501
|
Luo X, Zhu F, Chen Z, Li D, Li B. Regulation of Metabolism Across Different Subsets of T Cells in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1011:197-209. [DOI: 10.1007/978-94-024-1170-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
502
|
Quantification of lactate from various metabolic pathways and quantification issues of lactate isotopologues and isotopmers. Sci Rep 2017; 7:8489. [PMID: 28814730 PMCID: PMC5559627 DOI: 10.1038/s41598-017-08277-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022] Open
Abstract
13C-labeled glucose combined with chromatography and mass spectrometry enables us to decipher the percentage of lactate generated from various metabolic pathways. We showed that lactate derived from glycolysis, pentose phosphate pathway, Krebs cycle, and other sources accounted for 82-90%, 6.0-11%, 0.67-1.8% and 1.5-7.9%, respectively, depending on different types of cells. When using glucose isotopomers ([1-13C]-, [3-13C]-, [4-13C]-, and [6-13C]glucose) or isotopologues ([1,2-13C2]- and [1,2,3-13C3]glucose) for tracing, the ratio of lactate derived from glucose carbon 1, 2, 3 over 4, 5, 6 via glycolysis varied significantly, ranging from 1.6 (traced with [1,2-13C2]glucose) to 0.85 (traced with [6-13C]glucose), but the theoretical ratio should be 1. The odd results might be caused by intramolecular 13C, which may significantly affect lactate fragmentation under tandem mass spectrometry condition, leading to erroneous quantification. Indeed, the fragmentation efficiency of [U-13C]lactate, [2,3-13C]lactate, and [3-13C]lactate were 1.4, 1.5 and 1.2 folds higher than lactate, respectively, but [1-13C]lactate was similar to lactate, suggesting that carbon-13 at different positions could differentially influence lactate fragmentation. This observed phenomenon was inconsistent with the data based on theoretical calculation, according to which activation energies for all lactate isotopomers and isotopologues are nearly identical. The inconsistency suggested a need for further investigation. Our study suggests that calibration is required for quantifying metabolite isotopolugues and isotopomers.
Collapse
|
503
|
Tang CY, Mauro C. Similarities in the Metabolic Reprogramming of Immune System and Endothelium. Front Immunol 2017; 8:837. [PMID: 28785263 PMCID: PMC5519526 DOI: 10.3389/fimmu.2017.00837] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/03/2017] [Indexed: 01/06/2023] Open
Abstract
Cellular metabolism has been known for its role in bioenergetics. In recent years, much light has been shed on the reprogrammable cellular metabolism underlying many vital cellular processes, such as cell activation, proliferation, and differentiation. Metabolic reprogramming in immune and endothelial cells (ECs) is being studied extensively. These cell compartments are implicated in inflammation and pathogenesis of many diseases but their similarities in metabolic reprogramming have not been analyzed in detail. One of the most notable metabolic reprogramming is the Warburg-like effect, famously described as one of the hallmarks of cancer cells. Immune cells and ECs can display this phenotype that is characterized by a metabolic switch favoring glycolysis over oxidative phosphorylation (OXPHOS) in aerobic conditions. Though energy-inefficient, aerobic glycolysis confers many benefits to the respiring cells ranging from higher rate of adenosine triphosphate production to maintaining redox homeostasis. Chemical and biological regulators either promote or perturb this effect. In this review, nitric oxide, hypoxia-inducible factor, and adenosine monophosphate-activated protein kinase have been discussed for their common involvement in metabolic reprogramming of both systems. From in vitro and animal studies, various discrepancies exist regarding the effects of those regulators on metabolic switch. However, it is generally accepted that glycolysis favors inflammatory reactions while OXPHOS favors anti-inflammatory processes. The reasons for such observation are currently subject of intense studies and not completely understood. Finally, metabolic reprogramming in immune cells and ECs does not limit to the physiological state in health but can also be observed in pathological states, such as atherosclerosis and cancer. These new insights provide us with a better understanding of the similarities in metabolic reprogramming across a number of cell types, which could pave the way for future research and possible metabolic-based therapeutics.
Collapse
Affiliation(s)
- Chu-Yik Tang
- Barts and The London School of Medicine and Dentistry, Institute of Health Sciences Education, Queen Mary University of London, London, United Kingdom
| | - Claudio Mauro
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
504
|
Wu MC, Ye WR, Zheng YJ, Zhang SS. Oxamate Enhances the Anti-Inflammatory and Insulin-Sensitizing Effects of Metformin in Diabetic Mice. Pharmacology 2017; 100:218-228. [PMID: 28728159 DOI: 10.1159/000478909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/22/2017] [Indexed: 12/26/2022]
Abstract
Metformin (MET) is the first-line drug for treating type 2 diabetes mellitus (T2DM). However, MET increases blood lactate levels in patients with T2DM. Lactate possesses proinflammatory properties and causes insulin resistance (IR). Oxamate (OXA), a lactate dehydrogenase inhibitor, can decrease tissue lactate production and blood lactate levels. This study was conducted to examine the effects of the combination of OXA and MET on inflammation, and IR in diabetic db/db mice. Supplementation of OXA to MET led to lowered tissue lactate production and serum lactate levels compared to MET alone, accompanied with further decreased tissue and blood levels of pro-inflammatory cytokines, along with better insulin sensitivity, beta-cell mass, and glycemic control in diabetic db/db mice. These results show that OXA enhances the anti-inflammatory and insulin-sensitizing effects of MET through the inhibition of tissue lactate production in db/db mice.
Collapse
Affiliation(s)
- Mu-Chao Wu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | |
Collapse
|
505
|
Stienstra R, Netea-Maier RT, Riksen NP, Joosten LAB, Netea MG. Specific and Complex Reprogramming of Cellular Metabolism in Myeloid Cells during Innate Immune Responses. Cell Metab 2017; 26:142-156. [PMID: 28683282 DOI: 10.1016/j.cmet.2017.06.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/12/2017] [Accepted: 06/02/2017] [Indexed: 12/19/2022]
Abstract
Renewed interest in immune cell metabolism has led to the emergence of a research field aimed at studying the importance of metabolic processes for an effective immune response. In addition to the adaptive immune system, cells of the myeloid lineage have been shown to undergo robust metabolic changes upon activation. Whereas the specific metabolic requirements of myeloid cells after lipopolysaccharide/TLR4 stimulation have been extensively studied, recent evidence suggested that this model does not represent a metabolic blueprint for activated myeloid cells. Instead, different microbial stimuli, pathogens, or tissue microenvironments lead to specific and complex metabolic rewiring of myeloid cells. Here we present an overview of the metabolic heterogeneity in activated myeloid cells during health and disease. Directions for future research are suggested to ultimately provide new therapeutic opportunities. The uniqueness of metabolic signatures accompanying different conditions will require tailor-made interventions to ultimately modulate aberrant myeloid cell activation during disease.
Collapse
Affiliation(s)
- Rinke Stienstra
- Department of Internal Medicine (463), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Division of Human Nutrition, Wageningen University, 6700 AA Wageningen, the Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine (463), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine (463), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine (463), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine (463), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
506
|
|
507
|
Extracellular vesicles are independent metabolic units with asparaginase activity. Nat Chem Biol 2017; 13:951-955. [PMID: 28671681 PMCID: PMC5563455 DOI: 10.1038/nchembio.2422] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 05/19/2017] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane particles involved in the exchange of a broad range of bioactive molecules between cells and the microenvironment. While it has been shown that cells can traffic metabolic enzymes via EVs much remains to be elucidated with regard to their intrinsic metabolic activity. Accordingly, herein we assessed the ability of neural stem/progenitor cell (NSC)-derived EVs to consume and produce metabolites. Both our metabolomics and functional analyses revealed that EVs harbour L-asparaginase activity catalysed by the enzyme Asparaginase-like protein 1 (Asrgl1). Critically, we show that Asrgl1 activity is selective for asparagine and is devoid of glutaminase activity. We found that mouse and human NSC-derived EVs traffic ASRGL1. Our results demonstrate for the first time that NSC EVs function as independent, extracellular metabolic units able to modify the concentrations of critical nutrients, with the potential to affect the physiology of their microenvironment.
Collapse
|
508
|
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: 270] [Impact Index Per Article: 33.8] [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
|
509
|
Tumor-derived factors affecting immune cells. Cytokine Growth Factor Rev 2017; 36:79-87. [PMID: 28606733 DOI: 10.1016/j.cytogfr.2017.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022]
Abstract
Tumor progression is accompanied by the production of a wide array of immunosuppressive factors by tumor and non-tumor cells forming the tumor microenvironment. These factors belonging to cytokines, growth factors, metabolites, glycan-binding proteins and glycoproteins are responsible for the establishment of immunosuppressive networks leading towards tumor promotion, invasion and metastasis. In pre-clinical tumor models, the inactivation of some of these suppressive networks reprograms the phenotypic and functional features of tumor-infiltrating immune cells, ultimately favoring effective anti-tumor immune responses. We will discuss factors and mechanisms identified in both mouse and human tumors, and the possibility to associate drugs inhibiting these mechanisms with new immunotherapy strategies already entered in the clinical practice.
Collapse
|
510
|
Goers L, Ainsworth C, Goey CH, Kontoravdi C, Freemont PS, Polizzi KM. Whole-cell Escherichia coli lactate biosensor for monitoring mammalian cell cultures during biopharmaceutical production. Biotechnol Bioeng 2017; 114:1290-1300. [PMID: 28112405 PMCID: PMC5412874 DOI: 10.1002/bit.26254] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/11/2017] [Accepted: 01/19/2017] [Indexed: 02/06/2023]
Abstract
Many high-value added recombinant proteins, such as therapeutic glycoproteins, are produced using mammalian cell cultures. In order to optimize the productivity of these cultures it is important to monitor cellular metabolism, for example the utilization of nutrients and the accumulation of metabolic waste products. One metabolic waste product of interest is lactic acid (lactate), overaccumulation of which can decrease cellular growth and protein production. Current methods for the detection of lactate are limited in terms of cost, sensitivity, and robustness. Therefore, we developed a whole-cell Escherichia coli lactate biosensor based on the lldPRD operon and successfully used it to monitor lactate concentration in mammalian cell cultures. Using real samples and analytical validation we demonstrate that our biosensor can be used for absolute quantification of metabolites in complex samples with high accuracy, sensitivity, and robustness. Importantly, our whole-cell biosensor was able to detect lactate at concentrations more than two orders of magnitude lower than the industry standard method, making it useful for monitoring lactate concentrations in early phase culture. Given the importance of lactate in a variety of both industrial and clinical contexts we anticipate that our whole-cell biosensor can be used to address a range of interesting biological questions. It also serves as a blueprint for how to capitalize on the wealth of genetic operons for metabolite sensing available in nature for the development of other whole-cell biosensors. Biotechnol. Bioeng. 2017;114: 1290-1300. © 2017 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lisa Goers
- Department of Life SciencesImperial College LondonLondonSW7 2AZUK
- Centre for Synthetic Biology and InnovationImperial College LondonLondonUK
| | - Catherine Ainsworth
- Centre for Synthetic Biology and InnovationImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
| | - Cher Hui Goey
- Department of Chemical EngineeringImperial College LondonLondonUK
| | - Cleo Kontoravdi
- Centre for Synthetic Biology and InnovationImperial College LondonLondonUK
- Department of Chemical EngineeringImperial College LondonLondonUK
| | - Paul S. Freemont
- Centre for Synthetic Biology and InnovationImperial College LondonLondonUK
- Department of MedicineImperial College LondonLondonUK
| | - Karen M. Polizzi
- Department of Life SciencesImperial College LondonLondonSW7 2AZUK
- Centre for Synthetic Biology and InnovationImperial College LondonLondonUK
| |
Collapse
|
511
|
Weyand CM, Zeisbrich M, Goronzy JJ. Metabolic signatures of T-cells and macrophages in rheumatoid arthritis. Curr Opin Immunol 2017; 46:112-120. [PMID: 28538163 DOI: 10.1016/j.coi.2017.04.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022]
Abstract
In most autoimmune diseases, a decade-long defect in self-tolerance eventually leads to clinically relevant, tissue-destructive inflammatory disease. The pathogenic potential of chronic persistent immune responses during the pre-clinical and clinical phase is ultimately linked to the bioenergetic fitness of innate and adaptive immune cells. Chronic immune cell stimulation, high cellular turn-over, structural damage to the host tissue and maladaptive wound healing, all require a reliable supply of nutrients, oxygen, and biosynthetic precursors. Here, we use the model system of rheumatoid arthritis (RA) to discuss immunometabolism from the vantage point of T-cells and macrophages that encounter fundamentally different metabolic stress scenarios in the RA host. We outline the general principle that both insufficient nutrient supply, as well as nutrient excess generate cellular stress responses and guide immune function. ATPlow, NADPHhigh, ROSlow T-cells hyperproliferate and are forced into premature senescence. ATPhigh, ROShigh macrophages dimerize the glycolytic enzyme pyruvate kinase to amplify STAT3-dependent inflammatory effector functions. A corollary of this model is that simple nutraceutical interventions will be insufficient to re-educate the immune system in RA. Instead, interference with cell-type-exclusive and differentiation-stage-dependent metabolic setpoints will be needed to reprogram arthritogenic pathways.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA , United States.
| | - Markus Zeisbrich
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA , United States
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA , United States
| |
Collapse
|
512
|
Buck MD, Sowell RT, Kaech SM, Pearce EL. Metabolic Instruction of Immunity. Cell 2017; 169:570-586. [PMID: 28475890 PMCID: PMC5648021 DOI: 10.1016/j.cell.2017.04.004] [Citation(s) in RCA: 831] [Impact Index Per Article: 103.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/02/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Choices have consequences. Immune cells survey and migrate throughout the body and sometimes take residence in niche environments with distinct communities of cells, extracellular matrix, and nutrients that may differ from those in which they matured. Imbedded in immune cell physiology are metabolic pathways and metabolites that not only provide energy and substrates for growth and survival, but also instruct effector functions, differentiation, and gene expression. This review of immunometabolism will reference the most recent literature to cover the choices that environments impose on the metabolism and function of immune cells and highlight their consequences during homeostasis and disease.
Collapse
Affiliation(s)
- Michael D Buck
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Division of Biology and Biomedical Sciences Immunology Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan T Sowell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Susan M Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| |
Collapse
|
513
|
Ho PC, Kaech SM. Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Curr Opin Immunol 2017; 46:38-44. [PMID: 28458087 DOI: 10.1016/j.coi.2017.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022]
Abstract
T cells patrol our bodies preventing pathogenic infections and malignant cell outgrowth. However, T cells must be properly controlled because aberrant or persistent T cell responses can damage tissues and contribute to autoimmune diseases and other chronic inflammatory diseases including metabolic syndrome. One regulatory mechanism utilized in immune cells is immunometabolic regulation, which ensures immune cells properly respond to systemic and peripheral metabolic cues. Recent work has suggested that deregulated metabolism in tumor cells creates a microenvironmental barrier for mounting effective anti-tumor immune responses. Here, we discuss how tumor cells evade immunosurveillance by modulating metabolic checkpoints in immune cells and discuss how memory T cells could provide effective anti-tumor responses by sustaining metabolic fitness and longevity.
Collapse
Affiliation(s)
- Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Vaud 1066, Switzerland; Ludwig Lausanne Branch, University of Lausanne, Lausanne, Vaud 1066, Switzerland.
| | - Susan M Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
514
|
Beckermann KE, Dudzinski SO, Rathmell JC. Dysfunctional T cell metabolism in the tumor microenvironment. Cytokine Growth Factor Rev 2017; 35:7-14. [PMID: 28456467 DOI: 10.1016/j.cytogfr.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 12/17/2022]
Abstract
Metabolic and signaling pathways are integrated to determine T cell fate and function. As stimulated T cells gain distinct effector functions, specific metabolic programs and demands are also adopted. These changes are essential for T cell effector function, and alterations or dysregulation of metabolic pathways can modulate T cell function. One physiological setting that impacts T cell metabolism is the tumor microenvironment. The metabolism of cancer cells themselves can limit nutrients and accumulate waste products. In addition to the expression of inhibitory ligands that directly modify T cell physiology, T cell metabolism may be strongly inhibited in the tumor microenvironment. This suppression of T cell metabolism may inhibit effector T cell activity while promoting suppressive regulatory T cells, and act as a barrier to effective immunotherapies. A thorough understanding of the effect of the tumor microenvironment on the immune system will support the continued improvement of immune based therapies for cancer patients.
Collapse
Affiliation(s)
- Kathryn E Beckermann
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Stephanie O Dudzinski
- Department of Biomedical Engineering, 2301 Vanderbilt Place, Nashville, TN 37235, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
515
|
Abstract
One of the fundamental traits of immune cells in rheumatoid arthritis (RA) is their ability to proliferate, a property shared with the joint-resident cells that form the synovial pannus. The building of biomass imposes high demands for energy and biosynthetic precursors, implicating metabolic control as a basic disease mechanism. During preclinical RA, when autoreactive T cells expand and immunological tolerance is broken, the main sites of disease are the secondary lymphoid tissues. Naive CD4+ T cells from patients with RA have a distinct metabolic signature, characterized by dampened glycolysis, low ATP levels and enhanced shunting of glucose into the pentose phosphate pathway. Equipped with high levels of NADPH and depleted of intracellular reactive oxygen species, such T cells hyperproliferate and acquire proinflammatory effector functions. During clinical RA, immune cells coexist with stromal cells in the acidic milieu of the inflamed joint. This microenvironment is rich in metabolic intermediates that are released into the extracellular space to shape cell-cell communication and the functional activity of tissue-resident cells. Increasing awareness of how metabolites regulate signalling pathways, guide post-translational modifications and condition the tissue microenvironment will help to connect environmental factors with the pathogenic behaviour of T cells in RA.
Collapse
|
516
|
Zou Y, Zeng S, Huang M, Qiu Q, Xiao Y, Shi M, Zhan Z, Liang L, Yang X, Xu H. Inhibition of 6-phosphofructo-2-kinase suppresses fibroblast-like synoviocytes-mediated synovial inflammation and joint destruction in rheumatoid arthritis. Br J Pharmacol 2017; 174:893-908. [PMID: 28239846 DOI: 10.1111/bph.13762] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Abnormal glycolytic metabolism contributes to joint inflammation in rheumatoid arthritis (RA). The aims of this study were to investigate the role of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a bifunctional enzyme that controls the glycolytic rate, in regulating fibroblast-like synoviocyte (FLS)-mediated synovial inflammation and invasiveness in RA. EXPERIMENTAL APPROACH A specific inhibitor of PFKFB3, PFK15, and siRNA were used to evaluate the role of PFKFB3. Protein expression was measured by Western blotting or immunofluorescence staining. The expression of cytokines was determined by quantitative real-time PCR. Migration and invasion were measured using a Boyden chamber assay. A mouse model of collagen-induced arthritis (CIA) was used to evaluate the in vivo effect of PFK15. KEY RESULTS PFKFB3 expression was increased in the synovial tissue and FLSs from RA patients compared with osteoarthritis patients. PFKFB3 inhibition decreased the expression of IL-8, IL-6, CCL-2 and CXCL-10 and the proliferation, migration and invasion of RA FLSs. PFK15 suppressed TNF-α-induced activation of NF-κB and p38, JNK and ERK MAPK signals in RA FLSs. PFK15 treatment also suppressed glucose uptake and lactate secretion. Lactate reversed the inhibitory effect of PFK15 or PFKFB3 siRNA on cytokine expression and migration of RA FLSs. Lactate was also involved in PFKFB3-mediated activation of NF-κB and MAPKs. Intraperitoneal injection of PFK15 in mice with CIA attenuated joint inflammation. CONCLUSION AND IMPLICATIONS Elevated PFKFB3 expression might contribute to synovial inflammation and aggressive behaviours of RA FLSs, suggesting a novel strategy of targeting PFKFB3 to prevent synovial inflammation and joint destruction in RA.
Collapse
Affiliation(s)
- Yaoyao Zou
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shan Zeng
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mingcheng Huang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qian Qiu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youjun Xiao
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Maohua Shi
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhongping Zhan
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liuqin Liang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiuyan Yang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
517
|
Camps J, Joven J. Metabolite profiling can change health-care delivery to obese patients with fatty liver disease: the search for biomarkers. Clin Chem Lab Med 2017; 55:501-506. [PMID: 27816954 DOI: 10.1515/cclm-2016-0762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/01/2016] [Indexed: 12/25/2022]
Abstract
Comorbidities associated with obesity have become a worldwide public health concern. Obesity-associated hepatic steatosis is not benign, and the risk of developing severe liver disease is high. Currently, biopsy is the only clinical tool available for the diagnosis of pathological alterations in the liver. However, the procedure is painful and not without risk. As such, there is a need to identify non-invasive biomarkers of steatosis. There has been considerable progress in this area, but research appears to be limited to measurements of levels of certain parameters in patients with liver impairment relative to those of healthy controls. The clinically relevant aim should be to distinguish, at an early stage, those obese individuals with liver steatosis from those obese individuals without it. Plasma constituents that act as surrogates of altered hepatic energy metabolism in response to food intake are likely candidates. Targeted metabolomics, combined with quantitation of the metabolites involved, has been shown to be an efficient measurement tool. Indeed, the evaluation of exhaled volatile compounds might be sufficient, while other rapid, sensitive, and reproducible methods have been validated in preliminary studies in various clinical settings. Metabolomics methods are promising but require considerable expertise and sophisticated (and expensive) equipment not readily available in all centers. The challenge is to adapt this newly acquired, expanding knowledge to current, reasonably equipped clinical laboratories, while substantially reducing costs. Good outcomes are urgently required if effective prevention programs are to be developed to decrease the prevalence of liver disease.
Collapse
|
518
|
Hu X, Chao M, Wu H. Central role of lactate and proton in cancer cell resistance to glucose deprivation and its clinical translation. Signal Transduct Target Ther 2017; 2:16047. [PMID: 29263910 PMCID: PMC5661620 DOI: 10.1038/sigtrans.2016.47] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/21/2016] [Accepted: 12/26/2016] [Indexed: 12/14/2022] Open
Abstract
Targeting common weaknesses of cancer is an important strategy for cancer therapy. Glucose is a nutrient that maintains essential cellular metabolism, supporting cancer cell survival, growth and proliferation. Depriving glucose rapidly kills cancer cells. Most cancer cells possess a feature called Warburg effect, which refers to that cancer cells even with ample oxygen exhibit an exceptionally high glycolysis rate and convert most incoming glucose to lactate. Although it is recognized that Warburg effect confers growth advantage to cancer cells when glucose supply is sufficient, this feature could be considered as a fatal weakness of cancer cells when glucose supply is a problem. As glucose supply in many solid tumors is poor, and as most cancer cells have exceptionally high glycolytic capacity, maximizing cancer cell glycolysis rate would possibly exhaust intratumoral glucose, leading cancer cell to death. Lactate and proton are two common factors in solid tumors, they jointly protect cancer cells against glucose deprivation, and they are also powerful regulators dictating glucose metabolic phenotypes of cancer cells. Disrupting the joint action of lactate and proton, for example, by means of bicarbonate infusion into tumor, could maximize cancer cell glycolytic rate to rapidly use up glucose, expose their vulnerability to glucose deprivation and ultimately kill cancer cells. A pilot clinical study demonstrated that this approach achieved a remarkable improvement in local control of large and huge hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xun Hu
- Cancer Institute (a Key Laboratory For Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Chao
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wu
- Cancer Institute (a Key Laboratory For Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
519
|
Renner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, Kreutz M. Metabolic Hallmarks of Tumor and Immune Cells in the Tumor Microenvironment. Front Immunol 2017; 8:248. [PMID: 28337200 PMCID: PMC5340776 DOI: 10.3389/fimmu.2017.00248] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/20/2017] [Indexed: 12/14/2022] Open
Abstract
Cytotoxic T lymphocytes and NK cells play an important role in eliminating malignant tumor cells and the number and activity of tumor-infiltrating T cells represent a good marker for tumor prognosis. Based on these findings, immunotherapy, e.g., checkpoint blockade, has received considerable attention during the last couple of years. However, for the majority of patients, immune control of their tumors is gray theory as malignant cells use effective mechanisms to outsmart the immune system. Increasing evidence suggests that changes in tumor metabolism not only ensure an effective energy supply and generation of building blocks for tumor growth but also contribute to inhibition of the antitumor response. Immunosuppression in the tumor microenvironment is often based on the mutual metabolic requirements of immune cells and tumor cells. Cytotoxic T and NK cell activation leads to an increased demand for glucose and amino acids, a well-known feature shown by tumor cells. These close metabolic interdependencies result in metabolic competition, limiting the proliferation, and effector functions of tumor-specific immune cells. Moreover, not only nutrient restriction but also tumor-driven shifts in metabolite abundance and accumulation of metabolic waste products (e.g., lactate) lead to local immunosuppression, thereby facilitating tumor progression and metastasis. In this review, we describe the metabolic interplay between immune cells and tumor cells and discuss tumor cell metabolism as a target structure for cancer therapy. Metabolic (re)education of tumor cells is not only an approach to kill tumor cells directly but could overcome metabolic immunosuppression in the tumor microenvironment and thereby facilitate immunotherapy.
Collapse
Affiliation(s)
- Kathrin Renner
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Katrin Singer
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Gudrun E Koehl
- Department of Surgery, University Hospital Regensburg , Regensburg , Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg , Regensburg , Germany
| | - Katrin Peter
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Peter J Siska
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Marina Kreutz
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Regensburg Center for Interventional Immunology, Regensburg, Germany
| |
Collapse
|
520
|
Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 2017; 43:74-89. [PMID: 28267587 DOI: 10.1016/j.semcancer.2017.03.001] [Citation(s) in RCA: 405] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Abstract
The link between cancer metabolism and immunosuppression, inflammation and immune escape has generated major interest in investigating the effects of low pH on tumor immunity. Indeed, microenvironmental acidity may differentially impact on diverse components of tumor immune surveillance, eventually contributing to immune escape and cancer progression. Although the molecular pathways underlying acidity-related immune dysfunctions are just emerging, initial evidence indicates that antitumor effectors such as T and NK cells tend to lose their function and undergo a state of mostly reversible anergy followed by apoptosis, when exposed to low pH environment. At opposite, immunosuppressive components such as myeloid cells and regulatory T cells are engaged by tumor acidity to sustain tumor growth while blocking antitumor immune responses. Local acidity could also profoundly influence bioactivity and distribution of antibodies, thus potentially interfering with the clinical efficacy of therapeutic antibodies including immune checkpoint inhibitors. Hence tumor acidity is a central regulator of cancer immunity that orchestrates both local and systemic immunosuppression and that may offer a broad panel of therapeutic targets. This review outlines the fundamental pathways of acidity-driven immune dysfunctions and sheds light on the potential strategies that could be envisaged to potentiate immune-mediated tumor control in cancer patients.
Collapse
|
521
|
Pucino V, Bombardieri M, Pitzalis C, Mauro C. Lactate at the crossroads of metabolism, inflammation, and autoimmunity. Eur J Immunol 2017; 47:14-21. [PMID: 27883186 DOI: 10.1002/eji.201646477] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/02/2016] [Accepted: 11/23/2016] [Indexed: 12/19/2022]
Abstract
For a long time after its discovery at the beginning of the 20th century, lactate was considered a waste product of cellular metabolism. Starting in the early '90s, however, lactate has begun to be recognized as an active molecule capable of modulating the immune response. Inflammatory sites, including in rheumatoid arthritis (RA) synovitis, are characterized by the accumulation of lactate, which is partly responsible for the establishment of an acidic environment. We have recently reported that T cells sense lactate via the expression of specific transporters, leading to inhibition of their motility. Importantly, this "stop migration signal" is dependent upon lactate's interference with intracellular metabolic pathways, specifically glycolysis. Furthermore, lactate promotes the switch of CD4+ T cells to an IL-17+ subset, and reduces the cytolytic capacity of CD8+ T cells. These phenomena might be responsible for the formation of ectopic lymphoid structures and autoantibody production in inflammatory sites such as in RA synovitis, Sjogren syndrome salivary glands, and multiple sclerosis plaques. Here, we review the roles of lactate in the modulation of the inflammatory immune response.
Collapse
Affiliation(s)
- Valentina Pucino
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Michele Bombardieri
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
522
|
PHD2 Is a Regulator for Glycolytic Reprogramming in Macrophages. Mol Cell Biol 2016; 37:MCB.00236-16. [PMID: 27795296 DOI: 10.1128/mcb.00236-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 10/07/2016] [Indexed: 01/17/2023] Open
Abstract
The prolyl-4-hydroxylase domain (PHD) enzymes are regarded as the molecular oxygen sensors. There is an interplay between oxygen availability and cellular metabolism, which in turn has significant effects on the functionality of innate immune cells, such as macrophages. However, if and how PHD enzymes affect macrophage metabolism are enigmatic. We hypothesized that macrophage metabolism and function can be controlled via manipulation of PHD2. We characterized the metabolic phenotypes of PHD2-deficient RAW cells and primary PHD2 knockout bone marrow-derived macrophages (BMDM). Both showed typical features of anaerobic glycolysis, which were paralleled by increased pyruvate dehydrogenase kinase 1 (PDK1) protein levels and a decreased pyruvate dehydrogenase enzyme activity. Metabolic alterations were associated with an impaired cellular functionality. Inhibition of PDK1 or knockout of hypoxia-inducible factor 1α (HIF-1α) reversed the metabolic phenotype and impaired the functionality of the PHD2-deficient RAW cells and BMDM. Taking these results together, we identified a critical role of PHD2 for a reversible glycolytic reprogramming in macrophages with a direct impact on their function. We suggest that PHD2 serves as an adjustable switch to control macrophage behavior.
Collapse
|
523
|
Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, Kastenberger M, Bogdan C, Schleicher U, Mackensen A, Ullrich E, Fichtner-Feigl S, Kesselring R, Mack M, Ritter U, Schmid M, Blank C, Dettmer K, Oefner PJ, Hoffmann P, Walenta S, Geissler EK, Pouyssegur J, Villunger A, Steven A, Seliger B, Schreml S, Haferkamp S, Kohl E, Karrer S, Berneburg M, Herr W, Mueller-Klieser W, Renner K, Kreutz M. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab 2016; 24:657-671. [PMID: 27641098 DOI: 10.1016/j.cmet.2016.08.011] [Citation(s) in RCA: 1233] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 04/20/2016] [Accepted: 08/19/2016] [Indexed: 10/21/2022]
Abstract
Elevated lactate dehydrogenase A (LDHA) expression is associated with poor outcome in tumor patients. Here we show that LDHA-associated lactic acid accumulation in melanomas inhibits tumor surveillance by T and NK cells. In immunocompetent C57BL/6 mice, tumors with reduced lactic acid production (Ldhalow) developed significantly slower than control tumors and showed increased infiltration with IFN-γ-producing T and NK cells. However, in Rag2-/-γc-/- mice, lacking lymphocytes and NK cells, and in Ifng-/- mice, Ldhalow and control cells formed tumors at similar rates. Pathophysiological concentrations of lactic acid prevented upregulation of nuclear factor of activated T cells (NFAT) in T and NK cells, resulting in diminished IFN-γ production. Database analyses revealed negative correlations between LDHA expression and T cell activation markers in human melanoma patients. Our results demonstrate that lactic acid is a potent inhibitor of function and survival of T and NK cells leading to tumor immune escape.
Collapse
Affiliation(s)
- Almut Brand
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katrin Singer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gudrun E Koehl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Marlene Kolitzus
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gabriele Schoenhammer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Annette Thiel
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christina Bruss
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastian Klobuch
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katrin Peter
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Michael Kastenberger
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Evelyn Ullrich
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, 91054 Erlangen, Germany; Cellular Immunology, Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine of the University Hospital Frankfurt, Goethe-University, 60590 Frankfurt, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Rebecca Kesselring
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Matthias Mack
- Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany; Department of Internal Medicine II - Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Uwe Ritter
- Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Maximilian Schmid
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Blank
- Division of Immunology, the Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam 1066CX, the Netherlands
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Stefan Walenta
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jacques Pouyssegur
- Institute of Research on Cancer and Aging, University of Nice-Sophia Antipolis, Centre A. Lacassagne, 06189 Nice, France; Centre Scientifique de Monaco (CSM), 98000 Monaco, Monaco
| | - Andreas Villunger
- Medical University Innsbruck, Biocenter, Division of Developmental Immunology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, 6020 Innsbruck, Austria
| | - André Steven
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology Halle/Saale, 06112 Halle, Germany
| | - Barbara Seliger
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology Halle/Saale, 06112 Halle, Germany
| | - Stephan Schreml
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Elisabeth Kohl
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sigrid Karrer
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Mueller-Klieser
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
524
|
Emerging concepts of T cell metabolism as a target of immunotherapy. Nat Immunol 2016; 17:364-8. [PMID: 27002844 DOI: 10.1038/ni.3415] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/17/2016] [Indexed: 12/27/2022]
Abstract
T cells have a pivotal protective role in defense against infection and cancer but also are instrumental in the development of many autoimmune diseases. The regulation of nutrient uptake and utilization in T cells is critically important for the control of their differentiation, and manipulating metabolic pathways in these cells can alter their function and longevity. While the importance of T cell metabolic remodeling in different physiological settings is not fully understood, there is a growing realization that inappropriate metabolic remodeling underlies many aberrant immune responses and that manipulating cellular metabolism can beneficially enhance or temper immunity. Here we comment on the basic metabolic pathways in T cells, followed by a discussion on up-to-date findings about the relationship between metabolism and T cell function and longevity. Furthermore, we expand on potential approaches and applications in which T cells might be manipulated by the reprogramming of metabolic pathways for therapeutic purposes.
Collapse
|
525
|
Hernández-Aguilera A, Fernández-Arroyo S, Cuyàs E, Luciano-Mateo F, Cabre N, Camps J, Lopez-Miranda J, Menendez JA, Joven J. Epigenetics and nutrition-related epidemics of metabolic diseases: Current perspectives and challenges. Food Chem Toxicol 2016; 96:191-204. [PMID: 27503834 DOI: 10.1016/j.fct.2016.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/03/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
We live in a world fascinated by the relationship between disease and nutritional disequilibrium. The subtle and slow effects of chronic nutrient toxicity are a major public health concern. Since food is potentially important for the development of "metabolic memory", there is a need for more information on the type of nutrients causing adverse or toxic effects. We now know that metabolic alterations produced by excessive intake of some nutrients, drugs and chemicals directly impact epigenetic regulation. We envision that understanding how metabolic pathways are coordinated by environmental and genetic factors will provide novel insights for the treatment of metabolic diseases. New methods will enable the assembly and analysis of large sets of complex molecular and clinical data for understanding how inflammation and mitochondria affect bioenergetics, epigenetics and health. Collectively, the observations we highlight indicate that energy utilization and disease are intimately connected by epigenetics. The challenge is to incorporate metabolo-epigenetic data in better interpretations of disease, to expedite therapeutic targeting of key pathways linking nutritional toxicity and metabolism. An additional concern is that changes in the parental phenotype are detectable in the methylome of subsequent offspring. The effect might create a menace to future generations and preconceptional considerations.
Collapse
Affiliation(s)
- Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Noemi Cabre
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jose Lopez-Miranda
- Lipid and Atherosclerosis Unit, IMIBIC, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain; The Campus of International Excellence Southern Catalonia, Tarragona, Spain.
| |
Collapse
|
526
|
Chao M, Wu H, Jin K, Li B, Wu J, Zhang G, Yang G, Hu X. A nonrandomized cohort and a randomized study of local control of large hepatocarcinoma by targeting intratumoral lactic acidosis. eLife 2016; 5:15691. [PMID: 27481188 PMCID: PMC4970867 DOI: 10.7554/elife.15691] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Background: Previous works suggested that neutralizing intratumoral lactic acidosis combined with glucose deprivation may deliver an effective approach to control tumor. We did a pilot clinical investigation, including a nonrandomized (57 patients with large HCC) and a randomized controlled (20 patients with large HCC) study. Methods: The patients were treated with transarterial chemoembolization (TACE) with or without bicarbonate local infusion into tumor. Results: In the nonrandomized controlled study, geometric mean of viable tumor residues (VTR) in TACE with bicarbonate was 6.4-fold lower than that in TACE without bicarbonate (7.1% [95% CI: 4.6%–10.9%] vs 45.6% [28.9%–72.0%]; p<0.0001). This difference was recapitulated by a subsequent randomized controlled study. TACE combined with bicarbonate yielded a 100% objective response rate (ORR), whereas the ORR treated with TACE alone was 44.4% (nonrandomized) and 63.6% (randomized). The survival data suggested that bicarbonate may bring survival benefit. Conclusions: Bicarbonate markedly enhances the anticancer activity of TACE. Funding: Funded by National Natural Science Foundation of China. Clinical trial number: ChiCTR-IOR-14005319. Surgery is the main treatment for liver cancer, but the most common liver cancer – called hepatocellular carcinoma – can sometimes become too large to remove safely. An alternative option to kill the tumor is to block its blood supply via a process called embolization. This procedure deprives the tumor cells of oxygen and nutrients such as glucose. However, embolization also prevents a chemical called lactic acid – which is commonly found around tumors – from being removed. Lactic acid actually helps to protect cancer cells and also aids the growth of new blood vessels, and so the “trapped” lactic acid may reduce the anticancer activity of embolization. Previous works suggested that neutralizing the acidic environment in a tumor while depriving it of glucose via embolization could become a new treatment option for cancer patients. Chao et al. now report a small clinical trial that tested this idea and involved patients with large hepatocellular carcinomas. First, a group of thirty patients received the embolization treatment together with an injection of bicarbonate – a basic compound used to neutralize the lactic acid – that was delivered directly to the tumor. The neutralization killed these large tumors more effectively than what is typically seen in patients who just undergo embolization Chao et al. then recruited another twenty patients and randomly assigned them to receive either just the embolization or the embolization with bicarbonate treatment. This randomized trial showed that the tumors died more and patients survived for longer if they received the bicarbonate together with the embolization treatment compared to those patients that were only embolized. In fact, four patients initially assigned to, and treated in, the embolization-only group subsequently asked to cross over to, and indeed received, the bicarbonate treatment as well. These data indicate that this bicarbonate therapy may indeed be effective for patients with large tumors that are not amenable to surgery. In future, larger clinical trials will need to be carried out to verify these initial findings.
Collapse
Affiliation(s)
- Ming Chao
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wu
- Cancer Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Jin
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jianjun Wu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Guangqiang Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Gong Yang
- Vanderbilt University Medical Center, Nashville, United States
| | - Xun Hu
- Cancer Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
527
|
Corrado M, Scorrano L, Campello S. Changing perspective on oncometabolites: from metabolic signature of cancer to tumorigenic and immunosuppressive agents. Oncotarget 2016; 7:46692-46706. [PMID: 27083002 PMCID: PMC5216830 DOI: 10.18632/oncotarget.8727] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/31/2016] [Indexed: 12/12/2022] Open
Abstract
During tumorigenesis, the shift from oxidative phosphorylation to glycolysis in ATP production accounts for the dramatic change in the cellular metabolism and represents one of the major steps leading to tumour formation. The so-called Warburg effect is currently considered something more than a mere modification in the cellular metabolism. The paradox that during cancer cell proliferation the increase in energy need is supplied by glycolysis can be only explained by taking into account the many roles that intermediates of glycolysis or TCA cycle play in cellular physiology, besides energy production. Recent studies have shown that metabolic intermediates induce changes in chromatin structure or drive neo-angiogenesis. In this review, we present some of the latest findings in the study of cancer metabolism with particular attention to how tumour metabolism and its microenvironment can favour tumour growth and aggressiveness, by hijacking and dampening the anti-tumoral immune response.
Collapse
Affiliation(s)
- Mauro Corrado
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy
- IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Luca Scorrano
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Silvia Campello
- IRCCS Fondazione Santa Lucia, Roma, Italy
- Department of Biology, University of Roma Tor Vergata, Roma, Italy
| |
Collapse
|
528
|
Zhang G, Hailemariam D, Dervishi E, Goldansaz SA, Deng Q, Dunn SM, Ametaj BN. Dairy cows affected by ketosis show alterations in innate immunity and lipid and carbohydrate metabolism during the dry off period and postpartum. Res Vet Sci 2016; 107:246-256. [PMID: 27474003 DOI: 10.1016/j.rvsc.2016.06.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 05/17/2016] [Accepted: 06/18/2016] [Indexed: 12/17/2022]
Abstract
The objective of this investigation was to search for alterations in blood variables related to innate immunity and carbohydrate and lipid metabolism during the transition period in cows affected by ketosis. One hundred multiparous Holstein dairy cows were involved in the study. Blood samples were collected at -8, -4, week of disease diagnosis (+1 to +3weeks), and +4weeks relative to parturition from 6 healthy cows (CON) and 6 cows with ketosis and were analyzed for serum variables. Results showed that cows with ketosis had greater concentrations of serum β-hydroxybutyric acid (BHBA), interleukin (IL)-6, tumor necrosis factor (TNF), serum amyloid A (SAA), and lactate in comparison with the CON animals. Serum concentrations of BHBA, IL-6, TNF, and lactate were greater starting at -8 and -4weeks prior to parturition in cows with ketosis vs those of CON group. Cows with ketosis also had lower DMI and milk production vs CON cows. Milk fat also was lower in ketotic cows at diagnosis of disease. Cows affected by ketosis showed an activated innate immunity and altered carbohydrate and lipid metabolism several weeks prior to diagnosis of disease. Serum IL-6 and lactate were the strongest discriminators between ketosis cows and CON ones before the occurrence of ketosis, which might be useful as predictive biomarkers of the disease state.
Collapse
Affiliation(s)
- Guanshi Zhang
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Dagnachew Hailemariam
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Elda Dervishi
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Seyed Ali Goldansaz
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2M9, Canada
| | - Qilan Deng
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Suzanna M Dunn
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Burim N Ametaj
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada.
| |
Collapse
|
529
|
Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol 2016; 12:385-97. [PMID: 27225300 DOI: 10.1038/nrrheum.2016.69] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
530
|
DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. SCIENCE ADVANCES 2016; 2:e1600200. [PMID: 27386546 PMCID: PMC4928883 DOI: 10.1126/sciadv.1600200] [Citation(s) in RCA: 2007] [Impact Index Per Article: 223.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/29/2016] [Indexed: 04/14/2023]
Abstract
Tumors reprogram pathways of nutrient acquisition and metabolism to meet the bioenergetic, biosynthetic, and redox demands of malignant cells. These reprogrammed activities are now recognized as hallmarks of cancer, and recent work has uncovered remarkable flexibility in the specific pathways activated by tumor cells to support these key functions. In this perspective, we provide a conceptual framework to understand how and why metabolic reprogramming occurs in tumor cells, and the mechanisms linking altered metabolism to tumorigenesis and metastasis. Understanding these concepts will progressively support the development of new strategies to treat human cancer.
Collapse
Affiliation(s)
- Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Corresponding author. (R.J.D.); (N.S.C.)
| | - Navdeep S. Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Corresponding author. (R.J.D.); (N.S.C.)
| |
Collapse
|
531
|
Nguyen HD, Chatterjee S, Haarberg KMK, Wu Y, Bastian D, Heinrichs J, Fu J, Daenthanasanmak A, Schutt S, Shrestha S, Liu C, Wang H, Chi H, Mehrotra S, Yu XZ. Metabolic reprogramming of alloantigen-activated T cells after hematopoietic cell transplantation. J Clin Invest 2016; 126:1337-52. [PMID: 26950421 DOI: 10.1172/jci82587] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 01/21/2016] [Indexed: 12/13/2022] Open
Abstract
Alloreactive donor T cells are the driving force in the induction of graft-versus-host disease (GVHD), yet little is known about T cell metabolism in response to alloantigens after hematopoietic cell transplantation (HCT). Here, we have demonstrated that donor T cells undergo metabolic reprograming after allogeneic HCT. Specifically, we employed a murine allogeneic BM transplant model and determined that T cells switch from fatty acid β-oxidation (FAO) and pyruvate oxidation via the tricarboxylic (TCA) cycle to aerobic glycolysis, thereby increasing dependence upon glutaminolysis and the pentose phosphate pathway. Glycolysis was required for optimal function of alloantigen-activated T cells and induction of GVHD, as inhibition of glycolysis by targeting mTORC1 or 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) ameliorated GVHD mortality and morbidity. Together, our results indicate that donor T cells use glycolysis as the predominant metabolic process after allogeneic HCT and suggest that glycolysis has potential as a therapeutic target for the control of GVHD.
Collapse
|
532
|
Haas R, Cucchi D, Smith J, Pucino V, Macdougall CE, Mauro C. Intermediates of Metabolism: From Bystanders to Signalling Molecules. Trends Biochem Sci 2016; 41:460-471. [PMID: 26935843 DOI: 10.1016/j.tibs.2016.02.003] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/27/2016] [Accepted: 02/04/2016] [Indexed: 11/18/2022]
Abstract
The integration of biochemistry into immune cell biology has contributed immensely to our understanding of immune cell function and the associated pathologies. So far, most studies have focused on the regulation of metabolic pathways during an immune response and their contribution to its success. More recently, novel signalling functions of metabolic intermediates are being discovered that might play important roles in the regulation of immunity. Here we describe the three long-known small metabolites lactate, acetyl-CoA, and succinate in the context of immunometabolic signalling. Functions of these ubiquitous molecules are largely dependent on their intra- and extracellular concentrations as well as their subcompartmental localisation. Importantly, the signalling functions of these metabolic intermediates extend beyond self-regulatory roles and include cell-to-cell communication and sensing of microenvironmental conditions to elicit stress responses and cellular adaptation.
Collapse
Affiliation(s)
- Robert Haas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Danilo Cucchi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK; Istituto Pasteur, Fondazione Cenci Bolognetti, Rome, Italy
| | - Joanne Smith
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Valentina Pucino
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK; Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | | | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK.
| |
Collapse
|
533
|
Norata GD, Caligiuri G, Chavakis T, Matarese G, Netea MG, Nicoletti A, O'Neill LAJ, Marelli-Berg FM. The Cellular and Molecular Basis of Translational Immunometabolism. Immunity 2016; 43:421-34. [PMID: 26377896 DOI: 10.1016/j.immuni.2015.08.023] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Indexed: 12/11/2022]
Abstract
The immune response requires major changes to metabolic processes, and indeed, energy metabolism and functional activation are fully integrated in immune cells to determine their ability to divide, differentiate, and carry out effector functions. Immune cell metabolism has therefore become an attractive target area for therapeutic purposes. A neglected aspect in the translation of immunometabolism is the critical connection between systemic and cellular metabolism. Here, we discuss the importance of understanding and manipulating the integration of systemic and immune cell metabolism through in-depth analysis of immune cell phenotype and function in human metabolic diseases and, in parallel, of the effects of conventional metabolic drugs on immune cell differentiation and function. We examine how the recent identification of selective metabolic programs operating in distinct immune cell subsets and functions has the potential to deliver tools for cell- and function-specific immunometabolic targeting.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, 20092 Milan, Italy.
| | - Giuseppina Caligiuri
- Unité 1148, INSERM, Hôpital X Bichat, 75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France; Département Hospitalo-Universitaire "FIRE," 75018 Paris, France
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Baronissi, 84081 Salerno, Italy; IRCCS MultiMedica, 20138 Milan, Italy
| | - Mihai Gheorge Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Antonino Nicoletti
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
534
|
Maggioli E, McArthur S, Mauro C, Kieswich J, Kusters DHM, Reutelingsperger CPM, Yaqoob M, Solito E. Estrogen protects the blood-brain barrier from inflammation-induced disruption and increased lymphocyte trafficking. Brain Behav Immun 2016; 51:212-222. [PMID: 26321046 DOI: 10.1016/j.bbi.2015.08.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/19/2015] [Accepted: 08/27/2015] [Indexed: 12/11/2022] Open
Abstract
Sex differences have been widely reported in neuroinflammatory disorders, focusing on the contributory role of estrogen. The microvascular endothelium of the brain is a critical component of the blood-brain barrier (BBB) and it is recognized as a major interface for communication between the periphery and the brain. As such, the cerebral capillary endothelium represents an important target for the peripheral estrogen neuroprotective functions, leading us to hypothesize that estrogen can limit BBB breakdown following the onset of peripheral inflammation. Comparison of male and female murine responses to peripheral LPS challenge revealed a short-term inflammation-induced deficit in BBB integrity in males that was not apparent in young females, but was notable in older, reproductively senescent females. Importantly, ovariectomy and hence estrogen loss recapitulated an aged phenotype in young females, which was reversible upon estradiol replacement. Using a well-established model of human cerebrovascular endothelial cells we investigated the effects of estradiol upon key barrier features, namely paracellular permeability, transendothelial electrical resistance, tight junction integrity and lymphocyte transmigration under basal and inflammatory conditions, modeled by treatment with TNFα and IFNγ. In all cases estradiol prevented inflammation-induced defects in barrier function, action mediated in large part through up-regulation of the central coordinator of tight junction integrity, annexin A1. The key role of this protein was then further confirmed in studies of human or murine annexin A1 genetic ablation models. Together, our data provide novel mechanisms for the protective effects of estrogen, and enhance our understanding of the beneficial role it plays in neurovascular/neuroimmune disease.
Collapse
Affiliation(s)
- E Maggioli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - S McArthur
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; Department of Biomedical Sciences, Faculty of Science & Technology, University of Westminster, New Cavendish Street, London W1W 6UW, UK
| | - C Mauro
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - J Kieswich
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - D H M Kusters
- Cardiovascular Research Institute, Department of Biochemistry, Maastricht University, 6200 Maastricht, The Netherlands; Department of Pathology, University of Michigan Health System, 109 Zina Pitcher Place, 4062 BSRB, Ann Arbor, MI 48109-2200, United States
| | - C P M Reutelingsperger
- Cardiovascular Research Institute, Department of Biochemistry, Maastricht University, 6200 Maastricht, The Netherlands
| | - M Yaqoob
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - E Solito
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
535
|
Patsoukis N, Bardhan K, Weaver J, Herbel C, Seth P, Li L, Boussiotis VA. The role of metabolic reprogramming in T cell fate and function. CURRENT TRENDS IN IMMUNOLOGY 2016; 17:1-12. [PMID: 28356677 PMCID: PMC5367635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
T lymphocytes undergo extensive changes in their metabolic properties during their transition through various differentiation states, from naïve to effector to memory or regulatory roles. The cause and effect relationship between metabolism and differentiation is a field of intense investigation. Many recent studies demonstrate the dependency of T cell functional outcomes on metabolic pathways and the possibility of metabolic intervention to modify these functions. In this review, we describe the basic metabolic features of T cells and new findings on how these correlate with various differentiation fates and functions. We also highlight the latest information regarding the main factors that affect T cell metabolic reprogramming.
Collapse
Affiliation(s)
- Nikolaos Patsoukis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| | - Kankana Bardhan
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| | - Jessica Weaver
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| | - Christoph Herbel
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| | - Pankaj Seth
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Lequn Li
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| | - Vassiliki A. Boussiotis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|