Metabolites and Immune Response in Tumor Microenvironments
Abstract
:Simple Summary
Abstract
1. Introduction
2. Immunosuppressive TME
3. Cancer Metabolism
4. TME Metabolism
5. Fasting, Caloric Restriction, Metabolite Restriction, and Fasting Mimicking Diet (FMD)
6. Effects of Fasting and Metabolite Restriction on Cancer Cells
7. Effects of Fasting and Caloric Restriction on TME
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Elia, I.; Schmieder, R.; Christen, S.; Fendt, S.M. Organ-Specific Cancer Metabolism and Its Potential for Therapy. Handb. Exp. Pharmacol. 2016, 233, 321–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mayers, J.R.; Torrence, M.E.; Danai, L.V.; Papagiannakopoulos, T.; Davidson, S.M.; Bauer, M.R.; Lau, A.N.; Ji, B.W.; Dixit, P.D.; Hosios, A.M.; et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 2016, 353, 1161–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuneva, M.O.; Fan, T.W.; Allen, T.D.; Higashi, R.M.; Ferraris, D.V.; Tsukamoto, T.; Mates, J.M.; Alonso, F.J.; Wang, C.; Seo, Y.; et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 2012, 15, 157–170. [Google Scholar] [CrossRef] [Green Version]
- Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [Green Version]
- Hofer, S.J.; Davinelli, S.; Bergmann, M.; Scapagnini, G.; Madeo, F. Caloric Restriction Mimetics in Nutrition and Clinical Trials. Front. Nutr. 2021, 8, 717343. [Google Scholar] [CrossRef]
- Eriau, E.; Paillet, J.; Kroemer, G.; Pol, J.G. Metabolic Reprogramming by Reduced Calorie Intake or Pharmacological Caloric Restriction Mimetics for Improved Cancer Immunotherapy. Cancers 2021, 13, 1260. [Google Scholar] [CrossRef]
- Longo, V.D.; Anderson, R.M. Nutrition, longevity and disease: From molecular mechanisms to interventions. Cell 2022, 185, 1455–1470. [Google Scholar] [CrossRef]
- Nencioni, A.; Caffa, I.; Cortellino, S.; Longo, V.D. Fasting and cancer: Molecular mechanisms and clinical application. Nat. Rev. Cancer. 2018, 18, 707–719. [Google Scholar] [CrossRef]
- de Cabo, R.; Mattson, M.P. Effects of Intermittent Fasting on Health, Aging, and Disease. N. Engl. J. Med. 2019, 381, 2541–2551. [Google Scholar] [CrossRef]
- Longo, V.D.; Cortellino, S. Fasting, dietary restriction, and immunosenescence. J. Allergy Clin. Immunol. 2020, 146, 1002–1004. [Google Scholar] [CrossRef]
- Taylor, S.R.; Falcone, J.N.; Cantley, L.C.; Goncalves, M.D. Developing dietary interventions as therapy for cancer. Nat. Rev. Cancer. 2022, 22, 452–466. [Google Scholar] [CrossRef]
- Ribas, A. Adaptive Immune Resistance: How Cancer Protects from Immune Attack. Cancer Discov. 2015, 5, 915–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, P.C.; Liu, P.S. Metabolic communication in tumors: A new layer of immunoregulation for immune evasion. J. Immunother. Cancer 2016, 4, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghesquiere, B.; Wong, B.W.; Kuchnio, A.; Carmeliet, P. Metabolism of stromal and immune cells in health and disease. Nature 2014, 511, 167–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egeblad, M.; Nakasone, E.S.; Werb, Z. Tumors as organs: Complex tissues that interface with the entire organism. Dev. Cell 2010, 18, 884–901. [Google Scholar] [CrossRef] [Green Version]
- Hass, R. Role of MSC in the Tumor Microenvironment. Cancers 2020, 12, 2107. [Google Scholar] [CrossRef]
- Kersten, K.; Coffelt, S.B.; Hoogstraat, M.; Verstegen, N.J.M.; Vrijland, K.; Ciampricotti, M.; Doornebal, C.W.; Hau, C.S.; Wellenstein, M.D.; Salvagno, C.; et al. Mammary tumor-derived CCL2 enhances pro-metastatic systemic inflammation through upregulation of IL1beta in tumor-associated macrophages. Oncoimmunology 2017, 6, e1334744. [Google Scholar] [CrossRef]
- Brett, E.; Sauter, M.; Timmins, E.; Azimzadeh, O.; Rosemann, M.; Merl-Pham, J.; Hauck, S.M.; Nelson, P.J.; Becker, K.F.; Schunn, I.; et al. Oncogenic Linear Collagen VI of Invasive Breast Cancer Is Induced by CCL5. J. Clin. Med. 2020, 9, 991. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Zhang, J.; Sun, X.; Su, Q.; You, C. Down-regulation of miR-29b in carcinoma associated fibroblasts promotes cell growth and metastasis of breast cancer. Oncotarget 2017, 8, 39559–39570. [Google Scholar] [CrossRef] [Green Version]
- Le Naour, A.; Prat, M.; Thibault, B.; Mevel, R.; Lemaitre, L.; Leray, H.; Joubert, M.V.; Coulson, K.; Golzio, M.; Lefevre, L.; et al. Tumor cells educate mesenchymal stromal cells to release chemoprotective and immunomodulatory factors. J. Mol. Cell Biol. 2020, 12, 202–215. [Google Scholar] [CrossRef] [Green Version]
- Wendel, M.; Galani, I.E.; Suri-Payer, E.; Cerwenka, A. Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res. 2008, 68, 8437–8445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hensbergen, P.J.; Wijnands, P.G.; Schreurs, M.W.; Scheper, R.J.; Willemze, R.; Tensen, C.P. The CXCR3 targeting chemokine CXCL11 has potent antitumor activity in vivo involving attraction of CD8(+) T lymphocytes but not inhibition of angiogenesis. J. Immunother. 2005, 28, 343–351. [Google Scholar] [CrossRef]
- Peng, W.; Liu, C.; Xu, C.; Lou, Y.; Chen, J.; Yang, Y.; Yagita, H.; Overwijk, W.W.; Lizee, G.; Radvanyi, L.; et al. PD-1 blockade enhances T-cell migration to tumors by elevating IFN-gamma inducible chemokines. Cancer Res. 2012, 72, 5209–5218. [Google Scholar] [CrossRef] [Green Version]
- Andersson, A.; Yang, S.C.; Huang, M.; Zhu, L.; Kar, U.K.; Batra, R.K.; Elashoff, D.; Strieter, R.M.; Dubinett, S.M.; Sharma, S. IL-7 promotes CXCR3 ligand-dependent T cell antitumor reactivity in lung cancer. J. Immunol. 2009, 182, 6951–6958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
- Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
- Qian, B.Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004, 25, 677–686. [Google Scholar] [CrossRef]
- Aldinucci, D.; Colombatti, A. The inflammatory chemokine CCL5 and cancer progression. Mediat. Inflamm. 2014, 2014, 292376. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.W.; Chen, H.Y.; Wang, L.T.; Wang, F.H.; Fang, L.W.; Lai, H.Y.; Chen, H.H.; Lu, J.; Hung, M.S.; Cheng, Y.; et al. Mesenchymal stem cells tune the development of monocyte-derived dendritic cells toward a myeloid-derived suppressive phenotype through growth-regulated oncogene chemokines. J. Immunol. 2013, 190, 5065–5077. [Google Scholar] [CrossRef] [Green Version]
- Biswas, S.; Mandal, G.; Roy Chowdhury, S.; Purohit, S.; Payne, K.K.; Anadon, C.; Gupta, A.; Swanson, P.; Yu, X.; Conejo-Garcia, J.R.; et al. Exosomes Produced by Mesenchymal Stem Cells Drive Differentiation of Myeloid Cells into Immunosuppressive M2-Polarized Macrophages in Breast Cancer. J. Immunol. 2019, 203, 3447–3460. [Google Scholar] [CrossRef] [PubMed]
- Mandapathil, M.; Szczepanski, M.J.; Szajnik, M.; Ren, J.; Jackson, E.K.; Johnson, J.T.; Gorelik, E.; Lang, S.; Whiteside, T.L. Adenosine and prostaglandin E2 cooperate in the suppression of immune responses mediated by adaptive regulatory T cells. J. Biol. Chem. 2010, 285, 27571–27580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poggi, A.; Musso, A.; Dapino, I.; Zocchi, M.R. Mechanisms of tumor escape from immune system: Role of mesenchymal stromal cells. Immunol. Lett. 2014, 159, 55–72. [Google Scholar] [CrossRef]
- Najar, M.; Raicevic, G.; Fayyad-Kazan, H.; De Bruyn, C.; Bron, D.; Toungouz, M.; Lagneaux, L. Immune-related antigens, surface molecules and regulatory factors in human-derived mesenchymal stromal cells: The expression and impact of inflammatory priming. Stem Cell Rev. Rep. 2012, 8, 1188–1198. [Google Scholar] [CrossRef]
- Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef]
- Takahashi, H.; Sakakura, K.; Kudo, T.; Toyoda, M.; Kaira, K.; Oyama, T.; Chikamatsu, K. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget 2017, 8, 8633–8647. [Google Scholar] [CrossRef] [Green Version]
- Vaish, U.; Jain, T.; Are, A.C.; Dudeja, V. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma: An Update on Heterogeneity and Therapeutic Targeting. Int. J. Mol. Sci. 2021, 22, 13408. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, H.; Enomoto, A.; Woods, S.L.; Burt, A.D.; Takahashi, M.; Worthley, D.L. Cancer-associated fibroblasts in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 282–295. [Google Scholar] [CrossRef]
- Erez, N.; Truitt, M.; Olson, P.; Arron, S.T.; Hanahan, D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef] [Green Version]
- Cohen, N.; Shani, O.; Raz, Y.; Sharon, Y.; Hoffman, D.; Abramovitz, L.; Erez, N. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene 2017, 36, 4457–4468. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Lin, Y.; Shi, Y.; Li, B.; Liu, W.; Yin, W.; Dang, Y.; Chu, Y.; Fan, J.; He, R. FAP Promotes Immunosuppression by Cancer-Associated Fibroblasts in the Tumor Microenvironment via STAT3-CCL2 Signaling. Cancer Res. 2016, 76, 4124–4135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flavell, R.A.; Sanjabi, S.; Wrzesinski, S.H.; Licona-Limon, P. The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol. 2010, 10, 554–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, J.T.; Deng, Y.N.; Yi, H.M.; Wang, G.Y.; Fu, B.S.; Chen, W.J.; Liu, W.; Tai, Y.; Peng, Y.W.; Zhang, Q. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis 2016, 5, e198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gorchs, L.; Fernandez Moro, C.; Bankhead, P.; Kern, K.P.; Sadeak, I.; Meng, Q.; Rangelova, E.; Kaipe, H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4(+) and CD8(+) T-Cells. Front. Immunol. 2019, 10, 847. [Google Scholar] [CrossRef]
- Sousa, C.M.; Biancur, D.E.; Wang, X.; Halbrook, C.J.; Sherman, M.H.; Zhang, L.; Kremer, D.; Hwang, R.F.; Witkiewicz, A.K.; Ying, H.; et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016, 536, 479–483. [Google Scholar] [CrossRef] [Green Version]
- Bertero, T.; Oldham, W.M.; Grasset, E.M.; Bourget, I.; Boulter, E.; Pisano, S.; Hofman, P.; Bellvert, F.; Meneguzzi, G.; Bulavin, D.V.; et al. Tumor-Stroma Mechanics Coordinate Amino Acid Availability to Sustain Tumor Growth and Malignancy. Cell Metab. 2019, 29, 124–140.e10. [Google Scholar] [CrossRef] [Green Version]
- Salimian Rizi, B.; Caneba, C.; Nowicka, A.; Nabiyar, A.W.; Liu, X.; Chen, K.; Klopp, A.; Nagrath, D. Nitric oxide mediates metabolic coupling of omentum-derived adipose stroma to ovarian and endometrial cancer cells. Cancer Res. 2015, 75, 456–471. [Google Scholar] [CrossRef] [Green Version]
- Auciello, F.R.; Bulusu, V.; Oon, C.; Tait-Mulder, J.; Berry, M.; Bhattacharyya, S.; Tumanov, S.; Allen-Petersen, B.L.; Link, J.; Kendsersky, N.D.; et al. A Stromal Lysolipid-Autotaxin Signaling Axis Promotes Pancreatic Tumor Progression. Cancer Discov. 2019, 9, 617–627. [Google Scholar] [CrossRef] [Green Version]
- Olivares, O.; Mayers, J.R.; Gouirand, V.; Torrence, M.E.; Gicquel, T.; Borge, L.; Lac, S.; Roques, J.; Lavaut, M.N.; Berthezene, P.; et al. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat. Commun. 2017, 8, 16031. [Google Scholar] [CrossRef]
- Wu, C.; Dong, S.; Huang, R.; Chen, X. Cancer-Associated Adipocytes and Breast Cancer: Intertwining in the Tumor Microenvironment and Challenges for Cancer Therapy. Cancers 2023, 15, 726. [Google Scholar] [CrossRef]
- Sundaram, S.; Yan, L. High-fat Diet Enhances Mammary Tumorigenesis and Pulmonary Metastasis and Alters Inflammatory and Angiogenic Profiles in MMTV-PyMT Mice. AntiCancer Res. 2016, 36, 6279–6287. [Google Scholar] [CrossRef]
- Kim, S.; Yang, X.; Li, Q.; Wu, M.; Costyn, L.; Beharry, Z.; Bartlett, M.G.; Cai, H. Myristoylation of Src kinase mediates Src-induced and high-fat diet-accelerated prostate tumor progression in mice. J. Biol. Chem. 2017, 292, 18422–18433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Q.; Li, B.; Li, J.; Sun, S.; Yuan, J.; Sun, S. Cancer-associated adipocytes as immunomodulators in cancer. Biomark. Res. 2021, 9, 2. [Google Scholar] [CrossRef] [PubMed]
- Pu, X.; Chen, D. Targeting Adipokines in Obesity-Related Tumors. Front. Oncol. 2021, 11, 685923. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, A.S.; El-Shinawi, M.; Sabet, S.; Ibrahim, S.A.; Mohamed, M.M. Role of adipose tissue-derived cytokines in the progression of inflammatory breast cancer in patients with obesity. Lipids Health Dis. 2022, 21, 67. [Google Scholar] [CrossRef]
- Zhang, C.; Yue, C.; Herrmann, A.; Song, J.; Egelston, C.; Wang, T.; Zhang, Z.; Li, W.; Lee, H.; Aftabizadeh, M.; et al. STAT3 Activation-Induced Fatty Acid Oxidation in CD8(+) T Effector Cells Is Critical for Obesity-Promoted Breast Tumor Growth. Cell Metab. 2020, 31, 148–161.e5. [Google Scholar] [CrossRef]
- Ferjancic, S.; Gil-Bernabe, A.M.; Hill, S.A.; Allen, P.D.; Richardson, P.; Sparey, T.; Savory, E.; McGuffog, J.; Muschel, R.J. VCAM-1 and VAP-1 recruit myeloid cells that promote pulmonary metastasis in mice. Blood 2013, 121, 3289–3297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pittet, C.L.; Newcombe, J.; Prat, A.; Arbour, N. Human brain endothelial cells endeavor to immunoregulate CD8 T cells via PD-1 ligand expression in multiple sclerosis. J. Neuroinflamm. 2011, 8, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Motz, G.T.; Santoro, S.P.; Wang, L.P.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos, G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014, 20, 607–615. [Google Scholar] [CrossRef]
- De Palma, M.; Biziato, D.; Petrova, T.V. Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer. 2017, 17, 457–474. [Google Scholar] [CrossRef]
- Zhou, X.; Fang, D.; Liu, H.; Ou, X.; Zhang, C.; Zhao, Z.; Zhao, S.; Peng, J.; Cai, S.; He, Y.; et al. PMN-MDSCs accumulation induced by CXCL1 promotes CD8(+) T cells exhaustion in gastric cancer. Cancer Lett. 2022, 532, 215598. [Google Scholar] [CrossRef] [PubMed]
- Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [Green Version]
- Poh, A.R.; Ernst, M. Targeting Macrophages in Cancer: From Bench to Bedside. Front. Oncol. 2018, 8, 49. [Google Scholar] [CrossRef] [Green Version]
- Leek, R.D.; Hunt, N.C.; Landers, R.J.; Lewis, C.E.; Royds, J.A.; Harris, A.L. Macrophage infiltration is associated with VEGF and EGFR expression in breast cancer. J. Pathol. 2000, 190, 430–436. [Google Scholar] [CrossRef]
- Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [Green Version]
- Casazza, A.; Laoui, D.; Wenes, M.; Rizzolio, S.; Bassani, N.; Mambretti, M.; Deschoemaeker, S.; Van Ginderachter, J.A.; Tamagnone, L.; Mazzone, M. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 2013, 24, 695–709. [Google Scholar] [CrossRef] [Green Version]
- Rhee, I. Diverse macrophages polarization in tumor microenvironment. Arch. Pharm. Res. 2016, 39, 1588–1596. [Google Scholar] [CrossRef] [PubMed]
- Kuwabara, K.; Ogawa, S.; Matsumoto, M.; Koga, S.; Clauss, M.; Pinsky, D.J.; Lyn, P.; Leavy, J.; Witte, L.; Joseph-Silverstein, J.; et al. Hypoxia-mediated induction of acidic/basic fibroblast growth factor and platelet-derived growth factor in mononuclear phagocytes stimulates growth of hypoxic endothelial cells. Proc. Natl. Acad. Sci. USA 1995, 92, 4606–4610. [Google Scholar] [CrossRef]
- Harmey, J.H.; Dimitriadis, E.; Kay, E.; Redmond, H.P.; Bouchier-Hayes, D. Regulation of macrophage production of vascular endothelial growth factor (VEGF) by hypoxia and transforming growth factor beta-1. Ann. Surg. Oncol. 1998, 5, 271–278. [Google Scholar] [CrossRef]
- Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stange, G.; Van den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef] [PubMed]
- Lu, T.; Ramakrishnan, R.; Altiok, S.; Youn, J.I.; Cheng, P.; Celis, E.; Pisarev, V.; Sherman, S.; Sporn, M.B.; Gabrilovich, D. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J. Clin. Investig. 2011, 121, 4015–4029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Annamalai, R.T.; Turner, P.A.; Carson, W.F.t.; Levi, B.; Kunkel, S.; Stegemann, J.P. Harnessing macrophage-mediated degradation of gelatin microspheres for spatiotemporal control of BMP2 release. Biomaterials 2018, 161, 216–227. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Allavena, P. The interaction of anticancer therapies with tumor-associated macrophages. J. Exp. Med. 2015, 212, 435–445. [Google Scholar] [CrossRef] [PubMed]
- Bonavita, E.; Bromley, C.P.; Jonsson, G.; Pelly, V.S.; Sahoo, S.; Walwyn-Brown, K.; Mensurado, S.; Moeini, A.; Flanagan, E.; Bell, C.R.; et al. Antagonistic Inflammatory Phenotypes Dictate Tumor Fate and Response to Immune Checkpoint Blockade. Immunity 2020, 53, 1215–1229.e8. [Google Scholar] [CrossRef]
- Wculek, S.K.; Heras-Murillo, I.; Mastrangelo, A.; Mananes, D.; Galan, M.; Miguel, V.; Curtabbi, A.; Barbas, C.; Chandel, N.S.; Enriquez, J.A.; et al. Oxidative phosphorylation selectively orchestrates tissue macrophage homeostasis. Immunity 2023, 56, 516–530.e9. [Google Scholar] [CrossRef]
- Wejksza, K.; Lee-Chang, C.; Bodogai, M.; Bonzo, J.; Gonzalez, F.J.; Lehrmann, E.; Becker, K.; Biragyn, A. Cancer-produced metabolites of 5-lipoxygenase induce tumor-evoked regulatory B cells via peroxisome proliferator-activated receptor alpha. J. Immunol. 2013, 190, 2575–2584. [Google Scholar] [CrossRef] [Green Version]
- Han, S.; Feng, S.; Ren, M.; Ma, E.; Wang, X.; Xu, L.; Xu, M. Glioma cell-derived placental growth factor induces regulatory B cells. Int. J. Biochem. Cell Biol. 2014, 57, 63–68. [Google Scholar] [CrossRef]
- Schioppa, T.; Moore, R.; Thompson, R.G.; Rosser, E.C.; Kulbe, H.; Nedospasov, S.; Mauri, C.; Coussens, L.M.; Balkwill, F.R. B regulatory cells and the tumor-promoting actions of TNF-alpha during squamous carcinogenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 10662–10667. [Google Scholar] [CrossRef]
- Lindner, S.; Dahlke, K.; Sontheimer, K.; Hagn, M.; Kaltenmeier, C.; Barth, T.F.; Beyer, T.; Reister, F.; Fabricius, D.; Lotfi, R.; et al. Interleukin 21-induced granzyme B-expressing B cells infiltrate tumors and regulate T cells. Cancer Res. 2013, 73, 2468–2479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, X.; Su, Y.X.; Lao, X.M.; Liang, Y.J.; Liao, G.Q. CD19(+)IL-10(+) regulatory B cells affect survival of tongue squamous cell carcinoma patients and induce resting CD4(+) T cells to CD4(+)Foxp3(+) regulatory T cells. Oral Oncol. 2016, 53, 27–35. [Google Scholar] [CrossRef] [PubMed]
- Xiao, S.; Brooks, C.R.; Sobel, R.A.; Kuchroo, V.K. Tim-1 is essential for induction and maintenance of IL-10 in regulatory B cells and their regulation of tissue inflammation. J. Immunol. 2015, 194, 1602–1608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosser, E.C.; Mauri, C. Regulatory B cells: Origin, phenotype, and function. Immunity 2015, 42, 607–612. [Google Scholar] [CrossRef] [Green Version]
- Inoue, S.; Leitner, W.W.; Golding, B.; Scott, D. Inhibitory effects of B cells on antitumor immunity. Cancer Res. 2006, 66, 7741–7747. [Google Scholar] [CrossRef] [Green Version]
- Cai, C.; Zhang, J.; Li, M.; Wu, Z.J.; Song, K.H.; Zhan, T.W.; Wang, L.H.; Sun, Y.H. Interleukin 10-expressing B cells inhibit tumor-infiltrating T cell function and correlate with T cell Tim-3 expression in renal cell carcinoma. Tumour Biol. 2016, 37, 8209–8218. [Google Scholar] [CrossRef]
- Zhou, M.; Wen, Z.; Cheng, F.; Ma, J.; Li, W.; Ren, H.; Sheng, Y.; Dong, H.; Lu, L.; Hu, H.M.; et al. Tumor-released autophagosomes induce IL-10-producing B cells with suppressive activity on T lymphocytes via TLR2-MyD88-NF-kappaB signal pathway. Oncoimmunology 2016, 5, e1180485. [Google Scholar] [CrossRef]
- Chen, Y.Q.; Li, P.C.; Pan, N.; Gao, R.; Wen, Z.F.; Zhang, T.Y.; Huang, F.; Wu, F.Y.; Ou, X.L.; Zhang, J.P.; et al. Tumor-released autophagosomes induces CD4(+) T cell-mediated immunosuppression via a TLR2-IL-6 cascade. J. Immunother. Cancer 2019, 7, 178. [Google Scholar] [CrossRef] [Green Version]
- Horikawa, M.; Minard-Colin, V.; Matsushita, T.; Tedder, T.F. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J. Clin. Investig. 2011, 121, 4268–4280. [Google Scholar] [CrossRef] [Green Version]
- Olkhanud, P.B.; Baatar, D.; Bodogai, M.; Hakim, F.; Gress, R.; Anderson, R.L.; Deng, J.; Xu, M.; Briest, S.; Biragyn, A. Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells. Cancer Res. 2009, 69, 5996–6004. [Google Scholar] [CrossRef] [Green Version]
- Bodogai, M.; Moritoh, K.; Lee-Chang, C.; Hollander, C.M.; Sherman-Baust, C.A.; Wersto, R.P.; Araki, Y.; Miyoshi, I.; Yang, L.; Trinchieri, G.; et al. Immunosuppressive and Prometastatic Functions of Myeloid-Derived Suppressive Cells Rely upon Education from Tumor-Associated B Cells. Cancer Res. 2015, 75, 3456–3465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pylayeva-Gupta, Y.; Das, S.; Handler, J.S.; Hajdu, C.H.; Coffre, M.; Koralov, S.B.; Bar-Sagi, D. IL35-Producing B Cells Promote the Development of Pancreatic Neoplasia. Cancer Discov. 2016, 6, 247–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, K.; Liu, J.; Li, J. IL-35-producing B cells in gastric cancer patients. Medicine 2018, 97, e0710. [Google Scholar] [CrossRef]
- Shen, M.; Wang, J.; Yu, W.; Zhang, C.; Liu, M.; Wang, K.; Yang, L.; Wei, F.; Wang, S.E.; Sun, Q.; et al. A novel MDSC-induced PD-1(−)PD-L1(+) B-cell subset in breast tumor microenvironment possesses immuno-suppressive properties. Oncoimmunology 2018, 7, e1413520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, A.; Dadaglio, G.; Oberkampf, M.; Di Carlo, S.; Peduto, L.; Laubreton, D.; Desrues, B.; Sun, C.M.; Montagutelli, X.; Leclerc, C. B cells promote tumor progression in a mouse model of HPV-mediated cervical cancer. Int. J. Cancer 2016, 139, 1358–1371. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Shen, M.; Feng, Y.; He, R.; Xu, X.; Xie, Y.; Shi, X.; Zhou, M.; Pan, S.; Wang, M.; et al. Regulatory B cells induced by pancreatic cancer cell-derived interleukin-18 promote immune tolerance via the PD-1/PD-L1 pathway. Oncotarget 2018, 9, 14803–14814. [Google Scholar] [CrossRef] [Green Version]
- Tretter, T.; Venigalla, R.K.; Eckstein, V.; Saffrich, R.; Sertel, S.; Ho, A.D.; Lorenz, H.M. Induction of CD4+ T-cell anergy and apoptosis by activated human B cells. Blood 2008, 112, 4555–4564. [Google Scholar] [CrossRef] [Green Version]
- Mabrouk, I.; Buart, S.; Hasmim, M.; Michiels, C.; Connault, E.; Opolon, P.; Chiocchia, G.; Levi-Strauss, M.; Chouaib, S.; Karray, S. Prevention of autoimmunity and control of recall response to exogenous antigen by Fas death receptor ligand expression on T cells. Immunity 2008, 29, 922–933. [Google Scholar] [CrossRef] [Green Version]
- Ding, Q.; Mohib, K.; Kuchroo, V.K.; Rothstein, D.M. TIM-4 Identifies IFN-gamma-Expressing Proinflammatory B Effector 1 Cells That Promote Tumor and Allograft Rejection. J. Immunol. 2017, 199, 2585–2595. [Google Scholar] [CrossRef] [Green Version]
- Xiao, X.; Lao, X.M.; Chen, M.M.; Liu, R.X.; Wei, Y.; Ouyang, F.Z.; Chen, D.P.; Zhao, X.Y.; Zhao, Q.; Li, X.F.; et al. PD-1hi Identifies a Novel Regulatory B-cell Population in Human Hepatoma That Promotes Disease Progression. Cancer Discov. 2016, 6, 546–559. [Google Scholar] [CrossRef] [Green Version]
- Kaku, H.; Cheng, K.F.; Al-Abed, Y.; Rothstein, T.L. A novel mechanism of B cell-mediated immune suppression through CD73 expression and adenosine production. J. Immunol. 2014, 193, 5904–5913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saze, Z.; Schuler, P.J.; Hong, C.S.; Cheng, D.; Jackson, E.K.; Whiteside, T.L. Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood 2013, 122, 9–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nouel, A.; Pochard, P.; Simon, Q.; Segalen, I.; Le Meur, Y.; Pers, J.O.; Hillion, S. B-Cells induce regulatory T cells through TGF-beta/IDO production in A CTLA-4 dependent manner. J. Autoimmun. 2015, 59, 53–60. [Google Scholar] [CrossRef]
- Huang, B.; Faucette, A.N.; Pawlitz, M.D.; Pei, B.; Goyert, J.W.; Zhou, J.Z.; El-Hage, N.G.; Deng, J.; Lin, J.; Yao, F.; et al. Interleukin-33-induced expression of PIBF1 by decidual B cells protects against preterm labor. Nat. Med. 2017, 23, 128–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Fu, Y.; Yu, B.; Jiang, X.; Liu, H.; Liu, J.; Zha, B.; Chu, Y. HSP70, a Novel Regulatory Molecule in B Cell-Mediated Suppression of Autoimmune Diseases. J. Mol. Biol. 2021, 433, 166634. [Google Scholar] [CrossRef]
- Wang, L.; Simons, D.L.; Lu, X.; Tu, T.Y.; Solomon, S.; Wang, R.; Rosario, A.; Avalos, C.; Schmolze, D.; Yim, J.; et al. Connecting blood and intratumoral T(reg) cell activity in predicting future relapse in breast cancer. Nat. Immunol. 2019, 20, 1220–1230. [Google Scholar] [CrossRef] [PubMed]
- Sakaguchi, S.; Mikami, N.; Wing, J.B.; Tanaka, A.; Ichiyama, K.; Ohkura, N. Regulatory T Cells and Human Disease. Annu. Rev. Immunol. 2020, 38, 541–566. [Google Scholar] [CrossRef] [Green Version]
- Toker, A.; Ohashi, P.S. Expression of costimulatory and inhibitory receptors in FoxP3(+) regulatory T cells within the tumor microenvironment: Implications for combination immunotherapy approaches. Adv. Cancer Res. 2019, 144, 193–261. [Google Scholar] [CrossRef]
- Campbell, C.; Rudensky, A. Roles of Regulatory T Cells in Tissue Pathophysiology and Metabolism. Cell Metab. 2020, 31, 18–25. [Google Scholar] [CrossRef]
- Li, Z.; Jiang, J.; Wang, Z.; Zhang, J.; Xiao, M.; Wang, C.; Lu, Y.; Qin, Z. Endogenous interleukin-4 promotes tumor development by increasing tumor cell resistance to apoptosis. Cancer Res. 2008, 68, 8687–8694. [Google Scholar] [CrossRef] [Green Version]
- Bankaitis, K.V.; Fingleton, B. Targeting IL4/IL4R for the treatment of epithelial cancer metastasis. Clin. Exp. Metastasis 2015, 32, 847–856. [Google Scholar] [CrossRef] [Green Version]
- Fu, C.; Jiang, L.; Hao, S.; Liu, Z.; Ding, S.; Zhang, W.; Yang, X.; Li, S. Activation of the IL-4/STAT6 Signaling Pathway Promotes Lung Cancer Progression by Increasing M2 Myeloid Cells. Front. Immunol. 2019, 10, 2638. [Google Scholar] [CrossRef]
- Chen, K.; Kolls, J.K. Interluekin-17A (IL17A). Gene 2017, 614, 8–14. [Google Scholar] [CrossRef] [Green Version]
- Sullivan, J.A.; Tomita, Y.; Jankowska-Gan, E.; Lema, D.A.; Arvedson, M.P.; Nair, A.; Bracamonte-Baran, W.; Zhou, Y.; Meyer, K.K.; Zhong, W.; et al. Treg-Cell-Derived IL-35-Coated Extracellular Vesicles Promote Infectious Tolerance. Cell Rep. 2020, 30, 1039–1051.e5. [Google Scholar] [CrossRef]
- Kalia, V.; Penny, L.A.; Yuzefpolskiy, Y.; Baumann, F.M.; Sarkar, S. Quiescence of Memory CD8(+) T Cells Is Mediated by Regulatory T Cells through Inhibitory Receptor CTLA-4. Immunity 2015, 42, 1116–1129. [Google Scholar] [CrossRef] [Green Version]
- Ohta, A.; Kini, R.; Ohta, A.; Subramanian, M.; Madasu, M.; Sitkovsky, M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012, 3, 190. [Google Scholar] [CrossRef] [Green Version]
- Ihara, F.; Sakurai, D.; Takami, M.; Kamata, T.; Kunii, N.; Yamasaki, K.; Iinuma, T.; Nakayama, T.; Motohashi, S.; Okamoto, Y. Regulatory T cells induce CD4(−) NKT cell anergy and suppress NKT cell cytotoxic function. Cancer Immunol. Immunother. 2019, 68, 1935–1947. [Google Scholar] [CrossRef] [PubMed]
- Carmi, Y.; Rinott, G.; Dotan, S.; Elkabets, M.; Rider, P.; Voronov, E.; Apte, R.N. Microenvironment-derived IL-1 and IL-17 interact in the control of lung metastasis. J. Immunol. 2011, 186, 3462–3471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theocharis, A.D.; Skandalis, S.S.; Gialeli, C.; Karamanos, N.K. Extracellular matrix structure. Adv. Drug Deliv. Rev. 2016, 97, 4–27. [Google Scholar] [CrossRef] [PubMed]
- Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [Green Version]
- Butcher, D.T.; Alliston, T.; Weaver, V.M. A tense situation: Forcing tumour progression. Nat. Rev. Cancer. 2009, 9, 108–122. [Google Scholar] [CrossRef]
- Spill, F.; Reynolds, D.S.; Kamm, R.D.; Zaman, M.H. Impact of the physical microenvironment on tumor progression and metastasis. Curr. Opin. Biotechnol. 2016, 40, 41–48. [Google Scholar] [CrossRef] [Green Version]
- Shidal, C.; Singh, N.P.; Nagarkatti, P.; Nagarkatti, M. MicroRNA-92 Expression in CD133(+) Melanoma Stem Cells Regulates Immunosuppression in the Tumor Microenvironment via Integrin-Dependent Activation of TGFbeta. Cancer Res. 2019, 79, 3622–3635. [Google Scholar] [CrossRef] [Green Version]
- Steinert, G.; Scholch, S.; Niemietz, T.; Iwata, N.; Garcia, S.A.; Behrens, B.; Voigt, A.; Kloor, M.; Benner, A.; Bork, U.; et al. Immune escape and survival mechanisms in circulating tumor cells of colorectal cancer. Cancer Res. 2014, 74, 1694–1704. [Google Scholar] [CrossRef] [Green Version]
- Fallarino, F.; Grohmann, U.; Hwang, K.W.; Orabona, C.; Vacca, C.; Bianchi, R.; Belladonna, M.L.; Fioretti, M.C.; Alegre, M.L.; Puccetti, P. Modulation of tryptophan catabolism by regulatory T cells. Nat. Immunol. 2003, 4, 1206–1212. [Google Scholar] [CrossRef] [PubMed]
- Moghadasi, S.; Elveny, M.; Rahman, H.S.; Suksatan, W.; Jalil, A.T.; Abdelbasset, W.K.; Yumashev, A.V.; Shariatzadeh, S.; Motavalli, R.; Behzad, F.; et al. A paradigm shift in cell-free approach: The emerging role of MSCs-derived exosomes in regenerative medicine. J. Transl. Med. 2021, 19, 302. [Google Scholar] [CrossRef] [PubMed]
- Kalaany, N.Y.; Sabatini, D.M. Tumours with PI3K activation are resistant to dietary restriction. Nature 2009, 458, 725–731. [Google Scholar] [CrossRef] [Green Version]
- Keenan, M.M.; Chi, J.T. Alternative fuels for cancer cells. Cancer J. 2015, 21, 49–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [Green Version]
- Heintzman, D.R.; Fisher, E.L.; Rathmell, J.C. Microenvironmental influences on T cell immunity in cancer and inflammation. Cell. Mol. Immunol. 2022, 19, 316–326. [Google Scholar] [CrossRef] [PubMed]
- DeBerardinis, R.J.; Chandel, N.S. We need to talk about the Warburg effect. Nat. Metab. 2020, 2, 127–129. [Google Scholar] [CrossRef] [PubMed]
- Lunt, S.Y.; Vander Heiden, M.G. Aerobic glycolysis: Meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 2011, 27, 441–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reid, M.A.; Allen, A.E.; Liu, S.; Liberti, M.V.; Liu, P.; Liu, X.; Dai, Z.; Gao, X.; Wang, Q.; Liu, Y.; et al. Serine synthesis through PHGDH coordinates nucleotide levels by maintaining central carbon metabolism. Nat. Commun. 2018, 9, 5442. [Google Scholar] [CrossRef] [Green Version]
- Sullivan, M.R.; Mattaini, K.R.; Dennstedt, E.A.; Nguyen, A.A.; Sivanand, S.; Reilly, M.F.; Meeth, K.; Muir, A.; Darnell, A.M.; Bosenberg, M.W.; et al. Increased Serine Synthesis Provides an Advantage for Tumors Arising in Tissues Where Serine Levels Are Limiting. Cell Metab. 2019, 29, 1410–1421.e4. [Google Scholar] [CrossRef]
- Rohrig, F.; Schulze, A. The multifaceted roles of fatty acid synthesis in cancer. Nat. Rev. Cancer. 2016, 16, 732–749. [Google Scholar] [CrossRef] [PubMed]
- Possemato, R.; Marks, K.M.; Shaul, Y.D.; Pacold, M.E.; Kim, D.; Birsoy, K.; Sethumadhavan, S.; Woo, H.K.; Jang, H.G.; Jha, A.K.; et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 2011, 476, 346–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Comerford, S.A.; Huang, Z.; Du, X.; Wang, Y.; Cai, L.; Witkiewicz, A.K.; Walters, H.; Tantawy, M.N.; Fu, A.; Manning, H.C.; et al. Acetate dependence of tumors. Cell 2014, 159, 1591–1602. [Google Scholar] [CrossRef] [Green Version]
- Ho, P.C.; Bihuniak, J.D.; Macintyre, A.N.; Staron, M.; Liu, X.; Amezquita, R.; Tsui, Y.C.; Cui, G.; Micevic, G.; Perales, J.C.; et al. Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses. Cell 2015, 162, 1217–1228. [Google Scholar] [CrossRef] [Green Version]
- Ganeshan, K.; Chawla, A. Metabolic regulation of immune responses. Annu. Rev. Immunol. 2014, 32, 609–634. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [Green Version]
- Mendler, A.N.; Hu, B.; Prinz, P.U.; Kreutz, M.; Gottfried, E.; Noessner, E. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int. J. Cancer 2012, 131, 633–640. [Google Scholar] [CrossRef] [PubMed]
- Newsholme, P.; Procopio, J.; Lima, M.M.; Pithon-Curi, T.C.; Curi, R. Glutamine and glutamate--their central role in cell metabolism and function. Cell Biochem. Funct. 2003, 21, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Ahluwalia, G.S.; Grem, J.L.; Hao, Z.; Cooney, D.A. Metabolism and action of amino acid analog anti-cancer agents. Pharmacol. Ther. 1990, 46, 243–271. [Google Scholar] [CrossRef] [Green Version]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef] [Green Version]
- Bansal, A.; Simon, M.C. Glutathione metabolism in cancer progression and treatment resistance. J. Cell Biol. 2018, 217, 2291–2298. [Google Scholar] [CrossRef]
- Alkan, H.F.; Walter, K.E.; Luengo, A.; Madreiter-Sokolowski, C.T.; Stryeck, S.; Lau, A.N.; Al-Zoughbi, W.; Lewis, C.A.; Thomas, C.J.; Hoefler, G.; et al. Cytosolic Aspartate Availability Determines Cell Survival When Glutamine Is Limiting. Cell Metab. 2018, 28, 706–720.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, B.H.; Coloff, J.L. The Diverse Functions of Non-Essential Amino Acids in Cancer. Cancers 2019, 11, 675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muir, A.; Danai, L.V.; Gui, D.Y.; Waingarten, C.Y.; Lewis, C.A.; Vander Heiden, M.G. Environmental cystine drives glutamine anaplerosis and sensitizes cancer cells to glutaminase inhibition. eLife 2017, 6, e27713. [Google Scholar] [CrossRef] [PubMed]
- Konteatis, Z.; Travins, J.; Gross, S.; Marjon, K.; Barnett, A.; Mandley, E.; Nicolay, B.; Nagaraja, R.; Chen, Y.; Sun, Y.; et al. Discovery of AG-270, a First-in-Class Oral MAT2A Inhibitor for the Treatment of Tumors with Homozygous MTAP Deletion. J. Med. Chem. 2021, 64, 4430–4449. [Google Scholar] [CrossRef]
- Albers, E. Metabolic characteristics and importance of the universal methionine salvage pathway recycling methionine from 5′-methylthioadenosine. IUBMB Life 2009, 61, 1132–1142. [Google Scholar] [CrossRef]
- Kusano, T.; Berberich, T.; Tateda, C.; Takahashi, Y. Polyamines: Essential factors for growth and survival. Planta 2008, 228, 367–381. [Google Scholar] [CrossRef] [PubMed]
- Cellarier, E.; Durando, X.; Vasson, M.P.; Farges, M.C.; Demiden, A.; Maurizis, J.C.; Madelmont, J.C.; Chollet, P. Methionine dependency and cancer treatment. Cancer Treat. Rev. 2003, 29, 489–499. [Google Scholar] [CrossRef] [PubMed]
- Chaturvedi, S.; Hoffman, R.M.; Bertino, J.R. Exploiting methionine restriction for cancer treatment. Biochem. Pharmacol. 2018, 154, 170–173. [Google Scholar] [CrossRef] [PubMed]
- Palmer, R.M.; Ashton, D.S.; Moncada, S. Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature 1988, 333, 664–666. [Google Scholar] [CrossRef]
- Keshet, R.; Erez, A. Arginine and the metabolic regulation of nitric oxide synthesis in cancer. Dis. Model. Mech. 2018, 11, dmm033332. [Google Scholar] [CrossRef] [Green Version]
- Lala, P.K.; Chakraborty, C. Role of nitric oxide in carcinogenesis and tumour progression. Lancet Oncol. 2001, 2, 149–156. [Google Scholar] [CrossRef]
- Saxton, R.A.; Chantranupong, L.; Knockenhauer, K.E.; Schwartz, T.U.; Sabatini, D.M. Mechanism of arginine sensing by CASTOR1 upstream of mTORC1. Nature 2016, 536, 229–233. [Google Scholar] [CrossRef] [Green Version]
- Porstmann, T.; Santos, C.R.; Griffiths, B.; Cully, M.; Wu, M.; Leevers, S.; Griffiths, J.R.; Chung, Y.L.; Schulze, A. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8, 224–236. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Egervari, G.; Wang, Y.; Berger, S.L.; Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 2018, 19, 563–578. [Google Scholar] [CrossRef]
- Ou, Y.; Wang, S.J.; Jiang, L.; Zheng, B.; Gu, W. p53 Protein-mediated regulation of phosphoglycerate dehydrogenase (PHGDH) is crucial for the apoptotic response upon serine starvation. J. Biol. Chem. 2015, 290, 457–466. [Google Scholar] [CrossRef] [Green Version]
- Chaneton, B.; Hillmann, P.; Zheng, L.; Martin, A.C.L.; Maddocks, O.D.K.; Chokkathukalam, A.; Coyle, J.E.; Jankevics, A.; Holding, F.P.; Vousden, K.H.; et al. Serine is a natural ligand and allosteric activator of pyruvate kinase M2. Nature 2012, 491, 458–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, J.; Mancuso, A.; Tong, X.; Ward, P.S.; Fan, J.; Rabinowitz, J.D.; Thompson, C.B. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. Proc. Natl. Acad. Sci. USA 2012, 109, 6904–6909. [Google Scholar] [CrossRef] [PubMed]
- Labuschagne, C.F.; van den Broek, N.J.; Mackay, G.M.; Vousden, K.H.; Maddocks, O.D. Serine, but not glycine, supports one-carbon metabolism and proliferation of cancer cells. Cell Rep. 2014, 7, 1248–1258. [Google Scholar] [CrossRef] [Green Version]
- Pacold, M.E.; Brimacombe, K.R.; Chan, S.H.; Rohde, J.M.; Lewis, C.A.; Swier, L.J.; Possemato, R.; Chen, W.W.; Sullivan, L.B.; Fiske, B.P.; et al. A PHGDH inhibitor reveals coordination of serine synthesis and one-carbon unit fate. Nat. Chem. Biol. 2016, 12, 452–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maddocks, O.D.; Labuschagne, C.F.; Adams, P.D.; Vousden, K.H. Serine Metabolism Supports the Methionine Cycle and DNA/RNA Methylation through De Novo ATP Synthesis in Cancer Cells. Mol. Cell 2016, 61, 210–221. [Google Scholar] [CrossRef] [Green Version]
- Neinast, M.D.; Jang, C.; Hui, S.; Murashige, D.S.; Chu, Q.; Morscher, R.J.; Li, X.; Zhan, L.; White, E.; Anthony, T.G.; et al. Quantitative Analysis of the Whole-Body Metabolic Fate of Branched-Chain Amino Acids. Cell Metab. 2019, 29, 417–429.e4. [Google Scholar] [CrossRef] [Green Version]
- Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef] [Green Version]
- Puris, E.; Gynther, M.; Auriola, S.; Huttunen, K.M. L-Type amino acid transporter 1 as a target for drug delivery. Pharm. Res. 2020, 37, 88. [Google Scholar] [CrossRef]
- Yue, M.; Jiang, J.; Gao, P.; Liu, H.; Qing, G. Oncogenic MYC Activates a Feedforward Regulatory Loop Promoting Essential Amino Acid Metabolism and Tumorigenesis. Cell Rep. 2017, 21, 3819–3832. [Google Scholar] [CrossRef] [Green Version]
- Elorza, A.; Soro-Arnaiz, I.; Melendez-Rodriguez, F.; Rodriguez-Vaello, V.; Marsboom, G.; de Carcer, G.; Acosta-Iborra, B.; Albacete-Albacete, L.; Ordonez, A.; Serrano-Oviedo, L.; et al. HIF2alpha acts as an mTORC1 activator through the amino acid carrier SLC7A5. Mol. Cell 2012, 48, 681–691. [Google Scholar] [CrossRef] [Green Version]
- Grzes, K.M.; Swamy, M.; Hukelmann, J.L.; Emslie, E.; Sinclair, L.V.; Cantrell, D.A. Control of amino acid transport coordinates metabolic reprogramming in T-cell malignancy. Leukemia 2017, 31, 2771–2779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- She, P.; Reid, T.M.; Bronson, S.K.; Vary, T.C.; Hajnal, A.; Lynch, C.J.; Hutson, S.M. Disruption of BCATm in mice leads to increased energy expenditure associated with the activation of a futile protein turnover cycle. Cell Metab. 2007, 6, 181–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campbell, S.L.; Wellen, K.E. Metabolic Signaling to the Nucleus in Cancer. Mol. Cell 2018, 71, 398–408. [Google Scholar] [CrossRef] [Green Version]
- Pearce, E.L.; Poffenberger, M.C.; Chang, C.H.; Jones, R.G. Fueling immunity: Insights into metabolism and lymphocyte function. Science 2013, 342, 1242454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pearce, E.J.; Everts, B. Dendritic cell metabolism. Nat. Rev. Immunol. 2015, 15, 18–29. [Google Scholar] [CrossRef] [Green Version]
- Arner, E.N.; Rathmell, J.C. Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell 2023, 41, 421–433. [Google Scholar] [CrossRef]
- Phan, A.T.; Doedens, A.L.; Palazon, A.; Tyrakis, P.A.; Cheung, K.P.; Johnson, R.S.; Goldrath, A.W. Constitutive Glycolytic Metabolism Supports CD8(+) T Cell Effector Memory Differentiation during Viral Infection. Immunity 2016, 45, 1024–1037. [Google Scholar] [CrossRef] [Green Version]
- Kumagai, S.; Koyama, S.; Itahashi, K.; Tanegashima, T.; Lin, Y.T.; Togashi, Y.; Kamada, T.; Irie, T.; Okumura, G.; Kono, H.; et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell 2022, 40, 201–218.e9. [Google Scholar] [CrossRef]
- Xu, K.; Yin, N.; Peng, M.; Stamatiades, E.G.; Shyu, A.; Li, P.; Zhang, X.; Do, M.H.; Wang, Z.; Capistrano, K.J.; et al. Glycolysis fuels phosphoinositide 3-kinase signaling to bolster T cell immunity. Science 2021, 371, 405–410. [Google Scholar] [CrossRef]
- Guo, D.; Tong, Y.; Jiang, X.; Meng, Y.; Jiang, H.; Du, L.; Wu, Q.; Li, S.; Luo, S.; Li, M.; et al. Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IkappaBalpha. Cell Metab. 2022, 34, 1312–1324.e6. [Google Scholar] [CrossRef]
- Hermans, D.; Gautam, S.; Garcia-Canaveras, J.C.; Gromer, D.; Mitra, S.; Spolski, R.; Li, P.; Christensen, S.; Nguyen, R.; Lin, J.X.; et al. Lactate dehydrogenase inhibition synergizes with IL-21 to promote CD8(+) T cell stemness and antitumor immunity. Proc. Natl. Acad. Sci. USA 2020, 117, 6047–6055. [Google Scholar] [CrossRef]
- Chowdhury, P.S.; Chamoto, K.; Kumar, A.; Honjo, T. PPAR-Induced Fatty Acid Oxidation in T Cells Increases the Number of Tumor-Reactive CD8(+) T Cells and Facilitates Anti-PD-1 Therapy. Cancer Immunol. Res. 2018, 6, 1375–1387. [Google Scholar] [CrossRef]
- Voss, K.; Larsen, S.E.; Snow, A.L. Metabolic reprogramming and apoptosis sensitivity: Defining the contours of a T cell response. Cancer Lett. 2017, 408, 190–196. [Google Scholar] [CrossRef] [PubMed]
- Macintyre, A.N.; Gerriets, V.A.; Nichols, A.G.; Michalek, R.D.; Rudolph, M.C.; Deoliveira, D.; Anderson, S.M.; Abel, E.D.; Chen, B.J.; Hale, L.P.; et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 2014, 20, 61–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Al-Khami, A.A.; Rodriguez, P.C.; Ochoa, A.C. Metabolic reprogramming of myeloid-derived suppressor cells (MDSC) in cancer. Oncoimmunology 2016, 5, e1200771. [Google Scholar] [CrossRef] [Green Version]
- Hossain, D.M.; Panda, A.K.; Chakrabarty, S.; Bhattacharjee, P.; Kajal, K.; Mohanty, S.; Sarkar, I.; Sarkar, D.K.; Kar, S.K.; Sa, G. MEK inhibition prevents tumour-shed transforming growth factor-beta-induced T-regulatory cell augmentation in tumour milieu. Immunology 2015, 144, 561–573. [Google Scholar] [CrossRef]
- Rapoport, B.L.; Steel, H.C.; Theron, A.J.; Smit, T.; Anderson, R. Role of the Neutrophil in the Pathogenesis of Advanced Cancer and Impaired Responsiveness to Therapy. Molecules 2020, 25, 1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cham, C.M.; Driessens, G.; O’Keefe, J.P.; Gajewski, T.F. Glucose deprivation inhibits multiple key gene expression events and effector functions in CD8+ T cells. Eur. J. Immunol. 2008, 38, 2438–2450. [Google Scholar] [CrossRef] [Green Version]
- Perez-Tomas, R.; Perez-Guillen, I. Lactate in the Tumor Microenvironment: An Essential Molecule in Cancer Progression and Treatment. Cancers 2020, 12, 3244. [Google Scholar] [CrossRef]
- Wang, Z.H.; Peng, W.B.; Zhang, P.; Yang, X.P.; Zhou, Q. Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine 2021, 73, 103627. [Google Scholar] [CrossRef]
- Erra Diaz, F.; Ochoa, V.; Merlotti, A.; Dantas, E.; Mazzitelli, I.; Gonzalez Polo, V.; Sabatte, J.; Amigorena, S.; Segura, E.; Geffner, J. Extracellular Acidosis and mTOR Inhibition Drive the Differentiation of Human Monocyte-Derived Dendritic Cells. Cell Rep. 2020, 31, 107613. [Google Scholar] [CrossRef] [PubMed]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gottfried, E.; Kunz-Schughart, L.A.; Ebner, S.; Mueller-Klieser, W.; Hoves, S.; Andreesen, R.; Mackensen, A.; Kreutz, M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2006, 107, 2013–2021. [Google Scholar] [CrossRef] [Green Version]
- Nasi, A.; Fekete, T.; Krishnamurthy, A.; Snowden, S.; Rajnavolgyi, E.; Catrina, A.I.; Wheelock, C.E.; Vivar, N.; Rethi, B. Dendritic cell reprogramming by endogenously produced lactic acid. J. Immunol. 2013, 191, 3090–3099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colegio, O.R.; Chu, N.Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef] [Green Version]
- Husain, Z.; Huang, Y.; Seth, P.; Sukhatme, V.P. Tumor-derived lactate modifies antitumor immune response: Effect on myeloid-derived suppressor cells and NK cells. J. Immunol. 2013, 191, 1486–1495. [Google Scholar] [CrossRef] [Green Version]
- Munder, M.; Engelhardt, M.; Knies, D.; Medenhoff, S.; Wabnitz, G.; Luckner-Minden, C.; Feldmeyer, N.; Voss, R.H.; Kropf, P.; Muller, I.; et al. Cytotoxicity of tumor antigen specific human T cells is unimpaired by arginine depletion. PLoS ONE 2013, 8, e63521. [Google Scholar] [CrossRef] [Green Version]
- Brown, T.P.; Ganapathy, V. Lactate/GPR81 signaling and proton motive force in cancer: Role in angiogenesis, immune escape, nutrition, and Warburg phenomenon. Pharmacol. Ther. 2020, 206, 107451. [Google Scholar] [CrossRef]
- Bricker, D.K.; Taylor, E.B.; Schell, J.C.; Orsak, T.; Boutron, A.; Chen, Y.C.; Cox, J.E.; Cardon, C.M.; Van Vranken, J.G.; Dephoure, N.; et al. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science 2012, 337, 96–100. [Google Scholar] [CrossRef] [Green Version]
- Herzig, S.; Raemy, E.; Montessuit, S.; Veuthey, J.L.; Zamboni, N.; Westermann, B.; Kunji, E.R.; Martinou, J.C. Identification and functional expression of the mitochondrial pyruvate carrier. Science 2012, 337, 93–96. [Google Scholar] [CrossRef]
- Johnson, M.O.; Wolf, M.M.; Madden, M.Z.; Andrejeva, G.; Sugiura, A.; Contreras, D.C.; Maseda, D.; Liberti, M.V.; Paz, K.; Kishton, R.J.; et al. Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism. Cell 2018, 175, 1780–1795.e19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Best, S.A.; Gubser, P.M.; Sethumadhavan, S.; Kersbergen, A.; Negron Abril, Y.L.; Goldford, J.; Sellers, K.; Abeysekera, W.; Garnham, A.L.; McDonald, J.A.; et al. Glutaminase inhibition impairs CD8 T cell activation in STK11-/Lkb1-deficient lung cancer. Cell Metab. 2022, 34, 874–887.e6. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Franco, F.; Tsui, Y.C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernandez-Garcia, J.; Tsai, C.H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef]
- Leone, R.D.; Zhao, L.; Englert, J.M.; Sun, I.M.; Oh, M.H.; Sun, I.H.; Arwood, M.L.; Bettencourt, I.A.; Patel, C.H.; Wen, J.; et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019, 366, 1013–1021. [Google Scholar] [CrossRef]
- Hung, M.H.; Lee, J.S.; Ma, C.; Diggs, L.P.; Heinrich, S.; Chang, C.W.; Ma, L.; Forgues, M.; Budhu, A.; Chaisaingmongkol, J.; et al. Tumor methionine metabolism drives T-cell exhaustion in hepatocellular carcinoma. Nat. Commun. 2021, 12, 1455. [Google Scholar] [CrossRef]
- Zhao, T.; Lum, J.J. Methionine cycle-dependent regulation of T cells in cancer immunity. Front. Oncol. 2022, 12, 969563. [Google Scholar] [CrossRef]
- Geiger, R.; Rieckmann, J.C.; Wolf, T.; Basso, C.; Feng, Y.; Fuhrer, T.; Kogadeeva, M.; Picotti, P.; Meissner, F.; Mann, M.; et al. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell 2016, 167, 829–842.e13. [Google Scholar] [CrossRef] [Green Version]
- Fletcher, M.; Ramirez, M.E.; Sierra, R.A.; Raber, P.; Thevenot, P.; Al-Khami, A.A.; Sanchez-Pino, D.; Hernandez, C.; Wyczechowska, D.D.; Ochoa, A.C.; et al. l-Arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells. Cancer Res. 2015, 75, 275–283. [Google Scholar] [CrossRef] [Green Version]
- Raber, P.; Ochoa, A.C.; Rodriguez, P.C. Metabolism of L-arginine by myeloid-derived suppressor cells in cancer: Mechanisms of T cell suppression and therapeutic perspectives. Immunol. Investig. 2012, 41, 614–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carmona-Fontaine, C.; Deforet, M.; Akkari, L.; Thompson, C.B.; Joyce, J.A.; Xavier, J.B. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl. Acad. Sci. USA 2017, 114, 2934–2939. [Google Scholar] [CrossRef] [PubMed]
- Frumento, G.; Rotondo, R.; Tonetti, M.; Damonte, G.; Benatti, U.; Ferrara, G.B. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J. Exp. Med. 2002, 196, 459–468. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Sharma, M.D.; Baban, B.; Harding, H.P.; Zhang, Y.; Ron, D.; Mellor, A.L. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 2005, 22, 633–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peyraud, F.; Guegan, J.P.; Bodet, D.; Cousin, S.; Bessede, A.; Italiano, A. Targeting Tryptophan Catabolism in Cancer Immunotherapy Era: Challenges and Perspectives. Front. Immunol. 2022, 13, 807271. [Google Scholar] [CrossRef]
- Vasseur, S.; Guillaumond, F. Lipids in cancer: A global view of the contribution of lipid pathways to metastatic formation and treatment resistance. Oncogenesis 2022, 11, 46. [Google Scholar] [CrossRef] [PubMed]
- Manzo, T.; Prentice, B.M.; Anderson, K.G.; Raman, A.; Schalck, A.; Codreanu, G.S.; Nava Lauson, C.B.; Tiberti, S.; Raimondi, A.; Jones, M.A.; et al. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J. Exp. Med. 2020, 217, e20191920. [Google Scholar] [CrossRef]
- Zurier, R.B.; Rossetti, R.G.; Seiler, C.M.; Laposata, M. Human peripheral blood T lymphocyte proliferation after activation of the T cell receptor: Effects of unsaturated fatty acids. Prostaglandins Leukot. Essent. Fat. Acids 1999, 60, 371–375. [Google Scholar] [CrossRef]
- Lima, T.M.; Kanunfre, C.C.; Pompeia, C.; Verlengia, R.; Curi, R. Ranking the toxicity of fatty acids on Jurkat and Raji cells by flow cytometric analysis. Toxicol. Vitr. 2002, 16, 741–747. [Google Scholar] [CrossRef]
- Xu, S.; Chaudhary, O.; Rodriguez-Morales, P.; Sun, X.; Chen, D.; Zappasodi, R.; Xu, Z.; Pinto, A.F.M.; Williams, A.; Schulze, I.; et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8(+) T cells in tumors. Immunity 2021, 54, 1561–1577.e7. [Google Scholar] [CrossRef]
- Fernanda Cury-Boaventura, M.; Cristine Kanunfre, C.; Gorjao, R.; Martins de Lima, T.; Curi, R. Mechanisms involved in Jurkat cell death induced by oleic and linoleic acids. Clin. Nutr. 2006, 25, 1004–1014. [Google Scholar] [CrossRef]
- Stentz, F.B.; Kitabchi, A.E. Palmitic acid-induced activation of human T-lymphocytes and aortic endothelial cells with production of insulin receptors, reactive oxygen species, cytokines, and lipid peroxidation. Biochem. Biophys. Res. Commun. 2006, 346, 721–726. [Google Scholar] [CrossRef]
- Berod, L.; Friedrich, C.; Nandan, A.; Freitag, J.; Hagemann, S.; Harmrolfs, K.; Sandouk, A.; Hesse, C.; Castro, C.N.; Bahre, H.; et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 2014, 20, 1327–1333. [Google Scholar] [CrossRef]
- Beier, U.H.; Angelin, A.; Akimova, T.; Wang, L.; Liu, Y.; Xiao, H.; Koike, M.A.; Hancock, S.A.; Bhatti, T.R.; Han, R.; et al. Essential role of mitochondrial energy metabolism in Foxp3(+) T-regulatory cell function and allograft survival. FASEB J. 2015, 29, 2315–2326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, P.; Wang, Q.; Bi, E.; Ma, X.; Liu, L.; Yang, M.; Qian, J.; Yi, Q. Enhanced Lipid Accumulation and Metabolism Are Required for the Differentiation and Activation of Tumor-Associated Macrophages. Cancer Res. 2020, 80, 1438–1450. [Google Scholar] [CrossRef] [PubMed]
- Herber, D.L.; Cao, W.; Nefedova, Y.; Novitskiy, S.V.; Nagaraj, S.; Tyurin, V.A.; Corzo, A.; Cho, H.I.; Celis, E.; Lennox, B.; et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 2010, 16, 880–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pfrieger, F.W. Role of cholesterol in synapse formation and function. Biochim. Biophys. Acta 2003, 1610, 271–280. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.; Bi, E.; Lu, Y.; Su, P.; Huang, C.; Liu, L.; Wang, Q.; Yang, M.; Kalady, M.F.; Qian, J.; et al. Cholesterol Induces CD8(+) T Cell Exhaustion in the Tumor Microenvironment. Cell Metab. 2019, 30, 143–156.e5. [Google Scholar] [CrossRef]
- Mailer, R.K.W.; Gistera, A.; Polyzos, K.A.; Ketelhuth, D.F.J.; Hansson, G.K. Hypercholesterolemia Enhances T Cell Receptor Signaling and Increases the Regulatory T Cell Population. Sci. Rep. 2017, 7, 15655. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.; Longo, V.D. Fasting vs dietary restriction in cellular protection and cancer treatment: From model organisms to patients. Oncogene 2011, 30, 3305–3316. [Google Scholar] [CrossRef] [Green Version]
- Bagherniya, M.; Butler, A.E.; Barreto, G.E.; Sahebkar, A. The effect of fasting or calorie restriction on autophagy induction: A review of the literature. Ageing Res. Rev. 2018, 47, 183–197. [Google Scholar] [CrossRef]
- Longo, V.D.; Fontana, L. Calorie restriction and cancer prevention: Metabolic and molecular mechanisms. Trends Pharmacol. Sci. 2010, 31, 89–98. [Google Scholar] [CrossRef] [Green Version]
- Cangemi, A.; Fanale, D.; Rinaldi, G.; Bazan, V.; Galvano, A.; Perez, A.; Barraco, N.; Massihnia, D.; Castiglia, M.; Vieni, S.; et al. Dietary restriction: Could it be considered as speed bump on tumor progression road? Tumour Biol. 2016, 37, 7109–7118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Longo, V.D.; Mattson, M.P. Fasting: Molecular mechanisms and clinical applications. Cell Metab. 2014, 19, 181–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Groot, S.; Pijl, H.; van der Hoeven, J.J.M.; Kroep, J.R. Effects of short-term fasting on cancer treatment. J. Exp. Clin. Cancer Res. 2019, 38, 209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raffaghello, L.; Lee, C.; Safdie, F.M.; Wei, M.; Madia, F.; Bianchi, G.; Longo, V.D. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc. Natl. Acad. Sci. USA 2008, 105, 8215–8220. [Google Scholar] [CrossRef]
- Lee, C.; Safdie, F.M.; Raffaghello, L.; Wei, M.; Madia, F.; Parrella, E.; Hwang, D.; Cohen, P.; Bianchi, G.; Longo, V.D. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010, 70, 1564–1572. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.; Raffaghello, L.; Brandhorst, S.; Safdie, F.M.; Bianchi, G.; Martin-Montalvo, A.; Pistoia, V.; Wei, M.; Hwang, S.; Merlino, A.; et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci. Transl. Med. 2012, 4, 124ra127. [Google Scholar] [CrossRef] [Green Version]
- Phadngam, S.; Castiglioni, A.; Ferraresi, A.; Morani, F.; Follo, C.; Isidoro, C. PTEN dephosphorylates AKT to prevent the expression of GLUT1 on plasmamembrane and to limit glucose consumption in cancer cells. Oncotarget 2016, 7, 84999–85020. [Google Scholar] [CrossRef] [Green Version]
- Lien, E.C.; Westermark, A.M.; Zhang, Y.; Yuan, C.; Li, Z.; Lau, A.N.; Sapp, K.M.; Wolpin, B.M.; Vander Heiden, M.G. Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 2021, 599, 302–307. [Google Scholar] [CrossRef]
- Luo, H.; Chiang, H.H.; Louw, M.; Susanto, A.; Chen, D. Nutrient Sensing and the Oxidative Stress Response. Trends Endocrinol. Metab. 2017, 28, 449–460. [Google Scholar] [CrossRef]
- Yoshino, J.; Mills, K.F.; Yoon, M.J.; Imai, S. Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 2011, 14, 528–536. [Google Scholar] [CrossRef] [Green Version]
- Mai, A.; Cheng, D.; Bedford, M.T.; Valente, S.; Nebbioso, A.; Perrone, A.; Brosch, G.; Sbardella, G.; De Bellis, F.; Miceli, M.; et al. epigenetic multiple ligands: Mixed histone/protein methyltransferase, acetyltransferase, and class III deacetylase (sirtuin) inhibitors. J. Med. Chem. 2008, 51, 2279–2290. [Google Scholar] [CrossRef]
- Keppler, B.R.; Archer, T.K. Chromatin-modifying enzymes as therapeutic targets—Part 1. Expert Opin. Ther. Targets 2008, 12, 1301–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salvadori, G.; Zanardi, F.; Iannelli, F.; Lobefaro, R.; Vernieri, C.; Longo, V.D. Fasting-mimicking diet blocks triple-negative breast cancer and cancer stem cell escape. Cell Metab. 2021, 33, 2247–2259.e6. [Google Scholar] [CrossRef] [PubMed]
- Caffa, I.; Spagnolo, V.; Vernieri, C.; Valdemarin, F.; Becherini, P.; Wei, M.; Brandhorst, S.; Zucal, C.; Driehuis, E.; Ferrando, L.; et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 2020, 583, 620–624. [Google Scholar] [CrossRef]
- Fontana, L.; Adelaiye, R.M.; Rastelli, A.L.; Miles, K.M.; Ciamporcero, E.; Longo, V.D.; Nguyen, H.; Vessella, R.; Pili, R. Dietary protein restriction inhibits tumor growth in human xenograft models. Oncotarget 2013, 4, 2451–2461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amelio, I.; Melino, G.; Frezza, C. Exploiting tumour addiction with a serine and glycine-free diet. Cell Death Differ. 2017, 24, 1311–1313. [Google Scholar] [CrossRef]
- Mates, J.M.; Sanchez-Jimenez, F.M. Role of reactive oxygen species in apoptosis: Implications for cancer therapy. Int. J. Biochem. Cell Biol. 2000, 32, 157–170. [Google Scholar] [CrossRef] [PubMed]
- Di Tano, M.; Raucci, F.; Vernieri, C.; Caffa, I.; Buono, R.; Fanti, M.; Brandhorst, S.; Curigliano, G.; Nencioni, A.; de Braud, F.; et al. Synergistic effect of fasting-mimicking diet and vitamin C against KRAS mutated cancers. Nat. Commun. 2020, 11, 2332. [Google Scholar] [CrossRef]
- Komninou, D.; Leutzinger, Y.; Reddy, B.S.; Richie, J.P., Jr. Methionine restriction inhibits colon carcinogenesis. Nutr. Cancer 2006, 54, 202–208. [Google Scholar] [CrossRef]
- Sinha, R.; Cooper, T.K.; Rogers, C.J.; Sinha, I.; Turbitt, W.J.; Calcagnotto, A.; Perrone, C.E.; Richie, J.P., Jr. Dietary methionine restriction inhibits prostatic intraepithelial neoplasia in TRAMP mice. Prostate 2014, 74, 1663–1673. [Google Scholar] [CrossRef]
- Gao, X.; Sanderson, S.M.; Dai, Z.; Reid, M.A.; Cooper, D.E.; Lu, M.; Richie, J.P., Jr.; Ciccarella, A.; Calcagnotto, A.; Mikhael, P.G.; et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 2019, 572, 397–401. [Google Scholar] [CrossRef] [PubMed]
- Yoo, H.C.; Yu, Y.C.; Sung, Y.; Han, J.M. Glutamine reliance in cell metabolism. Exp. Mol. Med. 2020, 52, 1496–1516. [Google Scholar] [CrossRef] [PubMed]
- Schvartzman, J.M.; Thompson, C.B.; Finley, L.W.S. Metabolic regulation of chromatin modifications and gene expression. J. Cell Biol. 2018, 217, 2247–2259. [Google Scholar] [CrossRef]
- Tischler, J.; Gruhn, W.H.; Reid, J.; Allgeyer, E.; Buettner, F.; Marr, C.; Theis, F.; Simons, B.D.; Wernisch, L.; Surani, M.A. Metabolic regulation of pluripotency and germ cell fate through alpha-ketoglutarate. EMBO J. 2019, 38, e99518. [Google Scholar] [CrossRef] [PubMed]
- Qing, G.; Li, B.; Vu, A.; Skuli, N.; Walton, Z.E.; Liu, X.; Mayes, P.A.; Wise, D.R.; Thompson, C.B.; Maris, J.M.; et al. ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Cancer Cell 2012, 22, 631–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, M.; Reid, M.A.; Lowman, X.H.; Kulkarni, R.P.; Tran, T.Q.; Liu, X.; Yang, Y.; Hernandez-Davies, J.E.; Rosales, K.K.; Li, H.; et al. Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat. Cell Biol. 2016, 18, 1090–1101. [Google Scholar] [CrossRef] [PubMed]
- Cortellino, S.; Raveane, A.; Chiodoni, C.; Delfanti, G.; Pisati, F.; Spagnolo, V.; Visco, E.; Fragale, G.; Ferrante, F.; Magni, S.; et al. Fasting renders immunotherapy effective against low-immunogenic breast cancer while reducing side effects. Cell Rep. 2022, 40, 111256. [Google Scholar] [CrossRef]
- Lu, Z.; Xie, J.; Wu, G.; Shen, J.; Collins, R.; Chen, W.; Kang, X.; Luo, M.; Zou, Y.; Huang, L.J.; et al. Fasting selectively blocks development of acute lymphoblastic leukemia via leptin-receptor upregulation. Nat. Med. 2017, 23, 79–90. [Google Scholar] [CrossRef]
- Yoshida, K.; Inoue, T.; Nojima, K.; Hirabayashi, Y.; Sado, T. Calorie restriction reduces the incidence of myeloid leukemia induced by a single whole-body radiation in C3H/He mice. Proc. Natl. Acad. Sci. USA 1997, 94, 2615–2619. [Google Scholar] [CrossRef]
- Bradey, A.L.; Fitter, S.; Duggan, J.; Wilczek, V.; Williams, C.M.D.; Cheney, E.A.; Noll, J.E.; Tangseefa, P.; Panagopoulos, V.; Zannettino, A.C.W. Calorie restriction has no effect on bone marrow tumour burden in a Vk*MYC transplant model of multiple myeloma. Sci. Rep. 2022, 12, 13128. [Google Scholar] [CrossRef]
- Kouidhi, S.; Ben Ayed, F.; Benammar Elgaaied, A. Targeting Tumor Metabolism: A New Challenge to Improve Immunotherapy. Front. Immunol. 2018, 9, 353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, E.H.; Bantug, G.; Griss, T.; Condotta, S.; Johnson, R.M.; Samborska, B.; Mainolfi, N.; Suri, V.; Guak, H.; Balmer, M.L.; et al. Serine Is an Essential Metabolite for Effector T Cell Expansion. Cell Metab. 2017, 25, 345–357. [Google Scholar] [CrossRef]
- Roy, D.G.; Chen, J.; Mamane, V.; Ma, E.H.; Muhire, B.M.; Sheldon, R.D.; Shorstova, T.; Koning, R.; Johnson, R.M.; Esaulova, E.; et al. Methionine Metabolism Shapes T Helper Cell Responses through Regulation of Epigenetic Reprogramming. Cell Metab. 2020, 31, 250–266.e9. [Google Scholar] [CrossRef]
- O’Flanagan, C.H.; Smith, L.A.; McDonell, S.B.; Hursting, S.D. When less may be more: Calorie restriction and response to cancer therapy. BMC Med. 2017, 15, 106. [Google Scholar] [CrossRef] [Green Version]
- Hursting, S.D.; Dunlap, S.M.; Ford, N.A.; Hursting, M.J.; Lashinger, L.M. Calorie restriction and cancer prevention: A mechanistic perspective. Cancer Metab. 2013, 1, 10. [Google Scholar] [CrossRef] [Green Version]
- Pomatto-Watson, L.C.D.; Bodogai, M.; Bosompra, O.; Kato, J.; Wong, S.; Carpenter, M.; Duregon, E.; Chowdhury, D.; Krishna, P.; Ng, S.; et al. Daily caloric restriction limits tumor growth more effectively than caloric cycling regardless of dietary composition. Nat. Commun. 2021, 12, 6201. [Google Scholar] [CrossRef]
- Manukian, G.; Kivolowitz, C.; DeAngelis, T.; Shastri, A.A.; Savage, J.E.; Camphausen, K.; Rodeck, U.; Zarif, J.C.; Simone, N.L. Caloric Restriction Impairs Regulatory T cells Within the Tumor Microenvironment After Radiation and Primes Effector T cells. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 1341–1349. [Google Scholar] [CrossRef]
- Vernieri, C.; Fuca, G.; Ligorio, F.; Huber, V.; Vingiani, A.; Iannelli, F.; Raimondi, A.; Rinchai, D.; Frige, G.; Belfiore, A.; et al. Fasting-Mimicking Diet Is Safe and Reshapes Metabolism and Antitumor Immunity in Patients with Cancer. Cancer Discov. 2022, 12, 90–107. [Google Scholar] [CrossRef]
- Ferrere, G.; Tidjani Alou, M.; Liu, P.; Goubet, A.G.; Fidelle, M.; Kepp, O.; Durand, S.; Iebba, V.; Fluckiger, A.; Daillere, R.; et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight 2021, 6, e145207. [Google Scholar] [CrossRef] [PubMed]
- Ajona, D.; Ortiz-Espinosa, S.; Lozano, T.; Exposito, F.; Calvo, A.; Valencia, K.; Redrado, M.; Remirez, A.; Lecanda, F.; Alignani, D.; et al. Short-term starvation reduces IGF-1 levels to sensitize lung tumors to PD-1 immune checkpoint blockade. Nat. Cancer 2020, 1, 75–85. [Google Scholar] [CrossRef] [PubMed]
- Di Biase, S.; Lee, C.; Brandhorst, S.; Manes, B.; Buono, R.; Cheng, C.W.; Cacciottolo, M.; Martin-Montalvo, A.; de Cabo, R.; Wei, M.; et al. Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Cancer Cell 2016, 30, 136–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pietrocola, F.; Pol, J.; Vacchelli, E.; Rao, S.; Enot, D.P.; Baracco, E.E.; Levesque, S.; Castoldi, F.; Jacquelot, N.; Yamazaki, T.; et al. Caloric Restriction Mimetics Enhance Anticancer Immunosurveillance. Cancer Cell 2016, 30, 147–160. [Google Scholar] [CrossRef] [Green Version]
- Vella, V.; Lappano, R.; Bonavita, E.; Maggiolini, M.; Clarke, R.B.; Belfiore, A.; De Francesco, E.M. Insulin/IGF Axis and the Receptor for Advanced Glycation End Products: Role in Meta-inflammation and Potential in Cancer Therapy. Endocr. Rev. 2023, 44, 693–723. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yang, Q.; Liao, Q.; Li, M.; Zhang, P.; Santos, H.O.; Kord-Varkaneh, H.; Abshirini, M. Effects of intermittent fasting diets on plasma concentrations of inflammatory biomarkers: A systematic review and meta-analysis of randomized controlled trials. Nutrition 2020, 79–80, 110974. [Google Scholar] [CrossRef]
- Cortellino, S.Q.V.; Delfanti, G.; Blaževitš, O.; Chiodoni, C.; Maurea, N.; Di Mauro, A.; Tatangelo, F.; Pisati, F.; Shmahala, A.; Lazzeri, S.; et al. Fasting mimicking diet delays cancer growth and reduces immune thearapy- associated cardiovascular and systemic side effects. Nat. Commun. 2023, in press. [Google Scholar]
- Hasegawa, A.; Iwasaka, H.; Hagiwara, S.; Asai, N.; Nishida, T.; Noguchi, T. Alternate day calorie restriction improves systemic inflammation in a mouse model of sepsis induced by cecal ligation and puncture. J. Surg. Res. 2012, 174, 136–141. [Google Scholar] [CrossRef]
- Venereau, E.; Casalgrandi, M.; Schiraldi, M.; Antoine, D.J.; Cattaneo, A.; De Marchis, F.; Liu, J.; Antonelli, A.; Preti, A.; Raeli, L.; et al. Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release. J. Exp. Med. 2012, 209, 1519–1528. [Google Scholar] [CrossRef]
- Kazama, H.; Ricci, J.E.; Herndon, J.M.; Hoppe, G.; Green, D.R.; Ferguson, T.A. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity 2008, 29, 21–32. [Google Scholar] [CrossRef] [Green Version]
- Gdynia, G.; Sauer, S.W.; Kopitz, J.; Fuchs, D.; Duglova, K.; Ruppert, T.; Miller, M.; Pahl, J.; Cerwenka, A.; Enders, M.; et al. The HMGB1 protein induces a metabolic type of tumour cell death by blocking aerobic respiration. Nat. Commun. 2016, 7, 10764. [Google Scholar] [CrossRef] [Green Version]
- Hubert, P.; Roncarati, P.; Demoulin, S.; Pilard, C.; Ancion, M.; Reynders, C.; Lerho, T.; Bruyere, D.; Lebeau, A.; Radermecker, C.; et al. Extracellular HMGB1 blockade inhibits tumor growth through profoundly remodeling immune microenvironment and enhances checkpoint inhibitor-based immunotherapy. J. Immunother. Cancer 2021, 9, e001966. [Google Scholar] [CrossRef]
- Mao, Y.Q.; Huang, J.T.; Zhang, S.L.; Kong, C.; Li, Z.M.; Jing, H.; Chen, H.L.; Kong, C.Y.; Huang, S.H.; Cai, P.R.; et al. The antitumour effects of caloric restriction are mediated by the gut microbiome. Nat. Metab. 2023, 5, 96–110. [Google Scholar] [CrossRef] [PubMed]
- Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [Green Version]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
- He, Y.; Fu, L.; Li, Y.; Wang, W.; Gong, M.; Zhang, J.; Dong, X.; Huang, J.; Wang, Q.; Mackay, C.R.; et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab. 2021, 33, 988–1000.e1007. [Google Scholar] [CrossRef] [PubMed]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F., III; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, S.H.; Hu, L.P.; Wang, X.; Li, J.; Zhang, Z.G. Neurotransmitters: Emerging targets in cancer. Oncogene 2020, 39, 503–515. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Denna, T.H.; Storkersen, J.N.; Gerriets, V.A. Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol. Res. 2019, 140, 100–114. [Google Scholar] [CrossRef] [PubMed]
- Huh, J.R.; Veiga-Fernandes, H. Neuroimmune circuits in inter-organ communication. Nat. Rev. Immunol. 2020, 20, 217–228. [Google Scholar] [CrossRef]
- Cheng, C.W.; Adams, G.B.; Perin, L.; Wei, M.; Zhou, X.; Lam, B.S.; Da Sacco, S.; Mirisola, M.; Quinn, D.I.; Dorff, T.B.; et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression. Cell Stem Cell 2014, 14, 810–823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collins, N.; Han, S.J.; Enamorado, M.; Link, V.M.; Huang, B.; Moseman, E.A.; Kishton, R.J.; Shannon, J.P.; Dixit, D.; Schwab, S.R.; et al. The Bone Marrow Protects and Optimizes Immunological Memory during Dietary Restriction. Cell 2019, 178, 1088–1101.e15. [Google Scholar] [CrossRef] [PubMed]
- Vodnala, S.K.; Eil, R.; Kishton, R.J.; Sukumar, M.; Yamamoto, T.N.; Ha, N.H.; Lee, P.H.; Shin, M.; Patel, S.J.; Yu, Z.; et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 2019, 363, eaau0135. [Google Scholar] [CrossRef] [PubMed]
- Miller, B.C.; Sen, D.R.; Al Abosy, R.; Bi, K.; Virkud, Y.V.; LaFleur, M.W.; Yates, K.B.; Lako, A.; Felt, K.; Naik, G.S.; et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 2019, 20, 326–336. [Google Scholar] [CrossRef]
- Sade-Feldman, M.; Yizhak, K.; Bjorgaard, S.L.; Ray, J.P.; de Boer, C.G.; Jenkins, R.W.; Lieb, D.J.; Chen, J.H.; Frederick, D.T.; Barzily-Rokni, M.; et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 2018, 175, 998–1013.e20. [Google Scholar] [CrossRef] [Green Version]
- Utzschneider, D.T.; Charmoy, M.; Chennupati, V.; Pousse, L.; Ferreira, D.P.; Calderon-Copete, S.; Danilo, M.; Alfei, F.; Hofmann, M.; Wieland, D.; et al. T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 2016, 45, 415–427. [Google Scholar] [CrossRef] [Green Version]
- Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef] [Green Version]
- Wu, T.; Ji, Y.; Moseman, E.A.; Xu, H.C.; Manglani, M.; Kirby, M.; Anderson, S.M.; Handon, R.; Kenyon, E.; Elkahloun, A.; et al. The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 2016, 1, eaai8593. [Google Scholar] [CrossRef] [Green Version]
- Gabriel, S.S.; Tsui, C.; Chisanga, D.; Weber, F.; Llano-Leon, M.; Gubser, P.M.; Bartholin, L.; Souza-Fonseca-Guimaraes, F.; Huntington, N.D.; Shi, W.; et al. Transforming growth factor-beta-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity 2021, 54, 1698–1714.e5. [Google Scholar] [CrossRef]
- Silva-Santos, B.; Serre, K.; Norell, H. gammadelta T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef]
- Goldberg, E.L.; Shchukina, I.; Asher, J.L.; Sidorov, S.; Artyomov, M.N.; Dixit, V.D. Ketogenesis activates metabolically protective gammadelta T cells in visceral adipose tissue. Nat. Metab. 2020, 2, 50–61. [Google Scholar] [CrossRef] [PubMed]
- Yang, K.; Blanco, D.B.; Chen, X.; Dash, P.; Neale, G.; Rosencrance, C.; Easton, J.; Chen, W.; Cheng, C.; Dhungana, Y.; et al. Metabolic signaling directs the reciprocal lineage decisions of alphabeta and gammadelta T cells. Sci. Immunol. 2018, 3, eaas9818. [Google Scholar] [CrossRef] [Green Version]
- Jordan, S.; Tung, N.; Casanova-Acebes, M.; Chang, C.; Cantoni, C.; Zhang, D.; Wirtz, T.H.; Naik, S.; Rose, S.A.; Brocker, C.N.; et al. Dietary Intake Regulates the Circulating Inflammatory Monocyte Pool. Cell 2019, 178, 1102–1114.e17. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, P.; Abate, L.E.; Seyfried, T.N. Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin. Cancer Res. 2004, 10, 5622–5629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hettiarachchi, S.U.; Li, Y.H.; Roy, J.; Zhang, F.; Puchulu-Campanella, E.; Lindeman, S.D.; Srinivasarao, M.; Tsoyi, K.; Liang, X.; Ayaub, E.A.; et al. Targeted inhibition of PI3 kinase/mTOR specifically in fibrotic lung fibroblasts suppresses pulmonary fibrosis in experimental models. Sci. Transl. Med. 2020, 12, eaay3724. [Google Scholar] [CrossRef] [PubMed]
- Lane, H.A.; Wood, J.M.; McSheehy, P.M.; Allegrini, P.R.; Boulay, A.; Brueggen, J.; Littlewood-Evans, A.; Maira, S.M.; Martiny-Baron, G.; Schnell, C.R.; et al. mTOR inhibitor RAD001 (everolimus) has antiangiogenic/vascular properties distinct from a VEGFR tyrosine kinase inhibitor. Clin. Cancer Res. 2009, 15, 1612–1622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, W.; Gerald, D.; Perruzzi, C.A.; Rodriguez-Waitkus, P.; Enayati, L.; Krishnan, B.; Edmonds, J.; Hochman, M.L.; Lev, D.C.; Phung, T.L. Vascular tumors have increased p70 S6-kinase activation and are inhibited by topical rapamycin. Lab. Investig. 2013, 93, 1115–1127. [Google Scholar] [CrossRef] [Green Version]
- Lastwika, K.J.; Wilson, W., III; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res. 2016, 76, 227–238. [Google Scholar] [CrossRef] [Green Version]
- Lv, X.X.; Liu, S.S.; Hu, Z.W. Autophagy-inducing natural compounds: A treasure resource for developing therapeutics against tissue fibrosis. J. Asian Nat. Prod. Res. 2017, 19, 101–108. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cortellino, S.; Longo, V.D. Metabolites and Immune Response in Tumor Microenvironments. Cancers 2023, 15, 3898. https://doi.org/10.3390/cancers15153898
Cortellino S, Longo VD. Metabolites and Immune Response in Tumor Microenvironments. Cancers. 2023; 15(15):3898. https://doi.org/10.3390/cancers15153898
Chicago/Turabian StyleCortellino, Salvatore, and Valter D. Longo. 2023. "Metabolites and Immune Response in Tumor Microenvironments" Cancers 15, no. 15: 3898. https://doi.org/10.3390/cancers15153898
APA StyleCortellino, S., & Longo, V. D. (2023). Metabolites and Immune Response in Tumor Microenvironments. Cancers, 15(15), 3898. https://doi.org/10.3390/cancers15153898