1
|
Chowdhury NU, Cephus JY, Henriquez Pilier E, Wolf MM, Madden MZ, Kuehnle SN, McKernan KE, Jennings EQ, Arner EN, Heintzman DR, Chi C, Sugiura A, Stier MT, Voss K, Ye X, Scales K, Krystofiak ES, Gandhi VD, Guzy RD, Cahill KN, Sperling AI, Peebles RS, Rathmell JC, Newcomb DC. Androgen signaling restricts glutaminolysis to drive sex-specific Th17 metabolism in allergic airway inflammation. J Clin Invest 2024; 134:e177242. [PMID: 39404231 PMCID: PMC11601904 DOI: 10.1172/jci177242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/25/2024] [Indexed: 11/29/2024] Open
Abstract
Females have an increased prevalence of many Th17 cell-mediated diseases, including asthma. Androgen signaling decreases Th17 cell-mediated airway inflammation, and Th17 cells rely on glutaminolysis. However, it remains unclear whether androgen receptor (AR) signaling modifies glutamine metabolism to suppress Th17 cell-mediated airway inflammation. We show that Th17 cells from male humans and mice had decreased glutaminolysis compared to females, and that AR signaling attenuated Th17 cell mitochondrial respiration and glutaminolysis in mice. Using allergen-induced airway inflammation mouse models, we determined females had a selective reliance upon glutaminolysis for Th17-mediated airway inflammation, and AR signaling attenuated glutamine uptake in CD4+ T cells by reducing expression of glutamine transporters. Minimal reliance on glutamine uptake in male Th17 cells compared to female Th17 cells was also found in circulating T cells from patients with asthma. AR signaling thus attenuates glutaminolysis, demonstrating sex-specific metabolic regulation of Th17 cells with implications for Th17 or glutaminolysis targeted therapeutics.
Collapse
Affiliation(s)
- Nowrin U. Chowdhury
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | | | - Emely Henriquez Pilier
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Melissa M. Wolf
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Shelby N. Kuehnle
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kaitlin E. McKernan
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Erin Q. Jennings
- Vanderbilt Center for Immunobiology, and
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Emily N. Arner
- Vanderbilt Center for Immunobiology, and
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Darren R. Heintzman
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Channing Chi
- Department of Pathology, Microbiology, and Immunology
| | - Ayaka Sugiura
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Matthew T. Stier
- Vanderbilt Center for Immunobiology, and
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelsey Voss
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Xiang Ye
- Department of Pathology, Microbiology, and Immunology
| | - Kennedi Scales
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Evan S. Krystofiak
- Department of Cellular and Molecular Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Vivek D. Gandhi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robert D. Guzy
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Katherine N. Cahill
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anne I. Sperling
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - R. Stokes Peebles
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
| | - Dawn C. Newcomb
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, and
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
2
|
Liu W, Zhou H, Lai W, Hu C, Xu R, Gu P, Luo M, Zhang R, Li G. The immunosuppressive landscape in tumor microenvironment. Immunol Res 2024; 72:566-582. [PMID: 38691319 DOI: 10.1007/s12026-024-09483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.
Collapse
Affiliation(s)
- Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Peng Gu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Menglin Luo
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| |
Collapse
|
3
|
Nicolini A, Ferrari P. Involvement of tumor immune microenvironment metabolic reprogramming in colorectal cancer progression, immune escape, and response to immunotherapy. Front Immunol 2024; 15:1353787. [PMID: 39119332 PMCID: PMC11306065 DOI: 10.3389/fimmu.2024.1353787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/04/2024] [Indexed: 08/10/2024] Open
Abstract
Metabolic reprogramming is a k`ey hallmark of tumors, developed in response to hypoxia and nutrient deficiency during tumor progression. In both cancer and immune cells, there is a metabolic shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, also known as the Warburg effect, which then leads to lactate acidification, increased lipid synthesis, and glutaminolysis. This reprogramming facilitates tumor immune evasion and, within the tumor microenvironment (TME), cancer and immune cells collaborate to create a suppressive tumor immune microenvironment (TIME). The growing interest in the metabolic reprogramming of the TME, particularly its significance in colorectal cancer (CRC)-one of the most prevalent cancers-has prompted us to explore this topic. CRC exhibits abnormal glycolysis, glutaminolysis, and increased lipid synthesis. Acidosis in CRC cells hampers the activity of anti-tumor immune cells and inhibits the phagocytosis of tumor-associated macrophages (TAMs), while nutrient deficiency promotes the development of regulatory T cells (Tregs) and M2-like macrophages. In CRC cells, activation of G-protein coupled receptor 81 (GPR81) signaling leads to overexpression of programmed death-ligand 1 (PD-L1) and reduces the antigen presentation capability of dendritic cells. Moreover, the genetic and epigenetic cell phenotype, along with the microbiota, significantly influence CRC metabolic reprogramming. Activating RAS mutations and overexpression of epidermal growth factor receptor (EGFR) occur in approximately 50% and 80% of patients, respectively, stimulating glycolysis and increasing levels of hypoxia-inducible factor 1 alpha (HIF-1α) and MYC proteins. Certain bacteria produce short-chain fatty acids (SCFAs), which activate CD8+ cells and genes involved in antigen processing and presentation, while other mechanisms support pro-tumor activities. The use of immune checkpoint inhibitors (ICIs) in selected CRC patients has shown promise, and the combination of these with drugs that inhibit aerobic glycolysis is currently being intensively researched to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Paola Ferrari
- Unit of Oncology, Department of Medical and Oncological Area, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| |
Collapse
|
4
|
Lara O, Janssen P, Mambretti M, De Pauw L, Ates G, Mackens L, De Munck J, Walckiers J, Pan Z, Beckers P, Espinet E, Sato H, De Ridder M, Marks DL, Barbé K, Aerts JL, Hermans E, Rooman I, Massie A. Compartmentalized role of xCT in supporting pancreatic tumor growth, inflammation and mood disturbance in mice. Brain Behav Immun 2024; 118:275-286. [PMID: 38447884 DOI: 10.1016/j.bbi.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/05/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024] Open
Abstract
xCT (Slc7a11), the specific subunit of the cystine/glutamate antiporter system xc-, is present in the brain and on immune cells, where it is known to modulate behavior and inflammatory responses. In a variety of cancers -including pancreatic ductal adenocarcinoma (PDAC)-, xCT is upregulated by tumor cells to support their growth and spread. Therefore, we studied the impact of xCT deletion in pancreatic tumor cells (Panc02) and/or the host (xCT-/- mice) on tumor burden, inflammation, cachexia and mood disturbances. Deletion of xCT in the tumor strongly reduced tumor growth. Targeting xCT in the host and not the tumor resulted only in a partial reduction of tumor burden, while it did attenuate tumor-related systemic inflammation and prevented an increase in immunosuppressive regulatory T cells. The latter effect could be replicated by specific xCT deletion in immune cells. xCT deletion in the host or the tumor differentially modulated neuroinflammation. When mice were grafted with xCT-deleted tumor cells, hypothalamic inflammation was reduced and, accordingly, food intake improved. Tumor bearing xCT-/- mice showed a trend of reduced hippocampal neuroinflammation with less anxiety- and depressive-like behavior. Taken together, targeting xCT may have beneficial effects on pancreatic cancer-related comorbidities, beyond reducing tumor burden. The search for novel and specific xCT inhibitors is warranted as they may represent a holistic therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Olaya Lara
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Janssen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Marco Mambretti
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Gamze Ates
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Liselotte Mackens
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jolien De Munck
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jarne Walckiers
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Zhaolong Pan
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Beckers
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Elisa Espinet
- Pancreatic Cancer Lab, Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain; Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata 950-3198, Japan
| | - Mark De Ridder
- Department of Radiotherapy, UZ Brussels, VUB, Brussels 1090, Belgium
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kurt Barbé
- The Biostatistics and Medical Informatics Department, VUB, Brussels 1090, Belgium
| | - Joeri L Aerts
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Ilse Rooman
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium.
| | - Ann Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium.
| |
Collapse
|
5
|
Wang B, Pei J, Xu S, Liu J, Yu J. A glutamine tug-of-war between cancer and immune cells: recent advances in unraveling the ongoing battle. J Exp Clin Cancer Res 2024; 43:74. [PMID: 38459595 PMCID: PMC10921613 DOI: 10.1186/s13046-024-02994-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024] Open
Abstract
Glutamine metabolism plays a pivotal role in cancer progression, immune cell function, and the modulation of the tumor microenvironment. Dysregulated glutamine metabolism has been implicated in cancer development and immune responses, supported by mounting evidence. Cancer cells heavily rely on glutamine as a critical nutrient for survival and proliferation, while immune cells require glutamine for activation and proliferation during immune reactions. This metabolic competition creates a dynamic tug-of-war between cancer and immune cells. Targeting glutamine transporters and downstream enzymes involved in glutamine metabolism holds significant promise in enhancing anti-tumor immunity. A comprehensive understanding of the intricate molecular mechanisms underlying this interplay is crucial for developing innovative therapeutic approaches that improve anti-tumor immunity and patient outcomes. In this review, we provide a comprehensive overview of recent advances in unraveling the tug-of-war of glutamine metabolism between cancer and immune cells and explore potential applications of basic science discoveries in the clinical setting. Further investigations into the regulation of glutamine metabolism in cancer and immune cells are expected to yield valuable insights, paving the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Bolin Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
6
|
Shao L, Yang M, Sun T, Xia H, Du D, Li X, Jie Z. Role of solute carrier transporters in regulating dendritic cell maturation and function. Eur J Immunol 2024; 54:e2350385. [PMID: 38073515 DOI: 10.1002/eji.202350385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 02/27/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that initiate and regulate innate and adaptive immune responses. Solute carrier (SLC) transporters mediate diverse physiological functions and maintain cellular metabolite homeostasis. Recent studies have highlighted the significance of SLCs in immune processes. Notably, upon activation, immune cells undergo rapid and robust metabolic reprogramming, largely dependent on SLCs to modulate diverse immunological responses. In this review, we explore the central roles of SLC proteins and their transported substrates in shaping DC functions. We provide a comprehensive overview of recent studies on amino acid transporters, metal ion transporters, and glucose transporters, emphasizing their essential contributions to DC homeostasis under varying pathological conditions. Finally, we propose potential strategies for targeting SLCs in DCs to bolster immunotherapy for a spectrum of human diseases.
Collapse
Affiliation(s)
- Lin Shao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- School of Life Sciences, Fudan University, Shanghai, China
| | - Mengxin Yang
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Tao Sun
- Department of Laboratory Medicine, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haotang Xia
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Dan Du
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xun Li
- Department of Laboratory Medicine, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuliang Jie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
7
|
Hua Y, Yang S, Zhang Y, Li J, Wang M, Yeerkenbieke P, Liao Q, Liu Q. Modulating ferroptosis sensitivity: environmental and cellular targets within the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:19. [PMID: 38217037 PMCID: PMC10787430 DOI: 10.1186/s13046-023-02925-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/06/2023] [Indexed: 01/14/2024] Open
Abstract
Ferroptosis, a novel form of cell death triggered by iron-dependent phospholipid peroxidation, presents significant therapeutic potential across diverse cancer types. Central to cellular metabolism, the metabolic pathways associated with ferroptosis are discernible in both cancerous and immune cells. This review begins by delving into the intricate reciprocal regulation of ferroptosis between cancer and immune cells. It subsequently details how factors within the tumor microenvironment (TME) such as nutrient scarcity, hypoxia, and cellular density modulate ferroptosis sensitivity. We conclude by offering a comprehensive examination of distinct immunophenotypes and environmental and metabolic targets geared towards enhancing ferroptosis responsiveness within the TME. In sum, tailoring precise ferroptosis interventions and combination strategies to suit the unique TME of specific cancers may herald improved patient outcomes.
Collapse
Affiliation(s)
- Yuze Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yalu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Anhui Provincial Hospital, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230027, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Palashate Yeerkenbieke
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Xinjiang Yili Kazak Autonomous Prefecture Friendship Hospital, Xinjiang, 835099, China
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
8
|
Bacigalupa ZA, Landis MD, Rathmell JC. Nutrient inputs and social metabolic control of T cell fate. Cell Metab 2024; 36:10-20. [PMID: 38118440 PMCID: PMC10872404 DOI: 10.1016/j.cmet.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/25/2023] [Accepted: 12/05/2023] [Indexed: 12/22/2023]
Abstract
Cells in multicellular organisms experience diverse neighbors, signals, and evolving physical environments that drive functional and metabolic demands. To maintain proper development and homeostasis while avoiding inappropriate cell proliferation or death, individual cells interact with their neighbors via "social" cues to share and partition available nutrients. Metabolic signals also contribute to cell fate by providing biochemical links between cell-extrinsic signals and available resources. In addition to metabolic checkpoints that sense nutrients and directly supply molecular intermediates for biosynthetic pathways, many metabolites directly signal or provide the basis for post-translational modifications of target proteins and chromatin. In this review, we survey the landscape of T cell nutrient sensing and metabolic signaling that supports proper immunity while avoiding immunodeficiency or autoimmunity. The integration of cell-extrinsic microenvironmental cues with cell-intrinsic metabolic signaling provides a social metabolic control model to integrate cell signaling, metabolism, and fate.
Collapse
Affiliation(s)
- Zachary A Bacigalupa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madelyn D Landis
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
9
|
Vizcarra EA, Ulu A, Landrith TA, Qiu X, Godzik A, Wilson EH. Group 1 metabotropic glutamate receptor expression defines a T cell memory population during chronic Toxoplasma infection that enhances IFN-gamma and perforin production in the CNS. Brain Behav Immun 2023; 114:131-143. [PMID: 37604212 DOI: 10.1016/j.bbi.2023.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023] Open
Abstract
Within the brain, a pro-inflammatory response is essential to prevent clinical disease due to Toxoplasma gondii reactivation. Infection in the immunocompromised leads to lethal Toxoplasmic encephalitis while in the immunocompetent, there is persistent low-grade inflammation which is devoid of clinical symptoms. This signifies that there is a well-balanced and regulated inflammatory response to T. gondii in the brain. T cells are the dominant immune cells that prevent clinical disease, and this is mediated through the secretion of effector molecules such as perforins and IFN-γ. The presence of cognate antigen, the expression of survival cytokines, and the alteration of the epigenetic landscape drive the development of memory T cells. However, specific extrinsic signals that promote the formation and maintenance of memory T cells within tissue are poorly understood. During chronic infection, there is an increase in extracellular glutamate that, due to its function as an excitatory neurotransmitter, is normally tightly controlled in the CNS. Here we demonstrate that CD8+ T cells from the T. gondii-infected brain parenchyma are enriched for metabotropic glutamate receptors (mGluR's). Characterization studies determined that mGluR+ expression by CD8+ T cells defines a distinct memory population at the transcriptional and protein level. Finally, using receptor antagonists and agonists we demonstrate mGluR signaling is required for optimal CD8+ T cell production of the effector cytokine IFNγ. This work suggests that glutamate is an important environmental signal of inflammation that promotes T cell function. Understanding glutamate's influence on T cells in the brain can provide insights into the mechanisms that govern protective immunity against CNS-infiltrating pathogens and neuroinflammation.
Collapse
Affiliation(s)
- Edward A Vizcarra
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Tyler A Landrith
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States.
| |
Collapse
|
10
|
Vick LV, Canter RJ, Monjazeb AM, Murphy WJ. Multifaceted effects of obesity on cancer immunotherapies: Bridging preclinical models and clinical data. Semin Cancer Biol 2023; 95:88-102. [PMID: 37499846 PMCID: PMC10836337 DOI: 10.1016/j.semcancer.2023.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Obesity, defined by excessive body fat, is a highly complex condition affecting numerous physiological processes, such as metabolism, proliferation, and cellular homeostasis. These multifaceted effects impact cells and tissues throughout the host, including immune cells as well as cancer biology. Because of the multifaceted nature of obesity, common parameters used to define it (such as body mass index in humans) can be problematic, and more nuanced methods are needed to characterize the pleiotropic metabolic effects of obesity. Obesity is well-accepted as an overall negative prognostic factor for cancer incidence, progression, and outcome. This is in part due to the meta-inflammatory and immunosuppressive effects of obesity. Immunotherapy is increasingly used in cancer therapy, and there are many different types of immunotherapy approaches. The effects of obesity on immunotherapy have only recently been studied with the demonstration of an "obesity paradox", in which some immune therapies have been demonstrated to result in greater efficacy in obese subjects despite the direct adverse effects of obesity and excess body fat acting on the cancer itself. The multifactorial characteristics that influence the effects of obesity (age, sex, lean muscle mass, underlying metabolic conditions and drugs) further confound interpretation of clinical data and necessitate the use of more relevant preclinical models mirroring these variables in the human scenario. Such models will allow for more nuanced mechanistic assessment of how obesity can impact, both positively and negatively, cancer biology, host metabolism, immune regulation, and how these intersecting processes impact the delivery and outcome of cancer immunotherapy.
Collapse
Affiliation(s)
- Logan V Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA; Department of Internal Medicine, Division of Malignant Hematology, Cellular Therapy and Transplantation, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
11
|
Bertolini M, Wong MS, Mendive-Tapia L, Vendrell M. Smart probes for optical imaging of T cells and screening of anti-cancer immunotherapies. Chem Soc Rev 2023; 52:5352-5372. [PMID: 37376918 PMCID: PMC10424634 DOI: 10.1039/d2cs00928e] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 06/29/2023]
Abstract
T cells are an essential part of the immune system with crucial roles in adaptive response and the maintenance of tissue homeostasis. Depending on their microenvironment, T cells can be differentiated into multiple states with distinct functions. This myriad of cellular activities have prompted the development of numerous smart probes, ranging from small molecule fluorophores to nanoconstructs with variable molecular architectures and fluorescence emission mechanisms. In this Tutorial Review, we summarize recent efforts in the design, synthesis and application of smart probes for imaging T cells in tumors and inflammation sites by targeting metabolic and enzymatic biomarkers as well as specific surface receptors. Finally, we briefly review current strategies for how smart probes are employed to monitor the response of T cells to anti-cancer immunotherapies. We hope that this Review may help chemists, biologists and immunologists to design the next generation of molecular imaging probes for T cells and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Marco Bertolini
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Man Sing Wong
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| |
Collapse
|
12
|
Zhong Z, Zhang C, Ni S, Ma M, Zhang X, Sang W, Lv T, Qian Z, Yi C, Yu B. NFATc1-mediated expression of SLC7A11 drives sensitivity to TXNRD1 inhibitors in osteoclast precursors. Redox Biol 2023; 63:102711. [PMID: 37148740 PMCID: PMC10184050 DOI: 10.1016/j.redox.2023.102711] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023] Open
Abstract
Excess osteoclast activity is found in many bone metabolic diseases, and inhibiting osteoclast differentiation has proven to be an effective strategy. Here, we revealed that osteoclast precursors (pre-OCs) were more susceptible to thioredoxin reductase 1 (TXNRD1) inhibitors than bone marrow-derived monocytes (BMDMs) during receptor activator of nuclear factor kappa B ligand (RANKL)-mediated osteoclastogenesis. Mechanistically, we found that nuclear factor of activated T-cells 1 (NFATc1) upregulated solute carrier family 7 member 11 (SLC7A11) expression through transcriptional regulation during RANKL-induced osteoclastogenesis. During TXNRD1 inhibition, the rate of intracellular disulfide reduction is significantly reduced. Increased cystine transport leads to increased cystine accumulation, which leads to increased cellular disulfide stress and disulfidptosis. We further demonstrated that SLC7A11 inhibitors and treatments that prevent disulphide accumulation could rescue this type of cell death, but not the ferroptosis inhibitors (DFO, Ferro-1), the ROS scavengers (Trolox, Tempol), the apoptosis inhibitor (Z-VAD), the necroptosis inhibitor (Nec-1), or the autophagy inhibitor (CQ). An in vivo study indicated that TXNRD1 inhibitors increased bone cystine content, reduced the number of osteoclasts, and alleviated bone loss in an ovariectomized (OVX) mouse model. Together, our findings demonstrate that NFATc1-mediated upregulation of SLC7A11 induces targetable metabolic sensitivity to TXNRD1 inhibitors during osteoclast differentiation. Moreover, we innovatively suggest that TXNRD1 inhibitors, a classic drug for osteoclast-related diseases, selectively kill pre-OCs by inducing intracellular cystine accumulation and subsequent disulfidptosis.
Collapse
Affiliation(s)
- Zeyuan Zhong
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Shanghai Medical College, Fudan University, Shanghai, China
| | - Chongjing Zhang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuo Ni
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Miao Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaomeng Zhang
- Renal Medicine and Baxter Novum, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Weicong Sang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Lv
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhi Qian
- Institution of Orthopaedic Diseases, Zhangye People's Hospital Affiliated to Hexi University, Zhangye, China.
| | - Chengqing Yi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| | - Baoqing Yu
- Department of Orthopedics, Shanghai Pudong New Area People's Hospital, Shanghai, China.
| |
Collapse
|
13
|
Yamamoto T, Arita M, Tamura T, Saito M, Katayama H, Kuroda H, Suzuki T, Kawamata S. A Novel Approach for Determining the Critical Quality Attributes of Mesenchymal Stem Cells by Specifying Cell Population With Replication Potential. Stem Cells Transl Med 2023; 12:169-182. [PMID: 36917628 PMCID: PMC10021496 DOI: 10.1093/stcltm/szad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/19/2023] [Indexed: 03/16/2023] Open
Abstract
We introduce a novel approach to determine the critical quality attributes (CQAs) of mesenchymal stem cells (MSCs) expected to exert immunosuppressive effects. MSCs retained homeostatic replication potentials, such as sustainable growth and consistent cell morphology as a population, in early passages, but lost them in late passages. Characteristic surface markers of MSCs (ie, CD73, CD90, and CD105) were no longer expressed at 2 weeks after subcutaneous transplantation into NOG mice when MSCs from late passages were transplanted, but not when MSCs from early passages were transplanted, suggesting that the biological effects of the MSCs differed according to the timing of cell harvesting and highlighting the importance of specifying MSCs that retained homeostatic features to define the CQAs. The homeostatic features of MSCs related to the balance of the redox system, nutrient requirements, and mitochondrial function were also observed until a certain passage. Therefore, we could define the CQAs of MSCs related to manufacturing by selecting process parameters (PPs) underlying the homeostatic features of MSCs and measuring these PPs quantitatively to specify the cell population with homeostatic features by limiting the passage number. The validity of the PPs stipulated in our pilot study was verified using an SKG murine arthritis model, and critical PPs (CPPs) were then selected among the PPs. Thus, CQAs related to manufacturing in the developmental phase could be defined by the CPPs in this manner, and the concept of CQAs could be refined continuously toward commercial manufacturing.
Collapse
Affiliation(s)
- Takako Yamamoto
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Mao Arita
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Takashi Tamura
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Miho Saito
- R&D Center for Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | | | - Hirotaka Kuroda
- Shimadzu Corp., Kyoto, Japan
- Graduate School of Engineering, Osaka University, Osaka, Japan
| | | | - Shin Kawamata
- Corresponding author: Shin Kawamata, 1-5-4, Minatojima Minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| |
Collapse
|
14
|
Valvo V, Parietti E, Deans K, Ahn SW, Park NR, Ferland B, Thompson D, Dominas C, Bhagavatula SK, Davidson S, Jonas O. High-throughput in situ perturbation of metabolite levels in the tumor micro-environment reveals favorable metabolic condition for increased fitness of infiltrated T-cells. Front Cell Dev Biol 2022; 10:1032360. [PMID: 36619865 PMCID: PMC9815512 DOI: 10.3389/fcell.2022.1032360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor-infiltrating immune cells experience significant metabolic reprogramming in the tumor microenvironment (TME), and they share similar metabolic pathways and nutrient needs with malignant cells. This positions these cell types in direct nutrient competition in the TME. We currently lack a complete understanding of the similarities, differences, and functional consequences of the metabolic pathways utilized by activated immune cells from different lineages versus neoplastic cells. This study applies a novel in situ approach using implantable microdevices to expose the tumor to 27 controlled and localized metabolic perturbations in order to perform a systematic investigation into the metabolic regulation of the cellular fitness and persistence between immune and tumor cells directly within the native TME. Our findings identify the most potent metabolites, notably glutamine and arginine, that induce a favorable metabolic immune response in a mammary carcinoma model, and reveal novel insights on less characterized pathways, such as cysteine and glutathione. We then examine clinical samples from cancer patients to confirm the elevation of these pathways in tumor regions that are enriched in activated T cells. Overall, this work provides the first instance of a highly multiplexed in situ competition assay between malignant and immune cells within tumors using a range of localized microdose metabolic perturbations. The approach and findings may be used to potentiate the effects of T cell stimulating immunotherapies on a tumor-specific or personalized basis through targeted enrichment or depletion of specific metabolites.
Collapse
Affiliation(s)
- Veronica Valvo
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Parietti
- Department of Infectious Diseases and Hospital of Epidemiology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Kyle Deans
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Sebastian W. Ahn
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Noel Ruth Park
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Benjamin Ferland
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Devon Thompson
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Sharath K. Bhagavatula
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Shawn Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Oliver Jonas
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Cosovanu C, Resch P, Jordan S, Lehmann A, Ralser M, Farztdinov V, Spranger J, Mülleder M, Brachs S, Neumann C. Intestinal epithelial c-Maf expression determines enterocyte differentiation and nutrient uptake in mice. J Exp Med 2022; 219:213479. [PMID: 36121416 PMCID: PMC9486084 DOI: 10.1084/jem.20220233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/20/2022] [Accepted: 08/24/2022] [Indexed: 11/04/2022] Open
Abstract
The primary function of the small intestine (SI) is to absorb nutrients to maintain whole-body energy homeostasis. Enterocytes are the major epithelial cell type facilitating nutrient sensing and uptake. However, the molecular regulators governing enterocytes have remained undefined. Here, we identify c-Maf as an enterocyte-specific transcription factor within the SI epithelium. c-Maf expression was determined by opposing Noggin/BMP signals and overlapped with the zonated enrichment of nutrient transporters in the mid-villus region. Functionally, enterocytes required c-Maf to appropriately differentiate along the villus axis. Specifically, gene programs controlling carbohydrate and protein absorption were c-Maf-dependent. Consequently, epithelial cell-specific c-Maf deletion resulted in impaired enterocyte maturation and nutrient uptake, including defects in the adaptation to different nutrient availability. Concomitantly, intraepithelial lymphocytes were less abundant, while commensal epithelial cell-attaching SFB overgrew in a c-Maf-deficient environment, highlighting the close interdependence between the intestinal epithelium, immune system, and microbiota. Collectively, our data identified c-Maf as a key regulator of SI enterocyte differentiation and function, essential for nutrient, immune, and microbial homeostasis.
Collapse
Affiliation(s)
- Catalina Cosovanu
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Resch
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Jordan
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Lehmann
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK
| | - Vadim Farztdinov
- Core Facility - High-Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility - High-Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Christian Neumann
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
16
|
Reprogramming T-Cell Metabolism for Better Anti-Tumor Immunity. Cells 2022; 11:cells11193103. [PMID: 36231064 PMCID: PMC9562038 DOI: 10.3390/cells11193103] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
T cells play central roles in the anti-tumor immunity, whose activation and differentiation are profoundly regulated by intrinsic metabolic reprogramming. Emerging evidence has revealed that metabolic processes of T cells are generally altered by tumor cells or tumor released factors, leading to crippled anti-tumor immunity. Therefore, better understanding of T cell metabolic mechanism is crucial in developing the next generation of T cell-based anti-tumor immunotherapeutics. In this review, we discuss how metabolic pathways affect T cells to exert their anti-tumor effects and how to remodel the metabolic programs to improve T cell-mediated anti-tumor immune responses. We emphasize that glycolysis, carboxylic acid cycle, fatty acid oxidation, cholesterol metabolism, amino acid metabolism, and nucleotide metabolism work together to tune tumor-reactive T-cell activation and proliferation.
Collapse
|
17
|
Metabolic regulation and function of T helper cells in neuroinflammation. Semin Immunopathol 2022; 44:581-598. [PMID: 36068310 DOI: 10.1007/s00281-022-00959-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/19/2022] [Indexed: 12/12/2022]
Abstract
Neuroinflammatory conditions such as multiple sclerosis (MS) are initiated by pathogenic immune cells invading the central nervous system (CNS). Autoreactive CD4+ T helper cells are critical players that orchestrate the immune response both in MS and in other neuroinflammatory autoimmune diseases including animal models that have been developed for MS. T helper cells are classically categorized into different subsets, but heterogeneity exists within these subsets. Untangling the more complex regulation of these subsets will clarify their functional roles in neuroinflammation. Here, we will discuss how differentiation, immune checkpoint pathways, transcriptional regulation and metabolic factors determine the function of CD4+ T cell subsets in CNS autoimmunity. T cells rely on metabolic reprogramming for their activation and proliferation to meet bioenergetic demands. This includes changes in glycolysis, glutamine metabolism and polyamine metabolism. Importantly, these pathways were recently also implicated in the fine tuning of T cell fate decisions during neuroinflammation. A particular focus of this review will be on the Th17/Treg balance and intra-subset functional states that can either promote or dampen autoimmune responses in the CNS and thus affect disease outcome. An increased understanding of factors that could tip CD4+ T cell subsets and populations towards an anti-inflammatory phenotype will be critical to better understand neuroinflammatory diseases and pave the way for novel treatment paradigms.
Collapse
|
18
|
Purohit V, Wagner A, Yosef N, Kuchroo VK. Systems-based approaches to study immunometabolism. Cell Mol Immunol 2022; 19:409-420. [PMID: 35121805 PMCID: PMC8891302 DOI: 10.1038/s41423-021-00783-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Technical advances at the interface of biology and computation, such as single-cell RNA-sequencing (scRNA-seq), reveal new layers of complexity in cellular systems. An emerging area of investigation using the systems biology approach is the study of the metabolism of immune cells. The diverse spectra of immune cell phenotypes, sparsity of immune cell numbers in vivo, limitations in the number of metabolites identified, dynamic nature of cellular metabolism and metabolic fluxes, tissue specificity, and high dependence on the local milieu make investigations in immunometabolism challenging, especially at the single-cell level. In this review, we define the systemic nature of immunometabolism, summarize cell- and system-based approaches, and introduce mathematical modeling approaches for systems interrogation of metabolic changes in immune cells. We close the review by discussing the applications and shortcomings of metabolic modeling techniques. With systems-oriented studies of metabolism expected to become a mainstay of immunological research, an understanding of current approaches toward systems immunometabolism will help investigators make the best use of current resources and push the boundaries of the discipline.
Collapse
Affiliation(s)
- Vinee Purohit
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02141, USA
| | - Allon Wagner
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02141, USA.
| |
Collapse
|
19
|
Beckers P, Lara O, Belo do Nascimento I, Desmet N, Massie A, Hermans E. Validation of a System xc– Functional Assay in Cultured Astrocytes and Nervous Tissue Samples. Front Cell Neurosci 2022; 15:815771. [PMID: 35095428 PMCID: PMC8793334 DOI: 10.3389/fncel.2021.815771] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Disruption of the glutamatergic homeostasis is commonly observed in neurological diseases and has been frequently correlated with the altered expression and/or function of astrocytic high-affinity glutamate transporters. There is, however, a growing interest for the role of the cystine-glutamate exchanger system xc– in controlling glutamate transmission. This exchanger is predominantly expressed in glial cells, especially in microglia and astrocytes, and its dysregulation has been documented in diverse neurological conditions. While most studies have focused on measuring the expression of its specific subunit xCT by RT-qPCR or by Western blotting, the activity of this exchanger in tissue samples remains poorly examined. Indeed, the reported use of sulfur- and carbon-radiolabeled cystine in uptake assays shows several drawbacks related to its short radioactive half-life and its relatively high cost. We here report on the elaborate validation of a method using tritiated glutamate as a substrate for the reversed transport mediated by system xc–. The uptake assay was validated in primary cultured astrocytes, in transfected cells as well as in crude synaptosomes obtained from fresh nervous tissue samples. Working in buffers containing defined concentrations of Na+, allowed us to differentiate the glutamate uptake supported by system xc– or by high-affinity glutamate transporters, as confirmed by using selective pharmacological inhibitors. The specificity was further demonstrated in primary astrocyte cultures from transgenic mice lacking xCT or in cell lines where xCT expression was genetically induced or reduced. As such, this assay appears to be a robust and cost-efficient solution to investigate the activity of this exchanger in physiological and pathological conditions. It also provides a reliable tool for the screening and characterization of new system xc– inhibitors which have been frequently cited as valuable drugs for nervous disorders and cancer.
Collapse
Affiliation(s)
- Pauline Beckers
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Olaya Lara
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ines Belo do Nascimento
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie Desmet
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Ann Massie
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emmanuel Hermans
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Emmanuel Hermans,
| |
Collapse
|
20
|
Plasmacytoid dendritic cell activation is dependent on coordinated expression of distinct amino acid transporters. Immunity 2021; 54:2514-2530.e7. [PMID: 34717796 DOI: 10.1016/j.immuni.2021.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 07/01/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2023]
Abstract
Human plasmacytoid dendritic cells (pDCs) are interleukin-3 (IL-3)-dependent cells implicated in autoimmunity, but the role of IL-3 in pDC biology is poorly understood. We found that IL-3-induced Janus kinase 2-dependent expression of SLC7A5 and SLC3A2, which comprise the large neutral amino acid transporter, was required for mammalian target of rapamycin complex 1 (mTORC1) nutrient sensor activation in response to toll-like receptor agonists. mTORC1 facilitated increased anabolic activity resulting in type I interferon, tumor necrosis factor, and chemokine production and the expression of the cystine transporter SLC7A11. Loss of function of these amino acid transporters synergistically blocked cytokine production by pDCs. Comparison of in vitro-activated pDCs with those from lupus nephritis lesions identified not only SLC7A5, SLC3A2, and SLC7A11 but also ectonucleotide pyrophosphatase-phosphodiesterase 2 (ENPP2) as components of a shared transcriptional signature, and ENPP2 inhibition also blocked cytokine production. Our data identify additional therapeutic targets for autoimmune diseases in which pDCs are implicated.
Collapse
|
21
|
Hope HC, Salmond RJ. The Role of Non-essential Amino Acids in T Cell Function and Anti-tumour Immunity. Arch Immunol Ther Exp (Warsz) 2021; 69:29. [PMID: 34637000 PMCID: PMC8510955 DOI: 10.1007/s00005-021-00633-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/03/2021] [Indexed: 02/02/2023]
Abstract
T cell activation, differentiation and proliferation is dependent upon and intrinsically linked to a capacity to modulate and adapt cellular metabolism. Antigen-induced activation stimulates a transcriptional programme that results in metabolic reprogramming, enabling T cells to fuel anabolic metabolic pathways and provide the nutrients to sustain proliferation and effector responses. Amino acids are key nutrients for T cells and have essential roles as building blocks for protein synthesis as well as in numerous metabolic pathways. In this review, we discuss the roles for uptake and biosynthesis of non-essential amino acids in T cell metabolism, activation and effector function. Furthermore, we highlight the effects of amino acid metabolism and depletion by cancer cells on T cell anti-tumour function and discuss approaches to modulate and improve T cell metabolism for improved anti-tumour function in these nutrient-depleted microenvironments.
Collapse
Affiliation(s)
- Helen Carrasco Hope
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, 1066, Epalinges, Switzerland
| | - Robert J Salmond
- Leeds Institute of Medical Research at St. James's, University of Leeds, St. James University Hospital, Leeds, LS9 7TF, UK.
| |
Collapse
|
22
|
Voss K, Hong HS, Bader JE, Sugiura A, Lyssiotis CA, Rathmell JC. A guide to interrogating immunometabolism. Nat Rev Immunol 2021; 21:637-652. [PMID: 33859379 PMCID: PMC8478710 DOI: 10.1038/s41577-021-00529-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
The metabolic charts memorized in early biochemistry courses, and then later forgotten, have come back to haunt many immunologists with new recognition of the importance of these pathways. Metabolites and the activity of metabolic pathways drive energy production, macromolecule synthesis, intracellular signalling, post-translational modifications and cell survival. Immunologists who identify a metabolic phenotype in their system are often left wondering where to begin and what does it mean? Here, we provide a framework for navigating and selecting the appropriate biochemical techniques to explore immunometabolism. We offer recommendations for initial approaches to develop and test metabolic hypotheses and how to avoid common mistakes. We then discuss how to take things to the next level with metabolomic approaches, such as isotope tracing and genetic approaches. By proposing strategies and evaluating the strengths and weaknesses of different methodologies, we aim to provide insight, note important considerations and discuss ways to avoid common misconceptions. Furthermore, we highlight recent studies demonstrating the power of these metabolic approaches to uncover the role of metabolism in immunology. By following the framework in this Review, neophytes and seasoned investigators alike can venture into the emerging realm of cellular metabolism and immunity with confidence and rigour.
Collapse
Affiliation(s)
- Kelsey Voss
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hanna S Hong
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jackie E Bader
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ayaka Sugiura
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Costas A Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
23
|
Frades I, Foguet C, Cascante M, Araúzo-Bravo MJ. Genome Scale Modeling to Study the Metabolic Competition between Cells in the Tumor Microenvironment. Cancers (Basel) 2021; 13:4609. [PMID: 34572839 PMCID: PMC8470216 DOI: 10.3390/cancers13184609] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/31/2022] Open
Abstract
The tumor's physiology emerges from the dynamic interplay of numerous cell types, such as cancer cells, immune cells and stromal cells, within the tumor microenvironment. Immune and cancer cells compete for nutrients within the tumor microenvironment, leading to a metabolic battle between these cell populations. Tumor cells can reprogram their metabolism to meet the high demand of building blocks and ATP for proliferation, and to gain an advantage over the action of immune cells. The study of the metabolic reprogramming mechanisms underlying cancer requires the quantification of metabolic fluxes which can be estimated at the genome-scale with constraint-based or kinetic modeling. Constraint-based models use a set of linear constraints to simulate steady-state metabolic fluxes, whereas kinetic models can simulate both the transient behavior and steady-state values of cellular fluxes and concentrations. The integration of cell- or tissue-specific data enables the construction of context-specific models that reflect cell-type- or tissue-specific metabolic properties. While the available modeling frameworks enable limited modeling of the metabolic crosstalk between tumor and immune cells in the tumor stroma, future developments will likely involve new hybrid kinetic/stoichiometric formulations.
Collapse
Affiliation(s)
- Itziar Frades
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, 20009 San Sebastian, Spain;
| | - Carles Foguet
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of University of Barcelona, Faculty of Biology, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain; (C.F.); (M.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) (CB17/04/00023) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Instituto de Salud Carlos III (ISCIII), 28020 Madrid, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of University of Barcelona, Faculty of Biology, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain; (C.F.); (M.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) (CB17/04/00023) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Instituto de Salud Carlos III (ISCIII), 28020 Madrid, Spain
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, 20009 San Sebastian, Spain;
- Max Planck Institute of Molecular Biomedicine, 48167 Münster, Germany
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERfes), 28015 Madrid, Spain
- Translational Bioinformatics Network (TransBioNet), 8001 Barcelona, Spain
- Ikerbasque, Basque Foundation for Science, 48012 Bilbao, Spain
| |
Collapse
|
24
|
Nguyen DC, Duan M, Ali M, Ley A, Sanz I, Lee FEH. Plasma cell survival: The intrinsic drivers, migratory signals, and extrinsic regulators. Immunol Rev 2021; 303:138-153. [PMID: 34337772 PMCID: PMC8387437 DOI: 10.1111/imr.13013] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022]
Abstract
Antibody-secreting cells (ASC) are the effectors of protective humoral immunity and the only cell type that produces antibodies or immunoglobulins in mammals. In addition to their formidable capacity to secrete massive quantities of proteins, ASC are terminally differentiated and have unique features to become long-lived plasma cells (LLPC). Upon antigen encounter, B cells are activated through a complex multistep process to undergo fundamental morphological, subcellular, and molecular transformation to become an efficient protein factory with lifelong potential. The ASC survival potential is determined by factors at the time of induction, capacity to migration from induction to survival sites, and ability to mature in the specialized bone marrow microenvironments. In the past decade, considerable progress has been made in identifying factors regulating ASC longevity. Here, we review the intrinsic drivers, trafficking signals, and extrinsic regulators with particular focus on how they impact the survival potential to become a LLPC.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Meixue Duan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Mohammad Ali
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ariel Ley
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, United States
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States
| | - F. Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, United States
| |
Collapse
|
25
|
Yu T, Dong T, Eyvani H, Fang Y, Wang X, Zhang X, Lu X. Metabolic interventions: A new insight into the cancer immunotherapy. Arch Biochem Biophys 2021; 697:108659. [PMID: 33144083 PMCID: PMC8638212 DOI: 10.1016/j.abb.2020.108659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/15/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
Metabolic reprogramming confers cancer cells plasticity and viability under harsh conditions. Such active alterations lead to cell metabolic dependency, which can be exploited as an attractive target in development of effective antitumor therapies. Similar to cancer cells, activated T cells also execute global metabolic reprogramming for their proliferation and effector functions when recruited to the tumor microenvironment (TME). However, the high metabolic activity of rapidly proliferating cancer cells can compete for nutrients with immune cells in the TME, and consequently, suppressing their anti-tumor functions. Thus, therapeutic strategies could aim to restore T cell metabolism and anti-tumor responses in the TME by targeting the metabolic dependence of cancer cells. In this review, we highlight current research progress on metabolic reprogramming and the interplay between cancer cells and immune cells. We also discuss potential therapeutic intervention strategies for targeting metabolic pathways to improve cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Tao Yu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tianhan Dong
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Haniyeh Eyvani
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yuanzhang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiyu Wang
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xinna Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Barili V, Boni C, Rossi M, Vecchi A, Zecca A, Penna A, Missale G, Ferrari C, Fisicaro P. Metabolic regulation of the HBV-specific T cell function. Antiviral Res 2020; 185:104989. [PMID: 33248194 DOI: 10.1016/j.antiviral.2020.104989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
Chronically HBV infected subjects are more than 260 million worldwide; cirrhosis and liver cancer represent possible outcomes which affect around 700,000 patients per year. Both innate and adaptive immune responses are necessary for viral control and both have been shown to be defective in chronic patients. Metabolic remodeling is an essential process in T cell biology, particularly for T cell activation, differentiation and survival. Cellular metabolism relies on the conversion of nutrients into energy to support intracellular processes, and to generate fundamental intermediate components for cell proliferation and growth. Adaptive immune responses are the central mechanisms for the resolution of primary human infections leading to the activation of pathogen-specific B and T cell functions. In chronic HBV infection the anti-viral immune response fails to contain the virus and leads to persistent hepatic tissue damage which may finally result in liver cirrhosis and cancer. This T cell failure is associated with metabolic alterations suggesting that control of nutrient uptake and intracellular utilization as well as correct regulation of intracellular metabolic pathways are strategic for T cell differentiation during persistent chronic infections. This review will discuss some of the main features of the T cell metabolic processes which are relevant to the generation of an efficient antiviral response, with specific focus on their clinical relevance in chronic HBV infection in the perspective of possible strategies to correct deregulated metabolic pathways underlying T cell dysfunction of chronic HBV patients.
Collapse
Affiliation(s)
- Valeria Barili
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Carolina Boni
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marzia Rossi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Andrea Vecchi
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Alessandra Zecca
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Amalia Penna
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Gabriele Missale
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Carlo Ferrari
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.
| | - Paola Fisicaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
27
|
Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, Birnbaumer L, Yang Y. Reinvigorating exhausted CD8 + cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev 2020; 41:156-201. [PMID: 32844499 DOI: 10.1002/med.21727] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has revolutionized the treatment of cancer in recent years and achieved overall success and long-term clinical benefit in patients with a wide variety of cancer types. However, there is still a large proportion of patients exhibiting limited or no responses to immunotherapeutic strategy, some of which were even observed with hyperprogressive disease. One major obstacle restricting the efficacy is that tumor-reactive CD8+ T cells, which are central for tumor control, undergo exhaustion, and lose their ability to eliminate cancer cells after infiltrating into the strongly immunosuppressive tumor microenvironment. Thus, as a potential therapeutic rationale in the development of cancer immunotherapy, targeting or reinvigorating exhausted CD8+ T cells has been attracting much interest. Hitherto, both intrinsic and extrinsic mechanisms that govern CD8+ T-cell exhaustion have been explored. Specifically, the transcriptional and epigenetic landscapes have been depicted utilizing single-cell RNA sequencing or mass cytometry (CyTOF). In addition, cellular metabolism dictating the tumor-infiltrating CD8+ T-cell fate is currently under investigation. A series of clinical trials are being carried out to further establish the current strategies targeting CD8+ T-cell exhaustion. Taken together, despite the proven benefit of immunotherapy in cancer patients, additional efforts are still needed to fully circumvent limitations of exhausted T cells in the treatment. In this review, we will focus on the current cellular and molecular understanding of metabolic changes, epigenetic remodeling, and transcriptional regulation in CD8+ T-cell exhaustion and describe hypothetical treatment approaches based on immunotherapy aiming at reinvigorating exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Md Amir Hossain
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guilai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxuan Si
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qitao Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junaid Wazir
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA.,Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Kelly B, Pearce EL. Amino Assets: How Amino Acids Support Immunity. Cell Metab 2020; 32:154-175. [PMID: 32649859 DOI: 10.1016/j.cmet.2020.06.010] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/06/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
Amino acids are fundamental building blocks supporting life. Their role in protein synthesis is well defined, but they contribute to a host of other intracellular metabolic pathways, including ATP generation, nucleotide synthesis, and redox balance, to support cellular and organismal function. Immune cells critically depend on such pathways to acquire energy and biomass and to reprogram their metabolism upon activation to support growth, proliferation, and effector functions. Amino acid metabolism plays a key role in this metabolic rewiring, and it supports various immune cell functions beyond increased protein synthesis. Here, we review the mechanisms by which amino acid metabolism promotes immune cell function, and how these processes could be targeted to improve immunity in pathological conditions.
Collapse
Affiliation(s)
- Beth Kelly
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Erika L Pearce
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.
| |
Collapse
|
29
|
Allocco JB, Alegre ML. Exploiting immunometabolism and T cell function for solid organ transplantation. Cell Immunol 2020; 351:104068. [PMID: 32139072 PMCID: PMC7150626 DOI: 10.1016/j.cellimm.2020.104068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/25/2022]
Abstract
Cellular metabolism is central to T cell function and proliferation, with most of the research to date focusing on cancer and autoimmunity. Cellular metabolism is associated with a host of physiological phenomena, from epigenetic changes, to cellular function and fate. For the purpose of this review, we will discuss the metabolism of T cells relating to their differentiation and function. We will cover a variety of metabolic processes, ranging from glycolysis to amino acid metabolism. Understanding how T cell metabolism informs T cell function may be useful to understand alloimmune responses and design novel therapies to improve graft outcome.
Collapse
Affiliation(s)
- Jennifer B Allocco
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, United States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, United States.
| |
Collapse
|
30
|
Cassim S, Pouyssegur J. Tumor Microenvironment: A Metabolic Player that Shapes the Immune Response. Int J Mol Sci 2019; 21:E157. [PMID: 31881671 PMCID: PMC6982275 DOI: 10.3390/ijms21010157] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Immune cells survey and patrol throughout the body and sometimes take residence in niche environments with distinct cellular subtypes and nutrients that may fluctuate from those in which they matured. Rooted in immune cell physiology are metabolic pathways and metabolites that not only deliver substrates and energy for growth and survival, but also instruct effector functions and cell differentiation. Unlike cancer cells, immune cells are not subject to a "Darwinian evolutionary pressure" that would allow them to adapt to developing tumors but are often irrevocably affected to local nutrient deprivation. Thus, immune cells must metabolically adapt to these changing conditions in order to perform their necessary functions. On the other hand, there is now a growing appreciation that metabolic changes occurring in cancer cells can impact on immune cell functionality and contribute to tumor immune evasion, and as such, there is a considerable and growing interest in developing techniques that target metabolism for immunotherapy. In this review, we discuss the metabolic plasticity displayed by innate and adaptive immune cells and highlight how tumor-derived lactate and tumor acidity restrict immunity. To our knowledge, this review outlines the most recent insights on how tumor microenvironment metabolically instructs immune responsiveness.
Collapse
Affiliation(s)
- Shamir Cassim
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
| | - Jacques Pouyssegur
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
- University Côte d’Azur, IRCAN, CNRS, Centre A. Lacassagne, 06189 Nice, France
| |
Collapse
|
31
|
Geltink RIK, Kyle RL, Pearce EL. Unraveling the Complex Interplay Between T Cell Metabolism and Function. Annu Rev Immunol 2019; 36:461-488. [PMID: 29677474 DOI: 10.1146/annurev-immunol-042617-053019] [Citation(s) in RCA: 525] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metabolism drives function, on both an organismal and a cellular level. In T cell biology, metabolic remodeling is intrinsically linked to cellular development, activation, function, differentiation, and survival. After naive T cells are activated, increased demands for metabolic currency in the form of ATP, as well as biomass for cell growth, proliferation, and the production of effector molecules, are met by rewiring cellular metabolism. Consequently, pharmacological strategies are being developed to perturb or enhance selective metabolic processes that are skewed in immune-related pathologies. Here we review the most recent advances describing the metabolic changes that occur during the T cell lifecycle. We discuss how T cell metabolism can have profound effects on health and disease and where it might be a promising target to treat a variety of pathologies.
Collapse
Affiliation(s)
- Ramon I Klein Geltink
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany;
| | - Ryan L Kyle
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany;
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany;
| |
Collapse
|
32
|
Patel CH, Leone RD, Horton MR, Powell JD. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat Rev Drug Discov 2019; 18:669-688. [PMID: 31363227 DOI: 10.1038/s41573-019-0032-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2019] [Indexed: 12/15/2022]
Abstract
Metabolic programming is emerging as a critical mechanism to alter immune cell activation, differentiation and function. Targeting metabolism does not completely suppress or activate the immune system but selectively regulates immune responses. The different metabolic requirements of the diverse cells that constitute an immune response provide a unique opportunity to separate effector functions from regulatory functions. Likewise, cells can be metabolically reprogrammed to promote either their short-term effector functions or long-term memory capacity. Studies in the growing field of immunometabolism support a paradigm of 'cellular selectivity based on demand', in which generic inhibitors of ubiquitous metabolic processes selectively affect cells with the greatest metabolic demand and have few effects on other cells of the body. Targeting metabolism, rather than particular cell types or cytokines, in metabolically demanding processes such as autoimmunity, graft rejection, cancer and uncontrolled inflammation could lead to successful strategies in controlling the pathogenesis of these complex disorders.
Collapse
Affiliation(s)
- Chirag H Patel
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert D Leone
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maureen R Horton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
33
|
Xiao Y, Meierhofer D. Glutathione Metabolism in Renal Cell Carcinoma Progression and Implications for Therapies. Int J Mol Sci 2019; 20:E3672. [PMID: 31357507 PMCID: PMC6696504 DOI: 10.3390/ijms20153672] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
A significantly increased level of the reactive oxygen species (ROS) scavenger glutathione (GSH) has been identified as a hallmark of renal cell carcinoma (RCC). The proposed mechanism for increased GSH levels is to counteract damaging ROS to sustain the viability and growth of the malignancy. Here, we review the current knowledge about the three main RCC subtypes, namely clear cell RCC (ccRCC), papillary RCC (pRCC), and chromophobe RCC (chRCC), at the genetic, transcript, protein, and metabolite level and highlight their mutual influence on GSH metabolism. A further discussion addresses the question of how the manipulation of GSH levels can be exploited as a potential treatment strategy for RCC.
Collapse
Affiliation(s)
- Yi Xiao
- Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany
- Freie Universität Berlin, Fachbereich Biologie, Chemie, Pharmazie, Takustraße 3, 14195 Berlin, Germany
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany.
| |
Collapse
|
34
|
Beinat C, Gowrishankar G, Shen B, Alam IS, Robinson E, Haywood T, Patel CB, Azevedo EC, Castillo JB, Ilovich O, Koglin N, Schmitt-Willich H, Berndt M, Mueller A, Zerna M, Srinivasan A, Gambhir SS. The Characterization of 18F-hGTS13 for Molecular Imaging of xC− Transporter Activity with PET. J Nucl Med 2019; 60:1812-1817. [DOI: 10.2967/jnumed.119.225870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
|
35
|
Combs JA, DeNicola GM. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers (Basel) 2019; 11:cancers11050678. [PMID: 31100816 PMCID: PMC6562400 DOI: 10.3390/cancers11050678] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
The non-essential amino acid cysteine is used within cells for multiple processes that rely on the chemistry of its thiol group. Under physiological conditions, many non-transformed tissues rely on glutathione, circulating cysteine, and the de novo cysteine synthesis (transsulfuration) pathway as sources of intracellular cysteine to support cellular processes. In contrast, many cancers require exogeneous cystine for proliferation and viability. Herein, we review how the cystine transporter, xCT, and exogenous cystine fuel cancer cell proliferation and the mechanisms that regulate xCT expression and activity. Further, we discuss the potential contribution of additional sources of cysteine to the cysteine pool and what is known about the essentiality of these processes in cancer cells. Finally, we discuss whether cyst(e)ine dependency and associated metabolic alterations represent therapeutically targetable metabolic vulnerabilities.
Collapse
Affiliation(s)
- Joseph A Combs
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
36
|
Meyer AR, Engevik AC, Willet SG, Williams JA, Zou Y, Massion PP, Mills JC, Choi E, Goldenring JR. Cystine/Glutamate Antiporter (xCT) Is Required for Chief Cell Plasticity After Gastric Injury. Cell Mol Gastroenterol Hepatol 2019; 8:379-405. [PMID: 31071489 PMCID: PMC6713894 DOI: 10.1016/j.jcmgh.2019.04.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/10/2019] [Accepted: 04/16/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Many differentiated epithelial cell types are able to reprogram in response to tissue damage. Although reprogramming represents an important physiological response to injury, the regulation of cellular plasticity is not well understood. Damage to the gastric epithelium initiates reprogramming of zymogenic chief cells into a metaplastic cell lineage known as spasmolytic polypeptide-expressing metaplasia (SPEM). The present study seeks to identify the role of xCT, a cystine/glutamate antiporter, in chief cell reprogramming after gastric injury. We hypothesize that xCT-dependent reactive oxygen species (ROS) detoxification is required for the reprogramming of chief cells into SPEM. METHODS Sulfasalazine (an xCT inhibitor) and small interfering RNA knockdown were used to target xCT on metaplastic cells in vitro. Sulfasalazine-treated wild-type mice and xCT knockout mice were analyzed. L635 or DMP-777 treatment was used to chemically induce acute gastric damage. The anti-inflammatory metabolites of sulfasalazine (sulfapyridine and mesalazine) were used as controls. Normal gastric lineages, metaplastic markers, autophagy, proliferation, xCT activity, ROS, and apoptosis were assessed. RESULTS xCT was up-regulated early as chief cells transitioned into SPEM. Inhibition of xCT or small interfering RNA knockdown blocked cystine uptake and decreased glutathione production by metaplastic cells and prevented ROS detoxification and proliferation. Moreover, xCT activity was required for chief cell reprogramming into SPEM after gastric injury in vivo. Chief cells from xCT-deficient mice showed decreased autophagy, mucus granule formation and proliferation, as well as increased levels of ROS and apoptosis compared with wild-type mice. On the other hand, the anti-inflammatory metabolites of sulfasalazine did not affect SPEM development. CONCLUSIONS The results presented here suggest that maintaining redox balance is crucial for progression through the reprogramming process and that xCT-mediated cystine uptake is required for chief cell plasticity and ROS detoxification.
Collapse
Affiliation(s)
- Anne R Meyer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Amy C Engevik
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Spencer G Willet
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri; Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Janice A Williams
- Cell Imaging Shared Resources, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Yong Zou
- Cancer Early Detection and Prevention Initiative, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Division of Allergy, Vanderbilt University School of Medicine, Nashville, Tennessee; Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - Pierre P Massion
- Cancer Early Detection and Prevention Initiative, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Division of Allergy, Vanderbilt University School of Medicine, Nashville, Tennessee; Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - Jason C Mills
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri; Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Eunyoung Choi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James R Goldenring
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
37
|
Cystine-glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc Natl Acad Sci U S A 2019; 116:9533-9542. [PMID: 31019077 PMCID: PMC6511047 DOI: 10.1073/pnas.1814932116] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
xCT, the cystine–glutamate antiporter, has been implicated in supporting both tumor growth and T cell proliferation; thus, antitumor effects of systemic xCT inhibition may be blunted by compromised antitumor immunity. This report details the unexpected finding that xCT is dispensable for T cell proliferation in vivo and for antitumor immune responses. Consequently, tumor cell xCT loss acts synergistically with the immunotherapeutic agent anti–CTLA-4, laying the foundation for utilizing specific xCT inhibitors clinically to expand the efficacy of existing anticancer immunotherapeutics. T cell-invigorating cancer immunotherapies have near-curative potential. However, their clinical benefit is currently limited, as only a fraction of patients respond, suggesting that these regimens may benefit from combination with tumor-targeting treatments. As oncogenic progression is accompanied by alterations in metabolic pathways, tumors often become heavily reliant on antioxidant machinery and may be susceptible to increases in oxidative stress. The cystine–glutamate antiporter xCT is frequently overexpressed in cancer and fuels the production of the antioxidant glutathione; thus, tumors prone to redox stress may be selectively vulnerable to xCT disruption. However, systemic inhibition of xCT may compromise antitumor immunity, as xCT is implicated in supporting antigen-induced T cell proliferation. Therefore, we utilized immune-competent murine tumor models to investigate whether cancer cell expression of xCT was required for tumor growth in vivo and if deletion of host xCT impacted antitumor immune responses. Deletion of xCT in tumor cells led to defective cystine uptake, accumulation of reactive oxygen species, and impaired tumor growth, supporting a cancer cell-autonomous role for xCT. In contrast, we observed that, although T cell proliferation in culture was exquisitely dependent on xCT expression, xCT was dispensable for T cell proliferation in vivo and for the generation of primary and memory immune responses to tumors. These findings prompted the combination of tumor cell xCT deletion with the immunotherapeutic agent anti–CTLA-4, which dramatically increased the frequency and durability of antitumor responses. Together, these results identify a metabolic vulnerability specific to tumors and demonstrate that xCT disruption can expand the efficacy of anticancer immunotherapies.
Collapse
|
38
|
Small-Molecule Ferroptotic Agents with Potential to Selectively Target Cancer Stem Cells. Sci Rep 2019; 9:5926. [PMID: 30976078 PMCID: PMC6459861 DOI: 10.1038/s41598-019-42251-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 03/22/2019] [Indexed: 02/07/2023] Open
Abstract
Effective management of advanced cancer requires systemic treatment including small molecules that target unique features of aggressive tumor cells. At the same time, tumors are heterogeneous and current evidence suggests that a subpopulation of tumor cells, called tumor initiating or cancer stem cells, are responsible for metastatic dissemination, tumor relapse and possibly drug resistance. Classical apoptotic drugs are less effective against this critical subpopulation. In the course of generating a library of open-chain epothilones, we discovered a new class of small molecule anticancer agents that has no effect on tubulin but instead kills selected cancer cell lines by harnessing reactive oxygen species to induce ferroptosis. Interestingly, we find that drug sensitivity is highest in tumor cells with a mesenchymal phenotype. Furthermore, these compounds showed enhanced toxicity towards mesenchymal breast cancer populations with cancer stem cell properties in vitro. In summary, we have identified a new class of small molecule ferroptotic agents that warrant further investigation.
Collapse
|
39
|
Wilson CS, Spaeth JM, Karp J, Stocks BT, Hoopes EM, Stein RW, Moore DJ. B lymphocytes protect islet β cells in diabetes prone NOD mice treated with imatinib. JCI Insight 2019; 5:125317. [PMID: 30964447 PMCID: PMC6538336 DOI: 10.1172/jci.insight.125317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/02/2019] [Indexed: 01/10/2023] Open
Abstract
Imatinib (Gleevec) reverses type 1 diabetes (T1D) in NOD mice and is currently in clinical trials in individuals with recent-onset disease. While research has demonstrated that imatinib protects islet β cells from the harmful effects of ER stress, the role the immune system plays in its reversal of T1D has been less well understood, and specific cellular immune targets have not been identified. In this study, we demonstrate that B lymphocytes, an immune subset that normally drives diabetes pathology, are unexpectedly required for reversal of hyperglycemia in NOD mice treated with imatinib. In the presence of B lymphocytes, reversal was linked to an increase in serum insulin concentration, but not an increase in islet β cell mass or proliferation. However, improved β cell function was reflected by a partial recovery of MafA transcription factor expression, a sensitive marker of islet β cell stress that is important to adult β cell function. Imatinib treatment was found to increase the antioxidant capacity of B lymphocytes, improving reactive oxygen species (ROS) handling in NOD islets. This study reveals a novel mechanism through which imatinib enables B lymphocytes to orchestrate functional recovery of T1D β cells.
Collapse
Affiliation(s)
- Christopher S. Wilson
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jason M. Spaeth
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jay Karp
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Blair T. Stocks
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Emilee M. Hoopes
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roland W. Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Daniel J. Moore
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
40
|
Johnson MO, Wolf MM, Madden MZ, Andrejeva G, Sugiura A, Contreras DC, Maseda D, Liberti MV, Paz K, Kishton RJ, Johnson ME, de Cubas AA, Wu P, Li G, Zhang Y, Newcomb DC, Wells AD, Restifo NP, Rathmell WK, Locasale JW, Davila ML, Blazar BR, Rathmell JC. Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism. Cell 2018; 175:1780-1795.e19. [PMID: 30392958 PMCID: PMC6361668 DOI: 10.1016/j.cell.2018.10.001] [Citation(s) in RCA: 447] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/16/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022]
Abstract
Activated T cells differentiate into functional subsets with distinct metabolic programs. Glutaminase (GLS) converts glutamine to glutamate to support the tricarboxylic acid cycle and redox and epigenetic reactions. Here, we identify a key role for GLS in T cell activation and specification. Though GLS deficiency diminished initial T cell activation and proliferation and impaired differentiation of Th17 cells, loss of GLS also increased Tbet to promote differentiation and effector function of CD4 Th1 and CD8 CTL cells. This was associated with altered chromatin accessibility and gene expression, including decreased PIK3IP1 in Th1 cells that sensitized to IL-2-mediated mTORC1 signaling. In vivo, GLS null T cells failed to drive Th17-inflammatory diseases, and Th1 cells had initially elevated function but exhausted over time. Transient GLS inhibition, however, led to increased Th1 and CTL T cell numbers. Glutamine metabolism thus has distinct roles to promote Th17 but constrain Th1 and CTL effector cell differentiation.
Collapse
Affiliation(s)
- Marc O. Johnson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Melissa M. Wolf
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ayaka Sugiura
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Diana C. Contreras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maria V. Liberti
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Katelyn Paz
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rigel J. Kishton
- Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew E. Johnson
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aguirre A. de Cubas
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Pingsheng Wu
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Gongbo Li
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Yongliang Zhang
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Dawn C. Newcomb
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Andrew D. Wells
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas P. Restifo
- Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jason W. Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Marco L. Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Lead Contact,Correspondence:
| |
Collapse
|
41
|
Schulze A, Yuneva M. The big picture: exploring the metabolic cross-talk in cancer. Dis Model Mech 2018; 11:11/8/dmm036673. [PMID: 30154190 PMCID: PMC6124556 DOI: 10.1242/dmm.036673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Metabolic reprogramming is now well established as one of the hallmarks of cancer. The renewed interest in this topic has spurred a remarkable advance in our understanding of the metabolic alterations in cancer cells and in the tumour microenvironment. Initially, this research focussed on identifying the metabolic processes that provided cancer cells with building blocks for growth or to prevent oxidative damage and death. In addition to providing detailed insight into the mechanisms by which oncogenic signalling pathways modulate metabolic processes, this research also revealed multiple nodes within the metabolic network that can be targeted for the selective elimination of cancer cells. However, recent years have seen a paradigm shift in the field of cancer metabolism; while early studies focussed mainly on the metabolic processes within a cancer cell, recent approaches also consider the impact of metabolic cross-talk between different cell types within the tumour or study cancer within the organismal metabolic context. The Review articles presented in this themed Special Collection of Disease Models & Mechanisms aim to provide an overview of the recent advances in the field. The Collection also contains research articles that describe how metabolic inhibition can improve the efficacy of targeted therapy and introduce a new zebrafish model to study metabolic tumour-host interactions. We also present 'A model for life' interviews: a new interview with Karen Vousden and a previously published one with Lewis Cantley that provide insight into these two leaders' personal scientific journeys that resulted in seminal discoveries in the field of cancer metabolism. In this Editorial, we summarise some of the key insights obtained from studying cancer metabolism. We also describe some of the many exciting developments in the field and discuss its future challenges.
Collapse
Affiliation(s)
- Almut Schulze
- Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany .,Comprehensive Cancer Center Mainfranken, Josef-Schneider-Str.6, 97080 Würzburg, Germany
| | - Mariia Yuneva
- Oncogenes and Tumour Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
42
|
Singer K, Cheng WC, Kreutz M, Ho PC, Siska PJ. Immunometabolism in cancer at a glance. Dis Model Mech 2018; 11:11/8/dmm034272. [PMID: 30076128 PMCID: PMC6124550 DOI: 10.1242/dmm.034272] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The scientific knowledge about tumor metabolism has grown at a fascinating rate in recent decades. We now know that tumors are highly active both in their metabolism of available nutrients and in the secretion of metabolic by-products. However, cancer cells can modulate metabolic pathways and thus adapt to specific nutrients. Unlike tumor cells, immune cells are not subject to a ‘micro-evolution’ that would allow them to adapt to progressing tumors that continuously develop new mechanisms of immune escape. Consequently, immune cells are often irreversibly affected and may allow or even support cancer progression. The mechanisms of how tumors change immune cell function are not sufficiently explored. It is, however, clear that commonly shared features of tumor metabolism, such as local nutrient depletion or production of metabolic ‘waste’ can broadly affect immune cells and contribute to immune evasion. Moreover, immune cells utilize different metabolic programs based on their subtype and function, and these immunometabolic pathways can be modified in the tumor microenvironment. In this review and accompanying poster, we identify and describe the common mechanisms by which tumors metabolically affect the tumor-infiltrating cells of native and adaptive immunity, and discuss how these mechanisms may lead to novel therapeutic opportunities. Summary: This ‘At a Glance’ review and accompanying poster address how tumors can negatively affect immune cells through depletion of critical nutrients or through production of toxic metabolic products.
Collapse
Affiliation(s)
- Katrin Singer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Wan-Chen Cheng
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, CH-1066 Epalinges, Vaud, Switzerland.,Ludwig Lausanne Branch, CH-1066 Epalinges, Vaud, Switzerland
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, CH-1066 Epalinges, Vaud, Switzerland.,Ludwig Lausanne Branch, CH-1066 Epalinges, Vaud, Switzerland
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
43
|
Cohen AS, Khalil FK, Welsh EA, Schabath MB, Enkemann SA, Davis A, Zhou JM, Boulware DC, Kim J, Haura EB, Morse DL. Cell-surface marker discovery for lung cancer. Oncotarget 2017; 8:113373-113402. [PMID: 29371917 PMCID: PMC5768334 DOI: 10.18632/oncotarget.23009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/11/2017] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths in the United States. Novel lung cancer targeted therapeutic and molecular imaging agents are needed to improve outcomes and enable personalized care. Since these agents typically cannot cross the plasma membrane while carrying cytotoxic payload or imaging contrast, discovery of cell-surface targets is a necessary initial step. Herein, we report the discovery and characterization of lung cancer cell-surface markers for use in development of targeted agents. To identify putative cell-surface markers, existing microarray gene expression data from patient specimens were analyzed to select markers with differential expression in lung cancer compared to normal lung. Greater than 200 putative cell-surface markers were identified as being overexpressed in lung cancers. Ten cell-surface markers (CA9, CA12, CXorf61, DSG3, FAT2, GPR87, KISS1R, LYPD3, SLC7A11 and TMPRSS4) were selected based on differential mRNA expression in lung tumors vs. non-neoplastic lung samples and other normal tissues, and other considerations involving known biology and targeting moieties. Protein expression was confirmed by immunohistochemistry (IHC) staining and scoring of patient tumor and normal tissue samples. As further validation, marker expression was determined in lung cancer cell lines using microarray data and Kaplan–Meier survival analyses were performed for each of the markers using patient clinical data. High expression for six of the markers (CA9, CA12, CXorf61, GPR87, LYPD3, and SLC7A11) was significantly associated with worse survival. These markers should be useful for the development of novel targeted imaging probes or therapeutics for use in personalized care of lung cancer patients.
Collapse
Affiliation(s)
- Allison S Cohen
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Farah K Khalil
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric A Welsh
- Biomedical Informatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Steven A Enkemann
- Molecular Genomics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrea Davis
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jun-Min Zhou
- Biostatistics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David C Boulware
- Biostatistics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jongphil Kim
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Oncologic Sciences, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David L Morse
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Oncologic Sciences, College of Medicine, University of South Florida, Tampa, FL, USA.,Department of Physics, College of Arts and Sciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
44
|
Siska PJ, Beckermann KE, Mason FM, Andrejeva G, Greenplate AR, Sendor AB, Chiang YCJ, Corona AL, Gemta LF, Vincent BG, Wang RC, Kim B, Hong J, Chen CL, Bullock TN, Irish JM, Rathmell WK, Rathmell JC. Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma. JCI Insight 2017; 2:93411. [PMID: 28614802 PMCID: PMC5470888 DOI: 10.1172/jci.insight.93411] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/05/2017] [Indexed: 12/23/2022] Open
Abstract
Cancer cells can inhibit effector T cells (Teff) through both immunomodulatory receptors and the impact of cancer metabolism on the tumor microenvironment. Indeed, Teff require high rates of glucose metabolism, and consumption of essential nutrients or generation of waste products by tumor cells may impede essential T cell metabolic pathways. Clear cell renal cell carcinoma (ccRCC) is characterized by loss of the tumor suppressor von Hippel-Lindau (VHL) and altered cancer cell metabolism. Here, we assessed how ccRCC influences the metabolism and activation of primary patient ccRCC tumor infiltrating lymphocytes (TIL). CD8 TIL were abundant in ccRCC, but they were phenotypically distinct and both functionally and metabolically impaired. ccRCC CD8 TIL were unable to efficiently uptake glucose or perform glycolysis and had small, fragmented mitochondria that were hyperpolarized and generated large amounts of ROS. Elevated ROS was associated with downregulated mitochondrial SOD2. CD8 T cells with hyperpolarized mitochondria were also visible in the blood of ccRCC patients. Importantly, provision of pyruvate to bypass glycolytic defects or scavengers to neutralize mitochondrial ROS could partially restore TIL activation. Thus, strategies to improve metabolic function of ccRCC CD8 TIL may promote the immune response to ccRCC.
Collapse
Affiliation(s)
- Peter J. Siska
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kathryn E. Beckermann
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Frank M. Mason
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Allison R. Greenplate
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Adam B. Sendor
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yun-Chen J. Chiang
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Armando L. Corona
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lelisa F. Gemta
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Benjamin G. Vincent
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Richard C. Wang
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bumki Kim
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | | | - Timothy N. Bullock
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Jonathan M. Irish
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - W. Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|