory T-cell adaptation to self, environment, and host tissues: plasticity or loss of function in autoimmune disease, Front. Immunol., 11, 1269, 10.3389/fimmu.2020.01269 Komatsu, 2014, Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis, Nat. Med., 20, 62, 10.1038/nm.3432 Wagner, 2021, Metabolic modeling of single Th17 cells reveals regulators of autoimmunity, Cell, 184, 4168, 10.1016/j.cell.2021.05.045 Marino, 2017, Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes, Nat. Immunol., 18, 552, 10.1038/ni.3713 Watson, 2021, Metabolic support of tumour-infiltrating regulatory T cells by lactic acid, Nature, 591, 645, 10.1038/s41586-020-03045-2 Zappasodi, 2021, CTLA-4 blockade drives loss of Treg stability in glycolysis-low tumours, Nature, 591, 652, 10.1038/s41586-021-03326-4 Wang, 2020, CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors, Nat. Immunol., 21, 298, 10.1038/s41590-019-0589-5 Picard, 2020, Relationships between immune landscapes, genetic subtypes and responses to immunotherapy in colorectal cancer, Front. Immunol., 11, 369, 10.3389/fimmu.2020.00369 Topper, 2020, The emerging role of epigenetic therapeutics in immuno-oncology, Nat. Rev. Clin. Oncol., 17, 75, 10.1038/s41571-019-0266-5 Goswami, 2018, Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy, J. Clin. Invest., 128, 3813, 10.1172/JCI99760 Perillo, 2020, LSD1: more than demethylation of histone lysine residues, Exp. Mol. Med., 52, 1936, 10.1038/s12276-020-00542-2 Pezone, 2020, Targeted DNA oxidation by LSD1-SMAD2/3 primes TGF-beta1/ EMT genes for activation or repression, Nucleic Acids Res., 48, 8943, 10.1093/nar/gkaa599 Kim, 2021, High KDM1A expression associated with decreased CD8+T cells reduces the breast cancer survival rate in patients with breast cancer, J. Clin. Med., 10 Andrikopoulou, 2020, The emerging role of BET inhibitors in breast cancer, Breast, 53, 152, 10.1016/j.breast.2020.08.005 Adeegbe, 2018, BET bromodomain inhibition cooperates with PD-1 blockade to facilitate antitumor response in Kras-mutant non-small cell lung cancer, Cancer Immunol. Res., 6, 1234, 10.1158/2326-6066.CIR-18-0077 Diehl, 2020, Chromatin as a key consumer in the metabolite economy, Nat. Chem. Biol., 16, 620, 10.1038/s41589-020-0517-x Awad, 2021, Selective histone deacetylase inhibitor ACY-241 (citarinostat) plus nivolumab in advanced non-small cell lung cancer: results from a phase Ib study, Front. Oncol., 11, 10.3389/fonc.2021.696512 Yoon, 2016, HDAC and HDAC inhibitor: from cancer to cardiovascular diseases, Chonnam Med. J., 52, 1, 10.4068/cmj.2016.52.1.1 Qiu, 2013, Effects of treatment with histone deacetylase inhibitors in solid tumors: a review based on 30 clinical trials, Future Oncol., 9, 255, 10.2217/fon.12.173 Choy, 2015, Phase 1 study of oral abexinostat, a histone deacetylase inhibitor, in combination with doxorubicin in patients with metastatic sarcoma, Cancer, 121, 1223, 10.1002/cncr.29175 Pleyer, 2017, Azacitidine for front-line therapy of patients with AML: reproducible efficacy established by direct comparison of international phase 3 trial data with registry data from the Austrian Azacitidine Registry of the AGMT Study Group, Int. J. Mol. Sci., 18, 10.3390/ijms18020415 Zeidan, 2021, A randomized phase 2 trial of azacitidine +/- durvalumab as first-line therapy for higher-risk myelodysplastic syndromes, Blood Adv. Lu, 2007, Demethylation of CD40LG on the inactive X in T cells from women with lupus, J. Immunol., 179, 6352, 10.4049/jimmunol.179.9.6352 Dees, 2014, The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis, Ann. Rheum. Dis., 73, 1232, 10.1136/annrheumdis-2012-203194 Konsta, 2016, Epigenetic modifications in salivary glands from patients with Sjogren's syndrome affect cytokeratin 19 expression, Bull. Group. Int. Rech. Sci. Stomatol. Odontol., 53 Marks, 2005, Histone deacetylase inhibitors: discovery and development as anticancer agents, Expert Opin. Investig. Drugs, 14, 1497, 10.1517/13543784.14.12.1497 Patel, 2011, Chromatin remodeling resets the immune system to protect against autoimmune diabetes in mice, Immunol. Cell Biol., 89, 640, 10.1038/icb.2010.144 Vojinovic, 2011, Safety and efficacy of an oral histone deacetylase inhibitor in systemic-onset juvenile idiopathic arthritis, Arthritis Rheum., 63, 1452, 10.1002/art.30238 Italiano, 2018, Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study, Lancet Oncol., 19, 649, 10.1016/S1470-2045(18)30145-1 Rugo, 2020, The promise for histone methyltransferase inhibitors for epigenetic therapy in clinical oncology: a narrative review, Adv. Ther., 37, 3059, 10.1007/s12325-020-01379-x Cousin, 2022, Safety, pharmacokinetic, pharmacodynamic and clinical activity of molibresib for the treatment of nuclear protein in testis carcinoma and other cancers: results of a Phase I/II open-label, dose escalation study, Int. J. Cancer, 150, 993, 10.1002/ijc.33861 Shapiro, 2021, A Phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma, Br. J. Cancer, 124, 744, 10.1038/s41416-020-01180-1 Sun, 2020, Safety and efficacy of bromodomain and extra-terminal inhibitors for the treatment of hematological malignancies and solid tumors: a systematic study of clinical trials, Front. Pharmacol., 11 Reyes-Garau, 2019, Pharmacological targeting of BET bromodomain proteins in acute myeloid leukemia and malignant lymphomas: from molecular characterization to clinical applications, Cancers, 11, 10.3390/cancers11101483 Wass, 2021, A proof of concept phase I/II pilot trial of LSD1 inhibition by tranylcypromine combined with ATRA in refractory/relapsed AML patients not eligible for intensive therapy, Leukemia, 35, 701, 10.1038/s41375-020-0892-z Zhou, 2021, KDM1A inhibition is effective in reducing stemness and treating triple negative breast cancer, Breast Cancer Res. Treat., 185, 343, 10.1007/s10549-020-05963-1 Tullius, 2014, NAD+ protects against EAE by regulating CD4+ T-cell differentiation, Nat. Commun., 5, 5101, 10.1038/ncomms6101