1
|
Ricci JE. Tumor-induced metabolic immunosuppression: Mechanisms and therapeutic targets. Cell Rep 2025; 44:115206. [PMID: 39798090 DOI: 10.1016/j.celrep.2024.115206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
Metabolic reprogramming in both immune and cancer cells plays a crucial role in the antitumor immune response. Recent studies indicate that cancer metabolism not only sustains carcinogenesis and survival via altered signaling but also modulates immune cell function. Metabolic crosstalk within the tumor microenvironment results in nutrient competition and acidosis, thereby hindering immune cell functionality. Interestingly, immune cells also undergo metabolic reprogramming that enables their proliferation, differentiation, and effector functions. This review highlights the regulation of antitumor immune responses through metabolic reprogramming in cancer and immune cells and explores therapeutic strategies that target these metabolic pathways in cancer immunotherapy, including using chimeric antigen receptor (CAR)-T cells. We discuss innovative combinations of immunotherapy, cellular therapies, and metabolic interventions that could optimize the efficacy of existing treatment protocols.
Collapse
Affiliation(s)
- Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice, France; Équipe labellisée LIGUE Contre le Cancer, Nice, France.
| |
Collapse
|
2
|
Hecht F, Zocchi M, Tuttle ET, Ward NP, Smith B, Kang YP, Cazarin J, Soares ZG, Ozgurses ME, Zhao H, Sheehan C, Alimohammadi F, Munger LD, Trivedi D, Asantewaa G, Blick-Nitko SK, Zoeller JJ, Chen Y, Vasiliou V, Turner BM, Muir A, Coloff JL, Munger J, DeNicola GM, Harris IS. Catabolism of extracellular glutathione supplies amino acids to support tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617667. [PMID: 39416022 PMCID: PMC11482906 DOI: 10.1101/2024.10.10.617667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Restricting amino acids from tumors is an emerging therapeutic strategy with significant promise. While typically considered an intracellular antioxidant with tumor-promoting capabilities, glutathione (GSH) is a tripeptide of cysteine, glutamate, and glycine that can be catabolized, yielding amino acids. The extent to which GSH-derived amino acids are essential to cancers is unclear. Here, we find that GSH catabolism promotes tumor growth. We show that depletion of intracellular GSH does not perturb tumor growth, and extracellular GSH is highly abundant in the tumor microenvironment, highlighting the potential importance of GSH outside of tumors. We find supplementation with GSH can rescue cancer cell survival and growth in cystine-deficient conditions, and this rescue is dependent on the catabolic activity of γ-glutamyltransferases (GGTs). Finally, pharmacologic targeting of GGTs' activity prevents the breakdown of circulating GSH, lowers tumor cysteine levels, and slows tumor growth. Our findings indicate a non-canonical role for GSH in supporting tumors by acting as a reservoir of amino acids. Depriving tumors of extracellular GSH or inhibiting its breakdown is potentially a therapeutically tractable approach for patients with cancer. Further, these findings change our view of GSH and how amino acids, including cysteine, are supplied to cells.
Collapse
Affiliation(s)
- Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- These authors contributed equally
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- These authors contributed equally
| | - Emily T. Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Nathan P. Ward
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Bradley Smith
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Yun Pyo Kang
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Zamira G. Soares
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Mete Emir Ozgurses
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Huiping Zhao
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Colin Sheehan
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Fatemeh Alimohammadi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Lila D. Munger
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Dhvani Trivedi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Gloria Asantewaa
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Sara K. Blick-Nitko
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Jason J. Zoeller
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA, 02115
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06510
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA, 06510
| | - Bradley M. Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA, 14620
| | - Alexander Muir
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Joshua Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA, 14642
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA, 33612
| | - Isaac S. Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA, 14620
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA, 14620
| |
Collapse
|
3
|
Fan H, Xia S, Xiang J, Li Y, Ross MO, Lim SA, Yang F, Tu J, Xie L, Dougherty U, Zhang FQ, Zheng Z, Zhang R, Wu R, Dong L, Su R, Chen X, Althaus T, Riedell PA, Jonker PB, Muir A, Lesinski GB, Rafiq S, Dhodapkar MV, Stock W, Odenike O, Patel AA, Opferman J, Tsuji T, Matsuzaki J, Shah H, Faubert B, Elf SE, Layden B, Bissonnette BM, He YY, Kline J, Mao H, Odunsi K, Gao X, Chi H, He C, Chen J. Trans-vaccenic acid reprograms CD8 + T cells and anti-tumour immunity. Nature 2023; 623:1034-1043. [PMID: 37993715 PMCID: PMC10686835 DOI: 10.1038/s41586-023-06749-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/16/2023] [Indexed: 11/24/2023]
Abstract
Diet-derived nutrients are inextricably linked to human physiology by providing energy and biosynthetic building blocks and by functioning as regulatory molecules. However, the mechanisms by which circulating nutrients in the human body influence specific physiological processes remain largely unknown. Here we use a blood nutrient compound library-based screening approach to demonstrate that dietary trans-vaccenic acid (TVA) directly promotes effector CD8+ T cell function and anti-tumour immunity in vivo. TVA is the predominant form of trans-fatty acids enriched in human milk, but the human body cannot produce TVA endogenously1. Circulating TVA in humans is mainly from ruminant-derived foods including beef, lamb and dairy products such as milk and butter2,3, but only around 19% or 12% of dietary TVA is converted to rumenic acid by humans or mice, respectively4,5. Mechanistically, TVA inactivates the cell-surface receptor GPR43, an immunomodulatory G protein-coupled receptor activated by its short-chain fatty acid ligands6-8. TVA thus antagonizes the short-chain fatty acid agonists of GPR43, leading to activation of the cAMP-PKA-CREB axis for enhanced CD8+ T cell function. These findings reveal that diet-derived TVA represents a mechanism for host-extrinsic reprogramming of CD8+ T cells as opposed to the intrahost gut microbiota-derived short-chain fatty acids. TVA thus has translational potential for the treatment of tumours.
Collapse
Affiliation(s)
- Hao Fan
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Siyuan Xia
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Junhong Xiang
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Yuancheng Li
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Matthew O Ross
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Seon Ah Lim
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Fan Yang
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Jiayi Tu
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Lishi Xie
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Freya Q Zhang
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Zhong Zheng
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Rukang Zhang
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Rong Wu
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Lei Dong
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Xiufen Chen
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Thomas Althaus
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Peter A Riedell
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Patrick B Jonker
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Alexander Muir
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wendy Stock
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Anand A Patel
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Joseph Opferman
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Takemasa Tsuji
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL, USA
| | - Junko Matsuzaki
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL, USA
| | - Hardik Shah
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Brandon Faubert
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Shannon E Elf
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Brian Layden
- Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | | | - Yu-Ying He
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Hui Mao
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL, USA
| | - Xue Gao
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Hongbo Chi
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
| | - Jing Chen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Apiz Saab JJ, Dzierozynski LN, Jonker PB, AminiTabrizi R, Shah H, Menjivar RE, Scott AJ, Nwosu ZC, Zhu Z, Chen RN, Oh M, Sheehan C, Wahl DR, Pasca di Magliano M, Lyssiotis CA, Macleod KF, Weber CR, Muir A. Pancreatic tumors exhibit myeloid-driven amino acid stress and upregulate arginine biosynthesis. eLife 2023; 12:e81289. [PMID: 37254839 PMCID: PMC10260022 DOI: 10.7554/elife.81289] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 05/25/2023] [Indexed: 06/01/2023] Open
Abstract
Nutrient stress in the tumor microenvironment requires cancer cells to adopt adaptive metabolic programs for survival and proliferation. Therefore, knowledge of microenvironmental nutrient levels and how cancer cells cope with such nutrition is critical to understand the metabolism underpinning cancer cell biology. Previously, we performed quantitative metabolomics of the interstitial fluid (the local perfusate) of murine pancreatic ductal adenocarcinoma (PDAC) tumors to comprehensively characterize nutrient availability in the microenvironment of these tumors. Here, we develop Tumor Interstitial Fluid Medium (TIFM), a cell culture medium that contains nutrient levels representative of the PDAC microenvironment, enabling us to study PDAC metabolism ex vivo under physiological nutrient conditions. We show that PDAC cells cultured in TIFM adopt a cellular state closer to that of PDAC cells present in tumors compared to standard culture models. Further, using the TIFM model, we found arginine biosynthesis is active in PDAC and allows PDAC cells to maintain levels of this amino acid despite microenvironmental arginine depletion. We also show that myeloid derived arginase activity is largely responsible for the low levels of arginine in PDAC tumors. Altogether, these data indicate that nutrient availability in tumors is an important determinant of cancer cell metabolism and behavior, and cell culture models that incorporate physiological nutrient availability have improved fidelity to in vivo systems and enable the discovery of novel cancer metabolic phenotypes.
Collapse
Affiliation(s)
- Juan J Apiz Saab
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | | | - Patrick B Jonker
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | - Roya AminiTabrizi
- Metabolomics Platform, Comprehensive Cancer Center, University of ChicagoChicagoUnited States
| | - Hardik Shah
- Metabolomics Platform, Comprehensive Cancer Center, University of ChicagoChicagoUnited States
| | - Rosa Elena Menjivar
- Cellular and Molecular Biology Program, University of Michigan-Ann ArborAnn ArborUnited States
| | - Andrew J Scott
- Department of Radiation Oncology, University of MichiganAnn ArborUnited States
| | - Zeribe C Nwosu
- Department of Molecular and Integrative Physiology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Zhou Zhu
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | - Riona N Chen
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | - Moses Oh
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | - Colin Sheehan
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | - Daniel R Wahl
- Department of Radiation Oncology, University of MichiganAnn ArborUnited States
| | | | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Kay F Macleod
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| | | | - Alexander Muir
- Ben May Department for Cancer Research, University of ChicagoChicagoUnited States
| |
Collapse
|
5
|
Yuan M, Tu B, Li H, Pang H, Zhang N, Fan M, Bai J, Wang W, Shu Z, DuFort CC, Huo S, Zhai J, Yao K, Wang L, Ying H, Zhu WG, Fu D, Hu Z, Zhao Y. Cancer-associated fibroblasts employ NUFIP1-dependent autophagy to secrete nucleosides and support pancreatic tumor growth. NATURE CANCER 2022; 3:945-960. [PMID: 35982178 DOI: 10.1038/s43018-022-00426-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are one of the most prominent and active components in the pancreatic tumor microenvironment. Our data show that CAFs are critical for survival from pancreatic ductal adenocarcinoma (PDAC) on glutamine deprivation. Specifically, we uncovered a role for nucleosides, which are secreted by CAFs through autophagy in a nuclear fragile X mental retardation-interacting protein 1 (NUFIP1)-dependent manner, increased glucose utilization and promoted growth of PDAC. Moreover, we demonstrate that CAF-derived nucleosides induced glucose consumption under glutamine-deprived conditions and displayed a dependence on MYC. Using an orthotopic mouse model of PDAC, we found that inhibiting nucleoside secretion by targeting NUFIP1 in the stroma reduced tumor weight. This finding highlights a previously unappreciated metabolic network within pancreatic tumors in which diverse nutrients are used to promote growth in an austere tumor microenvironment.
Collapse
Affiliation(s)
- Meng Yuan
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University School of Medicine, Shenzhen, China
| | - Bo Tu
- Molecular and Cellular Oncology Department, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hengchao Li
- Department of Pancreatic Surgery, Huashan hospital, Institute of Pancreatic Disease, FuDan University, Shanghai, China
| | - Huanhuan Pang
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Nan Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Minghe Fan
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jingru Bai
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology, Beijing, China
| | - Zhaoqi Shu
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Christopher C DuFort
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sihan Huo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jie Zhai
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ke Yao
- Department of Pancreatic Surgery, Huashan hospital, Institute of Pancreatic Disease, FuDan University, Shanghai, China
| | - Lina Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Haoqiang Ying
- Molecular and Cellular Oncology Department, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei-Guo Zhu
- Marshall Laboratory of Biomedical Engineering, Shenzhen University School of Medicine, Shenzhen, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan hospital, Institute of Pancreatic Disease, FuDan University, Shanghai, China.
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| | - Ying Zhao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
6
|
Ternes D, Tsenkova M, Pozdeev VI, Meyers M, Koncina E, Atatri S, Schmitz M, Karta J, Schmoetten M, Heinken A, Rodriguez F, Delbrouck C, Gaigneaux A, Ginolhac A, Nguyen TTD, Grandmougin L, Frachet-Bour A, Martin-Gallausiaux C, Pacheco M, Neuberger-Castillo L, Miranda P, Zuegel N, Ferrand JY, Gantenbein M, Sauter T, Slade DJ, Thiele I, Meiser J, Haan S, Wilmes P, Letellier E. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat Metab 2022; 4:458-475. [PMID: 35437333 PMCID: PMC9046088 DOI: 10.1038/s42255-022-00558-0] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 02/25/2022] [Indexed: 02/07/2023]
Abstract
The gut microbiome is a key player in the immunomodulatory and protumorigenic microenvironment during colorectal cancer (CRC), as different gut-derived bacteria can induce tumour growth. However, the crosstalk between the gut microbiome and the host in relation to tumour cell metabolism remains largely unexplored. Here we show that formate, a metabolite produced by the CRC-associated bacterium Fusobacterium nucleatum, promotes CRC development. We describe molecular signatures linking CRC phenotypes with Fusobacterium abundance. Cocultures of F. nucleatum with patient-derived CRC cells display protumorigenic effects, along with a metabolic shift towards increased formate secretion and cancer glutamine metabolism. We further show that microbiome-derived formate drives CRC tumour invasion by triggering AhR signalling, while increasing cancer stemness. Finally, F. nucleatum or formate treatment in mice leads to increased tumour incidence or size, and Th17 cell expansion, which can favour proinflammatory profiles. Moving beyond observational studies, we identify formate as a gut-derived oncometabolite that is relevant for CRC progression.
Collapse
Affiliation(s)
- Dominik Ternes
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Mina Tsenkova
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Vitaly Igorevich Pozdeev
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Marianne Meyers
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Sura Atatri
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Martine Schmitz
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Jessica Karta
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Maryse Schmoetten
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Almut Heinken
- School of Medicine, National University of Ireland, Galway, Ireland
- Ryan Institute, National University of Galway, Galway, Ireland
| | - Fabien Rodriguez
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Catherine Delbrouck
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anthoula Gaigneaux
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Aurelien Ginolhac
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Tam Thuy Dan Nguyen
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Lea Grandmougin
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Audrey Frachet-Bour
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Camille Martin-Gallausiaux
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Maria Pacheco
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | | | - Paulo Miranda
- National Center of Pathology, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Nikolaus Zuegel
- Department of Surgery, Centre Hospitalier Emile Mayrisch, Esch-sur-Alzette, Luxembourg
| | - Jean-Yves Ferrand
- Clinical and Epidemiological Investigation Center, Department of Population Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Manon Gantenbein
- Clinical and Epidemiological Investigation Center, Department of Population Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Thomas Sauter
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Daniel Joseph Slade
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Ines Thiele
- School of Medicine, National University of Ireland, Galway, Ireland
- Ryan Institute, National University of Galway, Galway, Ireland
- Discipline of Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
- APC Microbiome, Cork, Ireland
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Serge Haan
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
7
|
Iitani K, Ramamurthy SS, Ge X, Rao G. Transdermal sensing: in-situ non-invasive techniques for monitoring of human biochemical status. Curr Opin Biotechnol 2021; 71:198-205. [PMID: 34455345 DOI: 10.1016/j.copbio.2021.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Improving life expectancy necessitates prevention and early diagnosis of any disease state based on active self-monitoring of symptoms and longitudinal biochemical profiling. Non-invasive and continuous measurement of molecular biomarkers that reflect metabolism and health must however be established to realize this plan. Human samples non-invasively obtained via the skin are suitable in this context for in-situ biochemical monitoring. We present a brief classification of transdermal sampling in aqueous and gaseous phases and then introduce a new generation of transdermal monitoring devices for rapid and accurate assessment of important parameters. Finally, we have summarized the diversity of body-wide skin characteristics that have possible effects for transdermal sampling. Because of its passive nature, in-situ biochemical monitoring via transdermal sampling will potentially lead to a greater understanding of important biochemical markers and their temporal variation.
Collapse
Affiliation(s)
- Kenta Iitani
- Center for Advanced Sensor Technology (CAST), Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD, 21250 USA; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Sai Sathish Ramamurthy
- Center for Advanced Sensor Technology (CAST), Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD, 21250 USA; STAR Laboratory, Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Puttaparthi, Anantapur, Andhra Pradesh 515134, India
| | - Xudong Ge
- Center for Advanced Sensor Technology (CAST), Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD, 21250 USA
| | - Govind Rao
- Center for Advanced Sensor Technology (CAST), Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County (UMBC), 1000 Hilltop Circle, Baltimore, MD, 21250 USA.
| |
Collapse
|