201
|
Huang L, Wang C, Xu H, Peng G. Targeting citrate as a novel therapeutic strategy in cancer treatment. Biochim Biophys Acta Rev Cancer 2019; 1873:188332. [PMID: 31751601 DOI: 10.1016/j.bbcan.2019.188332] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 01/09/2023]
Abstract
An important feature shared by many cancer cells is drastically altered metabolism that is critical for rapid growth and proliferation. The distinctly reprogrammed metabolism in cancer cells makes it possible to manipulate the levels of metabolites for cancer treatment. Citrate is a key metabolite that bridges many important metabolic pathways. Recent studies indicate that manipulating the level of citrate can impact the behaviors of both cancer and immune cells, resulting in induction of cancer cell apoptosis, boosting immune responses, and enhanced cancer immunotherapy. In this review, we discuss the recent developments in this emerging area of targeting citrate in cancer treatment. Specifically, we summarize the molecular basis of altered citrate metabolism in both tumors and immune cells, explore the seemingly conflicted growth promoting and growth inhibiting roles of citrate in various tumors, discuss the use of citrate in the clinic as a novel biomarker for cancer progression and outcomes, and highlight the new development of combining citrate with other therapeutic strategies in cancer therapy. An improved understanding of complex roles of citrate in the suppressive tumor microenvironment should open new avenues for cancer therapy.
Collapse
Affiliation(s)
- Lan Huang
- Division of Infectious Diseases, Allergy & Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA; Department of Immunology, Jiangsu University School of Medicine, Zhenjiang 212013, PR China
| | - Cindy Wang
- Division of Infectious Diseases, Allergy & Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Huaxi Xu
- Department of Immunology, Jiangsu University School of Medicine, Zhenjiang 212013, PR China
| | - Guangyong Peng
- Division of Infectious Diseases, Allergy & Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA.
| |
Collapse
|
202
|
Quinn KM, Palchaudhuri R, Palmer CS, La Gruta NL. The clock is ticking: the impact of ageing on T cell metabolism. Clin Transl Immunology 2019; 8:e01091. [PMID: 31832191 PMCID: PMC6859487 DOI: 10.1002/cti2.1091] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/26/2019] [Accepted: 10/27/2019] [Indexed: 12/13/2022] Open
Abstract
It is now clear that access to specific metabolic programmes controls the survival and function of various immune cell populations, including T cells. Efficient naïve and memory T cell homoeostasis requires the use of specific metabolic pathways and differentiation requires rapid and dramatic metabolic remodelling. While we are beginning to appreciate the crucial role of metabolic programming during normal T cell physiology, many of the potential impacts of ageing on metabolic homoeostasis and remodelling in T cells remain unexplored. This review will outline our current understanding of T cell metabolism and explore age‐related metabolic changes that are postulated or have been demonstrated to impact T cell function.
Collapse
Affiliation(s)
- Kylie M Quinn
- School of Health and Biomedical Sciences RMIT University Bundoora VIC Australia.,Department of Biochemistry Biomedicine Discovery Institute Monash University Clayton VIC Australia
| | - Riya Palchaudhuri
- Life Sciences Macfarlane Burnet Institute for Medical Research and Public Health Melbourne VIC Australia.,Department of Infectious Diseases Monash University Melbourne VIC Australia.,Department of Immunology and Pathology Monash University Melbourne VIC Australia
| | - Clovis S Palmer
- Life Sciences Macfarlane Burnet Institute for Medical Research and Public Health Melbourne VIC Australia.,Department of Infectious Diseases Monash University Melbourne VIC Australia
| | - Nicole L La Gruta
- Department of Biochemistry Biomedicine Discovery Institute Monash University Clayton VIC Australia
| |
Collapse
|
203
|
Koutroulis I, Batabyal R, McNamara B, Ledda M, Hoptay C, Freishtat RJ. Sepsis Immunometabolism: From Defining Sepsis to Understanding How Energy Production Affects Immune Response. Crit Care Explor 2019; 1:e0061. [PMID: 32166242 PMCID: PMC7063962 DOI: 10.1097/cce.0000000000000061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES This review will examine current definitions and trends in sepsis management as well pathophysiologic mechanisms in animal and ex vivo studies that correlate decreased energy production with deranged inflammatory response during the septic process. DATA SOURCES The latest articles in the literature that focus on the role of immunometabolism and associated mechanisms in sepsis were selected. STUDY SELECTION The most relevant, original articles were included in the review. DATA EXTRACTION All pertinent data for sepsis definitions as well as changes in immunometabolic pathways during the septic process was reviewed and assessed for inclusion in this article. DATA SYNTHESIS Sepsis is a major cause of multiple organ dysfunction. It is the principal cause of death resulting from infection and one of the most expensive conditions treated in the United States. Despite current efforts to accurately define sepsis, novel treatments and highly trained providers, mortality rates for sepsis remain high, prompting a need for further investigation of underlying immunometabolic mechanisms to identify potential treatment targets. The definition of sepsis has shifted and changed in the past few decades due to poorly defined criteria, as well as unclear guidelines for providers with regards to management of severe sepsis and septic shock. The early identification of patients with a systemic inflammatory response that will progress to septic shock is critical since recent traditional therapeutic approaches, such as early goal-directed therapy, IV immunoglobulin, and anti-tumor necrosis factor-α antibodies have failed. CONCLUSIONS There are no effective anti-sepsis drug therapies due to complex inflammatory and metabolic interactions. Further studies regarding the interface between innate immunity and metabolism should be investigated to effectively address septic patient mortality rates.
Collapse
Affiliation(s)
- Ioannis Koutroulis
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Rachael Batabyal
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Brittany McNamara
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Matthew Ledda
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
| | - Claire Hoptay
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Robert J Freishtat
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
204
|
Cui J, Zhu D, Su M, Tan D, Zhang X, Jia M, Chen G. The combined use of 1 H and 2D NMR-based metabolomics and chemometrics for non-targeted screening of biomarkers and identification of reconstituted milk. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:6455-6461. [PMID: 31294826 DOI: 10.1002/jsfa.9924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/29/2019] [Accepted: 07/09/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The illegal undeclared addition of reconstituted milk powder to ultra-heat treated (UHT) milk to lower production costs is an example of economically motivated adulteration. This activity not only defrauds consumers but also places honest traders at a disadvantage, which could damage the reputation of milk producers and reduce the integrity of the markets. In this research, a non-targeted analytical strategy that combines proton (1 H) nuclear magnetic resonance (NMR) spectroscopy with a chemometrics data mining tool was developed for the authentication of bovine UHT milk. RESULTS Unsupervised principal component analysis was used to distinguish UHT and tap-water-reconstituted powdered milk. Partial least squares-discriminant analysis (PLS-DA) with R2 (Y) and Q2 equal to 0.859 and 0.748, respectively, was used to differentiate UHT and reconstituted milk samples. Three compounds were selected as biomarkers to distinguish UHT and reconstituted milk and identified according to the standard NMR-spectra database. Finally, a PLS-DA model was established, according to the characteristic spectral bands, to identify UHT milk and reconstituted milk. CONCLUSION This procedure demonstrated the feasibility of using non-targeted NMR profiling combined with chemometric analysis to combat mislabeling and fraudulent practices in milk production. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jing Cui
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Dan Zhu
- Chemistry Department, University of Otago, Dunedin, New Zealand
| | - Meicheng Su
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Dongfei Tan
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xia Zhang
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Man Jia
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Gang Chen
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|
205
|
Munford H, Dimeloe S. Intrinsic and Extrinsic Determinants of T Cell Metabolism in Health and Disease. Front Mol Biosci 2019; 6:118. [PMID: 31709265 PMCID: PMC6823819 DOI: 10.3389/fmolb.2019.00118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/14/2019] [Indexed: 01/05/2023] Open
Abstract
T lymphocytes are a critical component of the adaptive immune system, with key roles in the immune response to infection and cancer. Their activity is fundamentally underpinned by dynamic, regulated changes in their metabolism. This ensures adequate availability of energy and biosynthetic precursors for clonal expansion and effector function, and also directly regulates cell signaling, gene transcription, and protein translation. In health, distinct T cells subtypes demonstrate differences in intrinsic metabolic capacity which correlate with their specialized immune functions. In disease, T cells with impaired immune function appear to be likewise metabolically impaired. Furthermore, diseased tissue environments-through inadequate provision of nutrients and oxygen, or accumulation of metabolic intermediates, end-products, and cytokines- can impose metabolic insufficiency upon these cells, and further compound intrinsic impairments. These intrinsic and extrinsic determinants of T cell metabolism and their potential compound effects, together with the mechanisms involved form the subject of this review. We will also discuss how dysfunctional metabolic pathways may be therapeutically targeted to restore normal T cell function in disease.
Collapse
Affiliation(s)
- Haydn Munford
- Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
206
|
Metabolic Profiling Using Stable Isotope Tracing Reveals Distinct Patterns of Glucose Utilization by Physiologically Activated CD8 + T Cells. Immunity 2019; 51:856-870.e5. [PMID: 31747582 DOI: 10.1016/j.immuni.2019.09.003] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/11/2019] [Accepted: 09/06/2019] [Indexed: 01/06/2023]
Abstract
Naive CD8+ T cells differentiating into effector T cells increase glucose uptake and shift from quiescent to anabolic metabolism. Although much is known about the metabolism of cultured T cells, how T cells use nutrients during immune responses in vivo is less well defined. Here, we combined bioenergetic profiling and 13C-glucose infusion techniques to investigate the metabolism of CD8+ T cells responding to Listeria infection. In contrast to in vitro-activated T cells, which display hallmarks of Warburg metabolism, physiologically activated CD8+ T cells displayed greater rates of oxidative metabolism, higher bioenergetic capacity, differential use of pyruvate, and prominent flow of 13C-glucose carbon to anabolic pathways, including nucleotide and serine biosynthesis. Glucose-dependent serine biosynthesis mediated by the enzyme Phgdh was essential for CD8+ T cell expansion in vivo. Our data highlight fundamental differences in glucose use by pathogen-specific T cells in vivo, illustrating the impact of environment on T cell metabolic phenotypes.
Collapse
|
207
|
Li C, Zhu B, Son Y, Wang Z, Jiang L, Xiang M, Ye Z, Beckermann KE, Wu Y, Jenkins J, Siska PJ, Vincent BG, Prakash Y, Peikert T, Edelson BT, Taneja R, Kaplan MH, Rathmell JC, Dong H, Hitosugi T, Sun J. The Transcription Factor Bhlhe40 Programs Mitochondrial Regulation of Resident CD8 + T Cell Fitness and Functionality. Immunity 2019; 51:491-507.e7. [PMID: 31533057 PMCID: PMC6903704 DOI: 10.1016/j.immuni.2019.08.013] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 04/13/2019] [Accepted: 08/14/2019] [Indexed: 12/28/2022]
Abstract
Tissue-resident memory CD8+ T (Trm) cells share core residency gene programs with tumor-infiltrating lymphocytes (TILs). However, the transcriptional, metabolic, and epigenetic regulation of Trm cell and TIL development and function is largely undefined. Here, we found that the transcription factor Bhlhe40 was specifically required for Trm cell and TIL development and polyfunctionality. Local PD-1 signaling inhibited TIL Bhlhe40 expression, and Bhlhe40 was critical for TIL reinvigoration following anti-PD-L1 blockade. Mechanistically, Bhlhe40 sustained Trm cell and TIL mitochondrial fitness and a functional epigenetic state. Building on these findings, we identified an epigenetic and metabolic regimen that promoted Trm cell and TIL gene signatures associated with tissue residency and polyfunctionality. This regimen empowered the anti-tumor activity of CD8+ T cells and possessed therapeutic potential even at an advanced tumor stage in mouse models. Our results provide mechanistic insights into the local regulation of Trm cell and TIL function.
Collapse
Affiliation(s)
- Chaofan Li
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Bibo Zhu
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Youngmin Son
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Zheng Wang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Jiang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Min Xiang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhenqing Ye
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Kathryn E. Beckermann
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yue Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - James Jenkins
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter J. Siska
- Internal Medicine III, University Hospital Regensburg, 93042 Regensburg, Germany,Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Y.S. Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tobias Peikert
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian T. Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reshma Taneja
- Department of Physiology, National University of Singapore, Singapore 117593
| | - Mark H. Kaplan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey C. Rathmell
- Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
208
|
Mondragón L, Mhaidly R, De Donatis GM, Tosolini M, Dao P, Martin AR, Pons C, Chiche J, Jacquin M, Imbert V, Proïcs E, Boyer L, Doye A, Luciano F, Neels JG, Coutant F, Fabien N, Sormani L, Rubio-Patiño C, Bossowski JP, Muller F, Marchetti S, Villa E, Peyron JF, Gaulard P, Lemonnier F, Asnafi V, Genestier L, Benhida R, Fournié JJ, Passeron T, Ricci JE, Verhoeyen E. GAPDH Overexpression in the T Cell Lineage Promotes Angioimmunoblastic T Cell Lymphoma through an NF-κB-Dependent Mechanism. Cancer Cell 2019; 36:268-287.e10. [PMID: 31447347 DOI: 10.1016/j.ccell.2019.07.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 04/17/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
GAPDH is emerging as a key player in T cell development and function. To investigate the role of GAPDH in T cells, we generated a transgenic mouse model overexpressing GAPDH in the T cell lineage. Aged mice developed a peripheral Tfh-like lymphoma that recapitulated key molecular, pathological, and immunophenotypic features of human angioimmunoblastic T cell lymphoma (AITL). GAPDH induced non-canonical NF-κB pathway activation in mouse T cells, which was strongly activated in human AITL. We developed a NIK inhibitor to reveal that targeting the NF-κB pathway prolonged AITL-bearing mouse survival alone and in combination with anti-PD-1. These findings suggest the therapeutic potential of targeting NF-κB signaling in AITL and provide a model for future AITL therapeutic investigations.
Collapse
Affiliation(s)
| | - Rana Mhaidly
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Marie Tosolini
- Pôle Technologique du CRCT - Plateau Bioinformatique INSERM-UMR 1037, Toulouse, France
| | - Pascal Dao
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Anthony R Martin
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Caroline Pons
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Marie Jacquin
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Emma Proïcs
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Laurent Boyer
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Anne Doye
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Jaap G Neels
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | - Frédéric Coutant
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon, Edouard Herriot Hospital, Lyon, France
| | - Nicole Fabien
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Laura Sormani
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | | | | | | | - Elodie Villa
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France
| | | | - Philippe Gaulard
- Université Paris-Est Créteil, Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France; Département de Pathologie, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Créteil, France
| | - François Lemonnier
- Université Paris-Est Créteil, Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Créteil, France
| | - Vahid Asnafi
- Université Paris 5, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker-Enfants Malades, Paris, France
| | - Laurent Genestier
- CRCL, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon I, 69921 Oullins Cedex, France
| | - Rachid Benhida
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice, France
| | - Jean-Jacques Fournié
- CRCT, INSERM U1037 - Université Paul Sabatier - CNRS ERL5294, Université de Toulouse, Laboratoire d'Excellence TOUCAN, Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France; IUCT, 31037 Toulouse, France
| | - Thierry Passeron
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France; Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Dermatology, 06204 Nice, France
| | | | - Els Verhoeyen
- Université Côte d'Azur, INSERM, C3M, 06204 Nice, France; CIRI, Université de Lyon, INSERM U1111, ENS de Lyon, Université Lyon 1, CNRS, UMR 5308, 69007 Lyon, France.
| |
Collapse
|
209
|
Bose S, Ramesh V, Locasale JW. Acetate Metabolism in Physiology, Cancer, and Beyond. Trends Cell Biol 2019; 29:695-703. [PMID: 31160120 PMCID: PMC6699882 DOI: 10.1016/j.tcb.2019.05.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/20/2022]
Abstract
Acetate and the related metabolism of acetyl-coenzyme A (acetyl-CoA) confer numerous metabolic functions, including energy production, lipid synthesis, and protein acetylation. Despite its importance as a nutrient for cellular metabolism, its source has been unclear. Recent studies have provided evidence to support the existence of a de novo pathway for acetate production derived from pyruvate, the end product of glycolysis. This mechanism of pyruvate-derived acetate generation could have far-reaching implications for the regulation of central carbon metabolism. In this Opinion, we discuss our current understanding of acetate metabolism in the context of cell-autonomous metabolic regulation, cell-cell interactions, and systemic physiology. Applications relevant to health and disease, particularly cancer, are emphasized.
Collapse
Affiliation(s)
- Shree Bose
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Vijyendra Ramesh
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
210
|
Vodnala SK, Eil R, Kishton RJ, Sukumar M, Yamamoto TN, Ha NH, Lee PH, Shin M, Patel SJ, Yu Z, Palmer DC, Kruhlak MJ, Liu X, Locasale JW, Huang J, Roychoudhuri R, Finkel T, Klebanoff CA, Restifo NP. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 2019; 363:363/6434/eaau0135. [PMID: 30923193 DOI: 10.1126/science.aau0135] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 11/06/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022]
Abstract
A paradox of tumor immunology is that tumor-infiltrating lymphocytes are dysfunctional in situ, yet are capable of stem cell-like behavior including self-renewal, expansion, and multipotency, resulting in the eradication of large metastatic tumors. We find that the overabundance of potassium in the tumor microenvironment underlies this dichotomy, triggering suppression of T cell effector function while preserving stemness. High levels of extracellular potassium constrain T cell effector programs by limiting nutrient uptake, thereby inducing autophagy and reduction of histone acetylation at effector and exhaustion loci, which in turn produces CD8+ T cells with improved in vivo persistence, multipotency, and tumor clearance. This mechanistic knowledge advances our understanding of T cell dysfunction and may lead to novel approaches that enable the development of enhanced T cell strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Suman Kumar Vodnala
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Robert Eil
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rigel J Kishton
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Madhusudhanan Sukumar
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Tori N Yamamoto
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ngoc-Han Ha
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ping-Hsien Lee
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - MinHwa Shin
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shashank J Patel
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Zhiya Yu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Douglas C Palmer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| | - Michael J Kruhlak
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Xiaojing Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jing Huang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Toren Finkel
- Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Christopher A Klebanoff
- Parker Institute for Cancer Immunotherapy, New York, NY 10065, USA.,Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Nicholas P Restifo
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA. .,Center for Cell-Based Therapy, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
211
|
Cui J, Lian Y, Zhao C, Du H, Han Y, Gao W, Xiao H, Zheng J. Dietary Fibers from Fruits and Vegetables and Their Health Benefits via Modulation of Gut Microbiota. Compr Rev Food Sci Food Saf 2019; 18:1514-1532. [DOI: 10.1111/1541-4337.12489] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Jiefen Cui
- Inst. of Food Science and TechnologyChinese Academy of Agricultural Sciences Beijing 100193 China
| | - Yunhe Lian
- Research and Development Dept.Chenguang Biotech Group Co., Ltd. Hebei 057250 China
| | - Chengying Zhao
- Inst. of Food Science and TechnologyChinese Academy of Agricultural Sciences Beijing 100193 China
| | - Hengjun Du
- Dept. of Food ScienceUniv. of Massachusetts Amherst MA 01003 U.S.A
| | - Yanhui Han
- Dept. of Food ScienceUniv. of Massachusetts Amherst MA 01003 U.S.A
| | - Wei Gao
- Research and Development Dept.Chenguang Biotech Group Co., Ltd. Hebei 057250 China
| | - Hang Xiao
- Dept. of Food ScienceUniv. of Massachusetts Amherst MA 01003 U.S.A
| | - Jinkai Zheng
- Inst. of Food Science and TechnologyChinese Academy of Agricultural Sciences Beijing 100193 China
| |
Collapse
|
212
|
The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients 2019; 11:nu11081943. [PMID: 31426593 PMCID: PMC6723943 DOI: 10.3390/nu11081943] [Citation(s) in RCA: 348] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
The interplay of gut microbiota, host metabolism, and metabolic health has gained increased attention. Gut microbiota may play a regulatory role in gastrointestinal health, substrate metabolism, and peripheral tissues including adipose tissue, skeletal muscle, liver, and pancreas via its metabolites short-chain fatty acids (SCFA). Animal and human data demonstrated that, in particular, acetate beneficially affects host energy and substrate metabolism via secretion of the gut hormones like glucagon-like peptide-1 and peptide YY, which, thereby, affects appetite, via a reduction in whole-body lipolysis, systemic pro-inflammatory cytokine levels, and via an increase in energy expenditure and fat oxidation. Thus, potential therapies to increase gut microbial fermentation and acetate production have been under vigorous scientific scrutiny. In this review, the relevance of the colonically and systemically most abundant SCFA acetate and its effects on the previously mentioned tissues will be discussed in relation to body weight control and glucose homeostasis. We discuss in detail the differential effects of oral acetate administration (vinegar intake), colonic acetate infusions, acetogenic fiber, and acetogenic probiotic administrations as approaches to combat obesity and comorbidities. Notably, human data are scarce, which highlights the necessity for further human research to investigate acetate’s role in host physiology, metabolic, and cardiovascular health.
Collapse
|
213
|
Riera-Domingo C, Audigé A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone M. Immunity, Hypoxia, and Metabolism-the Ménage à Trois of Cancer: Implications for Immunotherapy. Physiol Rev 2019; 100:1-102. [PMID: 31414610 DOI: 10.1152/physrev.00018.2019] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is generally accepted that metabolism is able to shape the immune response. Only recently we are gaining awareness that the metabolic crosstalk between different tumor compartments strongly contributes to the harsh tumor microenvironment (TME) and ultimately impairs immune cell fitness and effector functions. The major aims of this review are to provide an overview on the immune system in cancer; to position oxygen shortage and metabolic competition as the ground of a restrictive TME and as important players in the anti-tumor immune response; to define how immunotherapies affect hypoxia/oxygen delivery and the metabolic landscape of the tumor; and vice versa, how oxygen and metabolites within the TME impinge on the success of immunotherapies. By analyzing preclinical and clinical endeavors, we will discuss how a metabolic characterization of the TME can identify novel targets and signatures that could be exploited in combination with standard immunotherapies and can help to predict the benefit of new and traditional immunotherapeutic drugs.
Collapse
Affiliation(s)
- Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Annette Audigé
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Sara Granja
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Wan-Chen Cheng
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Ping-Chih Ho
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Fátima Baltazar
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Christian Stockmann
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| |
Collapse
|
214
|
Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol 2019; 20:55-70. [DOI: 10.1038/s41577-019-0203-y] [Citation(s) in RCA: 451] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
|
215
|
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: 562] [Impact Index Per Article: 93.7] [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
|
216
|
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: 183] [Impact Index Per Article: 30.5] [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
|
217
|
Bachem A, Makhlouf C, Binger KJ, de Souza DP, Tull D, Hochheiser K, Whitney PG, Fernandez-Ruiz D, Dähling S, Kastenmüller W, Jönsson J, Gressier E, Lew AM, Perdomo C, Kupz A, Figgett W, Mackay F, Oleshansky M, Russ BE, Parish IA, Kallies A, McConville MJ, Turner SJ, Gebhardt T, Bedoui S. Microbiota-Derived Short-Chain Fatty Acids Promote the Memory Potential of Antigen-Activated CD8 + T Cells. Immunity 2019; 51:285-297.e5. [PMID: 31272808 DOI: 10.1016/j.immuni.2019.06.002] [Citation(s) in RCA: 441] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 03/01/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022]
Abstract
Interactions with the microbiota influence many aspects of immunity, including immune cell development, differentiation, and function. Here, we examined the impact of the microbiota on CD8+ T cell memory. Antigen-activated CD8+ T cells transferred into germ-free mice failed to transition into long-lived memory cells and had transcriptional impairments in core genes associated with oxidative metabolism. The microbiota-derived short-chain fatty acid (SCFA) butyrate promoted cellular metabolism, enhanced memory potential of activated CD8+ T cells, and SCFAs were required for optimal recall responses upon antigen re-encounter. Mechanistic experiments revealed that butyrate uncoupled the tricarboxylic acid cycle from glycolytic input in CD8+ T cells, which allowed preferential fueling of oxidative phosphorylation through sustained glutamine utilization and fatty acid catabolism. Our findings reveal a role for the microbiota in promoting CD8+ T cell long-term survival as memory cells and suggest that microbial metabolites guide the metabolic rewiring of activated CD8+ T cells to enable this transition.
Collapse
Affiliation(s)
- Annabell Bachem
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christina Makhlouf
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katrina J Binger
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - David P de Souza
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Deidra Tull
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katharina Hochheiser
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Paul G Whitney
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sabrina Dähling
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Johanna Jönsson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Elise Gressier
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew M Lew
- Immunology Division, Walter and Eliza Hall Institute for Medical Research, Parkville, VIC 3010, Australia
| | - Carolina Perdomo
- Department of Immunology, Max-Planck Institute for Infection Biology, Berlin, Germany
| | - Andreas Kupz
- Department of Immunology, Max-Planck Institute for Infection Biology, Berlin, Germany; Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - William Figgett
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Fabienne Mackay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Moshe Oleshansky
- Department of Microbiology, Monash University, Clayton, VIC 3800, Australia
| | - Brendan E Russ
- Department of Microbiology, Monash University, Clayton, VIC 3800, Australia
| | - Ian A Parish
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephen J Turner
- Department of Microbiology, Monash University, Clayton, VIC 3800, Australia
| | - Thomas Gebhardt
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
218
|
O'Sullivan D. The metabolic spectrum of memory T cells. Immunol Cell Biol 2019; 97:636-646. [DOI: 10.1111/imcb.12274] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022]
Affiliation(s)
- David O'Sullivan
- Department of Immunometabolism Max Planck Institute of Immunobiology and Epigenetics Freiburg Germany
| |
Collapse
|
219
|
Kumar A, Smith C, Jobin C, Trinchieri G, Howcroft TK, Seifried H, Espey MG, Flores R, Kim YS, Daschner PJ. Workshop Report: Modulation of Antitumor Immune Responses by Dietary and Microbial Metabolites. J Natl Cancer Inst 2019; 109:3806188. [PMID: 30053241 DOI: 10.1093/jnci/djx040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/22/2017] [Indexed: 12/13/2022] Open
Abstract
The human microbiota maintains an enormous and diverse capacity to produce a diet-dependent metabolome that impacts both host tissue and microbial community homeostasis. Recent discoveries support a growing appreciation that microbial metabolites derived from bioactive foods are also important regulators of host immune and metabolic functions. To gain a better understanding of the current evidence for the roles of dietary and microbial metabolites in tumor immunity, the Division of Cancer Biology and the Division of Cancer Prevention, National Cancer Institute, cosponsored a workshop on August 31 and September 1, 2016, in Bethesda, Maryland. Workshop participants examined several lines of converging science that link nutrition, microbiology, and tumor immunology and identified key concepts and research opportunities that will accelerate our understanding of these interactions. In addition, the participants identified some of the critical gaps and research challenges that could be addressed through interdisciplinary collaborations, including future opportunities for translating new information into novel cancer prevention and treatment strategies based on targeting host immune functions that are altered by metabolite sensing pathways.
Collapse
Affiliation(s)
- Amit Kumar
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Carolyne Smith
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Christian Jobin
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Giorgio Trinchieri
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - T Kevin Howcroft
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Harold Seifried
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Michael Graham Espey
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Roberto Flores
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Young S Kim
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Phillip J Daschner
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| |
Collapse
|
220
|
Luci C, Vieira E, Perchet T, Gual P, Golub R. Natural Killer Cells and Type 1 Innate Lymphoid Cells Are New Actors in Non-alcoholic Fatty Liver Disease. Front Immunol 2019; 10:1192. [PMID: 31191550 PMCID: PMC6546848 DOI: 10.3389/fimmu.2019.01192] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity and associated liver diseases (Non Alcoholic Fatty Liver Disease, NAFLD) are a major public health problem with increasing incidence in Western countries (25% of the affected population). These complications develop from a fatty liver (steatosis) to an inflammatory state (steatohepatitis) evolving toward fibrosis and hepatocellular carcinoma. Lipid accumulation in the liver contributes to hepatocyte cell death and promotes liver injury. Local immune cells are activated either by Danger Associated Molecular Patterns (DAMPS) released by dead hepatocytes or by bacterial products (PAMPS) reaching the liver due to increased intestinal permeability. The resulting low-grade inflammatory state promotes the progression of liver complications toward more severe grades. Innate lymphoid cells (ILC) are an heterogeneous family of five subsets including circulating Natural Killer (NK) cells, ILC1, ILC2, ILC3, and lymphocytes tissue-inducer cells (LTi). NK cells and tissue-resident ILCs, mainly located at epithelial surfaces, are prompt to rapidly react to environmental changes to mount appropriate immune responses. Recent works have demonstrated the interplay between ILCs subsets and the environment within metabolic active organs such as liver, adipose tissue and gut during diet-induced obesity leading or not to hepatic abnormalities. Here, we provide an overview of the newly roles of NK cells and ILC1 in metabolism focusing on their contribution to the development of NAFLD. We also discuss recent studies that demonstrate the ability of these two subsets to influence tissue-specific metabolism and how their function and homeostasis are affected during metabolic disorders.
Collapse
Affiliation(s)
- Carmelo Luci
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Elodie Vieira
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Thibaut Perchet
- Unité Lymphopoïèse, Institut Pasteur, INSERM U1223, Université Paris Diderot, Paris, France
| | - Philippe Gual
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Rachel Golub
- Unité Lymphopoïèse, Institut Pasteur, INSERM U1223, Université Paris Diderot, Paris, France
| |
Collapse
|
221
|
Wensveen FM, Šestan M, Turk Wensveen T, Polić B. 'Beauty and the beast' in infection: How immune-endocrine interactions regulate systemic metabolism in the context of infection. Eur J Immunol 2019; 49:982-995. [PMID: 31106860 DOI: 10.1002/eji.201847895] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 02/07/2023]
Abstract
The immune and endocrine systems ensure two vital functions in the body. The immune system protects us from lethal pathogens, whereas the endocrine system ensures proper metabolic function of peripheral organs by regulating systemic homeostasis. These two systems were long thought to operate independently. The immune system uses cytokines and immune receptors, whereas the endocrine system uses hormones to regulate metabolism. However, recent findings show that the immune and endocrine systems closely interact, especially regarding regulation of glucose metabolism. In response to pathogen encounter, cytokines modify responsiveness of peripheral organs to endocrine signals, resulting in altered levels of blood hormones such as insulin, which promotes the ability of the body to fight infection. Here we provide an overview of recent literature describing various mechanisms, which the immune system utilizes to modify endocrine regulation of systemic metabolism. Moreover, we will describe how these immune-endocrine interactions derail in the context of obesity. From a clinical perspective we will elaborate how infection and obesity aggravate the development of metabolic diseases such as diabetes mellitus type 2 in humans. In summary, this review provides a comprehensive overview of immune-induced changes in systemic metabolism following infection, with a focus on regulation of glucose metabolism.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Marko Šestan
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Tamara Turk Wensveen
- Department of Endocrinology, Diabetes and Metabolic Diseases, Clinical hospital center Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, University of Rijeka School of Medicine, Rijeka, Croatia
| |
Collapse
|
222
|
Luu M, Visekruna A. Short-chain fatty acids: Bacterial messengers modulating the immunometabolism of T cells. Eur J Immunol 2019; 49:842-848. [PMID: 31054154 DOI: 10.1002/eji.201848009] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/28/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Abstract
Short-chain fatty acids (SCFAs) are mainly generated by bacterial fermentation of non-digestible carbohydrates such as dietary fiber. In the last decade, new investigations have revealed that SCFAs have a very specific function and serve as active microbial metabolites, which are able to modulate the function of immune cells in the intestine and other tissues. Recent studies have highlighted the immunomodulatory potential of SCFAs in several autoimmune and inflammatory disorders such as multiple sclerosis, colitis, type 1 diabetes and rheumatoid arthritis. While the SCFA-mediated activation of GPR41/GPR43 signalling pathways and their inhibitory activity on histone deacetylases have been extensively investigated, the impact of SCFAs on the T cell metabolism is poorly understood. SCFAs induce metabolic alterations in T cells by enhancing the activity of the mTOR complex and by regulating their glucose metabolism. Once taken up into T lymphocytes, SCFA-derived acetyl groups contribute to the cellular acetyl-CoA pool, which influences the histone acetylation and cytokine gene expression. This article reviews how SCFAs modulate the metabolic status of T cells, thereby impacting on epigenetic modifications and T cell function. We will also discuss how the recent findings from SCFA biology might be utilized for potential immune therapies of various autoimmune diseases.
Collapse
Affiliation(s)
- Maik Luu
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
223
|
Sierro F, Grau GER. The Ins and Outs of Cerebral Malaria Pathogenesis: Immunopathology, Extracellular Vesicles, Immunometabolism, and Trained Immunity. Front Immunol 2019; 10:830. [PMID: 31057552 PMCID: PMC6478768 DOI: 10.3389/fimmu.2019.00830] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/28/2019] [Indexed: 12/16/2022] Open
Abstract
Complications from malaria parasite infections still cost the lives of close to half a million people every year. The most severe is cerebral malaria (CM). Employing murine models of CM, autopsy results, in vitro experiments, neuroimaging and microscopic techniques, decades of research activity have investigated the development of CM immunopathology in the hope of identifying steps that could be therapeutically targeted. Yet important questions remain. This review summarizes recent findings, primarily mechanistic insights on the essential cellular and molecular players involved gained within the murine experimental cerebral malaria model. It also highlights recent developments in (a) cell-cell communication events mediated through extracellular vesicles (EVs), (b) mounting evidence for innate immune memory, leading to “trained“ increased or tolerised responses, and (c) modulation of immune cell function through metabolism, that could shed light on why some patients develop this life-threatening condition whilst many do not.
Collapse
Affiliation(s)
- Frederic Sierro
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia.,Human Health, Nuclear Science, Technology, and Landmark Infrastructure, Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Georges E R Grau
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
224
|
Lötscher J, Balmer ML. Sensing between reactions - how the metabolic microenvironment shapes immunity. Clin Exp Immunol 2019; 197:161-169. [PMID: 30868561 DOI: 10.1111/cei.13291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
Perception of potential threat is key for survival. The immune system constantly patrols the organism scanning for potential pathogenic or malignant danger. Recent evidence suggests that immunosurveillance not only relies on classic receptors [e.g. Toll-like receptors (TLRs) or antibodies] but is also based on sensing of the metabolic environment. Metabolites interact in numerous ways with immune cells, and are therefore more than just reaction intermediates. This new perspective opens the door for potential, future therapeutic strategies. Here we describe how immune functionality during infections, cancer or autoimmunity, as exemplified by short-chain fatty acids, lactate and reactive oxygen species (ROS), can be shaped by metabolic intermediates.
Collapse
Affiliation(s)
- J Lötscher
- Department of Biomedicine, Immunobiology, University of Basel, Basel, Switzerland
| | - M L Balmer
- Department of Biomedicine, Immunobiology, University of Basel, Basel, Switzerland
| |
Collapse
|
225
|
The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat Commun 2019; 10:760. [PMID: 30770822 PMCID: PMC6377655 DOI: 10.1038/s41467-019-08711-2] [Citation(s) in RCA: 308] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acids (SCFAs) have immunomodulatory effects, but the underlying mechanisms are not well understood. Here we show that pentanoate, a physiologically abundant SCFA, is a potent regulator of immunometabolism. Pentanoate induces IL-10 production in lymphocytes by reprogramming their metabolic activity towards elevated glucose oxidation. Mechanistically, this reprogramming is mediated by supplying additional pentanoate-originated acetyl-CoA for histone acetyltransferases, and by pentanoate-triggered enhancement of mTOR activity. In experimental mouse models of colitis and multiple sclerosis, pentanoate-induced regulatory B cells mediate protection from autoimmune pathology. Additionally, pentanoate shows a potent histone deacetylase-inhibitory activity in CD4+ T cells, thereby reducing their IL-17A production. In germ-free mice mono-colonized with segmented filamentous bacteria (SFB), pentanoate inhibits the generation of small-intestinal Th17 cells and ameliorates SFB-promoted inflammation in the central nervous system. Taken together, by enhancing IL-10 production and suppressing Th17 cells, the SCFA pentanoate might be of therapeutic relevance for inflammatory and autoimmune diseases. Short-chain fatty acids (SCFAs) have immunomodulatory effects, but the underlying mechanisms are still unclear. Here the authors show that a SCFA, pentanoate, suppresses autoimmune inflammation in mouse models of colitis and multiple sclerosis via epigenetic modulation of immune cell metabolic and functional pathways.
Collapse
|
226
|
Raud B, McGuire PJ, Jones RG, Sparwasser T, Berod L. Fatty acid metabolism in CD8 + T cell memory: Challenging current concepts. Immunol Rev 2019; 283:213-231. [PMID: 29664569 DOI: 10.1111/imr.12655] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD8+ T cells are key members of the adaptive immune response against infections and cancer. As we discuss in this review, these cells can present diverse metabolic requirements, which have been intensely studied during the past few years. Our current understanding suggests that aerobic glycolysis is a hallmark of activated CD8+ T cells, while naive and memory (Tmem ) cells often rely on oxidative phosphorylation, and thus mitochondrial metabolism is a crucial determinant of CD8+ Tmem cell development. Moreover, it has been proposed that CD8+ Tmem cells have a specific requirement for the oxidation of long-chain fatty acids (LC-FAO), a process modulated in lymphocytes by the enzyme CPT1A. However, this notion relies heavily on the metabolic analysis of in vitro cultures and on chemical inhibition of CPT1A. Therefore, we introduce more recent studies using genetic models to demonstrate that CPT1A-mediated LC-FAO is dispensable for the development of CD8+ T cell memory and protective immunity, and question the use of chemical inhibitors to target this enzyme. We discuss insights obtained from those and other studies analyzing the metabolic characteristics of CD8+ Tmem cells, and emphasize how T cells exhibit flexibility in their choice of metabolic fuel.
Collapse
Affiliation(s)
- Brenda Raud
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Peter J McGuire
- Metabolism, Infection, and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Russell G Jones
- Department of Physiology, Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| |
Collapse
|
227
|
Pandey SK, Yadav S, Goel Y, Singh SM. Cytotoxic action of acetate on tumor cells of thymic origin: Role of MCT-1, pH homeostasis and altered cell survival regulation. Biochimie 2019; 157:1-9. [DOI: 10.1016/j.biochi.2018.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023]
|
228
|
Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, Pernot J, Ubags N, Fajas L, Nicod LP, Marsland BJ. Dietary Fiber Confers Protection against Flu by Shaping Ly6c - Patrolling Monocyte Hematopoiesis and CD8 + T Cell Metabolism. Immunity 2019; 48:992-1005.e8. [PMID: 29768180 DOI: 10.1016/j.immuni.2018.04.022] [Citation(s) in RCA: 476] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 02/08/2018] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
Dietary fiber protects against chronic inflammatory diseases by dampening immune responses through short-chain fatty acids (SCFAs). Here we examined the effect of dietary fiber in viral infection, where the anti-inflammatory properties of SCFAs in principle could prevent protective immunity. Instead, we found that fermentable dietary fiber increased survival of influenza-infected mice through two complementary mechanisms. High-fiber diet (HFD)-fed mice exhibited altered bone marrow hematopoiesis, characterized by enhanced generation of Ly6c- patrolling monocytes, which led to increased numbers of alternatively activated macrophages with a limited capacity to produce the chemokine CXCL1 in the airways. Blunted CXCL1 production reduced neutrophil recruitment to the airways, thus limiting tissue immunopathology during infection. In parallel, diet-derived SCFAs boosted CD8+ T cell effector function by enhancing cellular metabolism. Hence, dietary fermentable fiber and SCFAs set an immune equilibrium, balancing innate and adaptive immunity so as to promote the resolution of influenza infection while preventing immune-associated pathology.
Collapse
Affiliation(s)
- Aurélien Trompette
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Eva S Gollwitzer
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland; Global Health Institute, EPFL-SV-GHI Station 19, EPFL, 1015 Lausanne, Switzerland
| | - Céline Pattaroni
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland; Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Isabel C Lopez-Mejia
- Department of Physiology, University of Lausanne, 1011 Lausanne, Switzerland; Center for Integrative Genomics (CIG), University of Lausanne, 1015 Lausanne, Switzerland
| | - Erika Riva
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Julie Pernot
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Niki Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Lluis Fajas
- Department of Physiology, University of Lausanne, 1011 Lausanne, Switzerland; Center for Integrative Genomics (CIG), University of Lausanne, 1015 Lausanne, Switzerland
| | - Laurent P Nicod
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Benjamin J Marsland
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, CLED_02.206, Chemin des Boveresses 155, 1066 Epalinges, Switzerland; Department of Immunology and Pathology, Monash University, Melbourne, Australia.
| |
Collapse
|
229
|
Tracking acetate through a journey of living world: Evolution as alternative cellular fuel with potential for application in cancer therapeutics. Life Sci 2018; 215:86-95. [DOI: 10.1016/j.lfs.2018.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 12/21/2022]
|
230
|
Maimela NR, Liu S, Zhang Y. Fates of CD8+ T cells in Tumor Microenvironment. Comput Struct Biotechnol J 2018; 17:1-13. [PMID: 30581539 PMCID: PMC6297055 DOI: 10.1016/j.csbj.2018.11.004] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/14/2018] [Accepted: 11/18/2018] [Indexed: 12/24/2022] Open
Abstract
Studies have reported a positive correlation between elevated CD8+ T cells in the tumor microenvironment (TME) and good prognosis in cancer. However, the mechanisms linking T cell tumor-infiltration and tumor rejection are yet to be fully understood. The cells and factors of the TME facilitate tumor development in various ways. CD8+ T cell function is influenced by a number of factors, including CD8+ T cell trafficking and localization into tumor sites; as well as CD8+ T cell growth and differentiation. This review highlights recent literature as well as currently evolving concepts regarding the fates of CD8+ T cells in the TME from three different aspects CD8+ T cell trafficking, differentiation and function. A thorough understanding of factors contributing to the fates of CD8+ T cells will allow researchers to develop new strategies and improve on already existing strategies to facilitate CD8+ T cell mediated anti-tumor function, impede T cell dysfunction and modulate the TME into a less immunosuppressive TME.
Collapse
Affiliation(s)
| | - Shasha Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou 450052, China
| |
Collapse
|
231
|
Liu X, Cooper DE, Cluntun AA, Warmoes MO, Zhao S, Reid MA, Liu J, Lund PJ, Lopes M, Garcia BA, Wellen KE, Kirsch DG, Locasale JW. Acetate Production from Glucose and Coupling to Mitochondrial Metabolism in Mammals. Cell 2018; 175:502-513.e13. [PMID: 30245009 PMCID: PMC6173642 DOI: 10.1016/j.cell.2018.08.040] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/22/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022]
Abstract
Acetate is a major nutrient that supports acetyl-coenzyme A (Ac-CoA) metabolism and thus lipogenesis and protein acetylation. However, its source is unclear. Here, we report that pyruvate, the end product of glycolysis and key node in central carbon metabolism, quantitatively generates acetate in mammals. This phenomenon becomes more pronounced in the context of nutritional excess, such as during hyperactive glucose metabolism. Conversion of pyruvate to acetate occurs through two mechanisms: (1) coupling to reactive oxygen species (ROS) and (2) neomorphic enzyme activity from keto acid dehydrogenases that enable function as pyruvate decarboxylases. Further, we demonstrate that de novo acetate production sustains Ac-CoA pools and cell proliferation in limited metabolic environments, such as during mitochondrial dysfunction or ATP citrate lyase (ACLY) deficiency. By virtue of de novo acetate production being coupled to mitochondrial metabolism, there are numerous possible regulatory mechanisms and links to pathophysiology.
Collapse
Affiliation(s)
- Xiaojing Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Daniel E Cooper
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ahmad A Cluntun
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Marc O Warmoes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Steven Zhao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Reid
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA
| | - Peder J Lund
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mariana Lopes
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA; Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
232
|
Regulation of the effector function of CD8 + T cells by gut microbiota-derived metabolite butyrate. Sci Rep 2018; 8:14430. [PMID: 30258117 PMCID: PMC6158259 DOI: 10.1038/s41598-018-32860-x] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota produces metabolites such as short-chain fatty acids (SCFAs) that regulate the energy homeostasis and impact on immune cell function of the host. Recently, innovative approaches based on the oral administration of SCFAs have been discussed for therapeutic modification of inflammatory immune responses in autoimmune diseases. So far, most studies have investigated the SCFA-mediated effects on CD4+ T cells and antigen presenting cells. Here we show that butyrate and, to a lesser degree, propionate directly modulate the gene expression of CD8+ cytotoxic T lymphocytes (CTLs) and Tc17 cells. Increased IFN-γ and granzyme B expression by CTLs as well as the molecular switch of Tc17 cells towards the CTL phenotype was mediated by butyrate independently of its interaction with specific SCFA-receptors GPR41 and GPR43. Our results indicate that butyrate strongly inhibited histone-deacetylases (HDACs) in CD8+ T cells thereby affecting the gene expression of effector molecules. Accordingly, the pan-HDAC inhibitors trichostatin A (TSA) and sodium valproate exerted similar influence on CD8+ T cells. Furthermore, higher acetate concentrations were also able to increase IFN-γ production in CD8+ T lymphocytes by modulating cellular metabolism and mTOR activity. These findings might have significant implications in adoptive immunotherapy of cancers and in anti-viral immunity.
Collapse
|
233
|
Fischer M, Bantug GR, Dimeloe S, Gubser PM, Burgener AV, Grählert J, Balmer ML, Develioglu L, Steiner R, Unterstab G, Sauder U, Hoenger G, Hess C. Early effector maturation of naïve human CD8 + T cells requires mitochondrial biogenesis. Eur J Immunol 2018; 48:1632-1643. [PMID: 30028501 DOI: 10.1002/eji.201747443] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 06/08/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022]
Abstract
The role of mitochondrial biogenesis during naïve to effector differentiation of CD8+ T cells remains ill explored. In this study, we describe a critical role for early mitochondrial biogenesis in supporting cytokine production of nascent activated human naïve CD8+ T cells. Specifically, we found that prior to the first round of cell division activated naïve CD8+ T cells rapidly increase mitochondrial mass, mitochondrial respiration, and mitochondrial reactive oxygen species (mROS) generation, which were all inter-linked and important for CD8+ T cell effector maturation. Inhibition of early mitochondrial biogenesis diminished mROS dependent IL-2 production - as well as subsequent IL-2 dependent TNF, IFN-γ, perforin, and granzyme B production. Together, these findings point to the importance of mitochondrial biogenesis during early effector maturation of CD8+ T cells.
Collapse
Affiliation(s)
- Marco Fischer
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Glenn R Bantug
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Sarah Dimeloe
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Patrick M Gubser
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Anne-Valérie Burgener
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Jasmin Grählert
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Maria L Balmer
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Leyla Develioglu
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Rebekah Steiner
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Gunhild Unterstab
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Ursula Sauder
- Microscopy Center, Biocenter, University of Basel, Basel, Switzerland
| | - Gideon Hoenger
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
234
|
Gaber T, Chen Y, Krauß PL, Buttgereit F. Metabolism of T Lymphocytes in Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:95-148. [PMID: 30635095 DOI: 10.1016/bs.ircmb.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adaptive immune responses that occur in infection, cancer, and autoimmune as well as allergic diseases involve the participation of T cells. T cells travel throughout the body searching for antigens, which are recognized via the major histocompatibility complexes. In the healthy organism, these T cells maintain metabolic quiescence until they encounter a potentially cognate antigen. Once activated, e.g., during an infection or tissue damage, T cells switch their metabolic program to gain energy and building blocks to maintain cellular homeostasis and to fulfill their specific immune functions involving clonal expansion and/or differentiation into effector and memory T cells to ultimately ensure host survival. Thus, differences in metabolism in healthy and pathogenic T cells provide an explanation for dysfunctionality of T-cell responses in metabolic disorders, autoimmunity, and cancer. Here, we summarize current knowledge on T-cell metabolism during the maintenance of homeostasis, activation, and differentiation as well as over the course of time that memory is generated in health and in diseased states such as autoimmunity and cancer.
Collapse
Affiliation(s)
- Timo Gaber
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Yuling Chen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Pierre-Louis Krauß
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Frank Buttgereit
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
235
|
Ratajczak W, Niedźwiedzka-Rystwej P, Tokarz-Deptuła B, Deptuła W. Immunological memory cells. Cent Eur J Immunol 2018; 43:194-203. [PMID: 30135633 PMCID: PMC6102609 DOI: 10.5114/ceji.2018.77390] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 02/16/2018] [Indexed: 02/03/2023] Open
Abstract
This article reviews immunological memory cells, currently represented by T and B lymphocytes and natural killer (NK) cells, which determine a rapid and effective response against a second encounter with the same antigen. Among T lymphocytes, functions of memory cells are provided by their subsets: central memory, effector memory, tissue-resident memory, regulatory memory and stem memory T cells. Memory T and B lymphocytes have an essential role in the immunity against microbial pathogens but are also involved in autoimmunity and maternal-fetal tolerance. Furthermore, the evidence of immunological memory has been established for NK cells. NK cells can respond to haptens or viruses, which results in generation of antigen-specific memory cells. T, B and NK cells, which have a role in immunological memory, have been characterized phenotypically and functionally. During the secondary immune response, these cells are involved in the reaction against foreign antigens, including pathogens, and take part in autoimmune diseases, but also are crucial to immunological tolerance and vaccine therapy.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Scientific Circle of Microbiologists, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | | | - Beata Tokarz-Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | - Wiesław Deptuła
- Department of Microbiology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| |
Collapse
|
236
|
Metabolic Stress in the Immune Function of T Cells, Macrophages and Dendritic Cells. Cells 2018; 7:cells7070068. [PMID: 29966302 PMCID: PMC6070887 DOI: 10.3390/cells7070068] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022] Open
Abstract
Innate and adaptive immune cells from myeloid and lymphoid lineages resolve host infection or cell stress by mounting an appropriate and durable immune response. Upon sensing of cellular insults, immune cells become activated and undergo rapid and efficient functional changes to unleash biosynthesis of macromolecules, proliferation, survival, and trafficking; unprecedented events among other mammalian cells within the host. These changes must become operational within restricted timing to rapidly control the insult and to avoid tissue damage and pathogen spread. Such changes occur at high energy cost. Recent advances have established that plasticity of immune functions occurs in distinct metabolic stress features. Evidence has accumulated to indicate that specific metabolic signatures dictate appropriate immune functions in both innate and adaptive immunity. Importantly, recent studies have shed light on whether successfully manipulating particular metabolic targets is sufficient to modulate immune function and polarization, thereby offering strong therapeutic potential for various common immune-mediated diseases, including inflammation and autoimmune-associated diseases and cancer. In this review, we detail how cellular metabolism controls immune function and phenotype within T cells and macrophages particularly, and the distinct molecular metabolic programming and targets instrumental to engage this regulation.
Collapse
|
237
|
Sun L, Suo C, Li ST, Zhang H, Gao P. Metabolic reprogramming for cancer cells and their microenvironment: Beyond the Warburg Effect. Biochim Biophys Acta Rev Cancer 2018; 1870:51-66. [PMID: 29959989 DOI: 10.1016/j.bbcan.2018.06.005] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023]
Abstract
While metabolic reprogramming of cancer cells has long been considered from the standpoint of how and why cancer cells preferentially utilize glucose via aerobic glycolysis, the so-called Warburg Effect, the progress in the following areas during the past several years has substantially advanced our understanding of the rewired metabolic network in cancer cells that is intertwined with oncogenic signaling. First, in addition to the major nutrient substrates glucose and glutamine, cancer cells have been discovered to utilize a variety of unconventional nutrient sources for survival. Second, the deregulated biomass synthesis is intertwined with cell cycle progression to coordinate the accelerated progression of cancer cells. Third, the reciprocal regulation of cancer cell's metabolic alterations and the microenvironment, involving extensive host immune cells and microbiota, have come into view as critical mechanisms to regulate cancer progression. These and other advances are shaping the current and future paradigm of cancer metabolism.
Collapse
Affiliation(s)
- Linchong Sun
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Caixia Suo
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shi-Ting Li
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Huafeng Zhang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| | - Ping Gao
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
238
|
Degauque N, Brosseau C, Brouard S. Regulation of the Immune Response by the Inflammatory Metabolic Microenvironment in the Context of Allotransplantation. Front Immunol 2018; 9:1465. [PMID: 29988548 PMCID: PMC6026640 DOI: 10.3389/fimmu.2018.01465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Antigen challenge induced by allotransplantation results in the activation of T and B cells, followed by their differentiation and proliferation to mount an effective immune response. Metabolic fitness has been shown to be crucial for supporting the major shift from quiescent to active immune cells and for tuning the immune response. Metabolic reprogramming includes regulation of the balance between glycolysis and mitochondrial respiration processes. Recent research has shed new light on the functions served by the end products of metabolism such as lactate, acetate, and ATP. At enhanced local concentrations, these metabolites have complex effects in which they not only induce T and B cell responses, cell mobility, and cytokine secretion but also favor the resolution of inflammation by promoting regulatory functions. Such mechanisms are instrumental in the context of the immune response in transplantation, not only to protect the graft and/or eliminate cells targeting it but also to maintain cell homeostasis per se. Metabolic adaptation thus plays an instrumental role on the outcome of the cellular and humoral responses. This, of course, raises the possibility of drugs that would interfere in these metabolic pathways to control the immune response but also highlights the risk that some drugs may perturb this metabolism and cell homeostasis and be deleterious for graft outcome. This review focuses on how metabolic alterations of the local immune microenvironment regulate the immune response and the impact of metabolic manipulation in allotransplantation.
Collapse
Affiliation(s)
- Nicolas Degauque
- CRTI UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Carole Brosseau
- CRTI UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Sophie Brouard
- CRTI UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
239
|
Amsen D, van Gisbergen KPJM, Hombrink P, van Lier RAW. Tissue-resident memory T cells at the center of immunity to solid tumors. Nat Immunol 2018; 19:538-546. [PMID: 29777219 DOI: 10.1038/s41590-018-0114-2] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/17/2018] [Indexed: 02/07/2023]
Abstract
Immune responses in tissues are constrained by the physiological properties of the tissue involved. Tissue-resident memory T cells (TRM cells) are a recently discovered lineage of T cells specialized for life and function within tissues. Emerging evidence has shown that TRM cells have a special role in the control of solid tumors. A high frequency of TRM cells in tumors correlates with favorable disease progression in patients with cancer, and studies of mice have shown that TRM cells are necessary for optimal immunological control of solid tumors. Here we describe what defines TRM cells as a separate lineage and how these cells are generated. Furthermore, we discuss the properties that allow TRM cells to operate in normal and transformed tissues, as well as implications for the treatment of patients with cancer.
Collapse
Affiliation(s)
- Derk Amsen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Pleun Hombrink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rene A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
240
|
MacPherson S, Kilgour M, Lum JJ. Understanding lymphocyte metabolism for use in cancer immunotherapy. FEBS J 2018; 285:2567-2578. [PMID: 29611301 DOI: 10.1111/febs.14454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/16/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022]
Abstract
Like all dividing cells, naïve T cells undergo a predictable sequence of events to enter the cell cycle starting from G0 and progressing to G1 , S and finally G2 /M. This methodical series of steps ensures fidelity in the generation of two identical T cells during a single round of division. To achieve this, T cells must activate or inactivate metabolic pathways at discrete times during each phase of the cell cycle. This permits the generation of substrates to support biosynthesis, bioenergetics and the epigenetic changes required for proper differentiation and function. The precursors that feed into these pathways are often shared, highlighting the complex relationship between metabolism and cellular processes that are essential to lymphocytes. It is therefore not surprising that different T cell subtypes exhibit unique metabolic dependencies that change as they mature and go through specialized differentiation programmes. The importance of the influence of metabolism on T cells is underscored by the emerging field of cancer immunotherapy, where autologous T cells can be manufactured ex vivo then infused as a form of curative treatment for human cancers. This review will highlight some of the recent knowledge on T lymphocyte metabolism and give a perspective on the practical implications for cellular-based immunotherapy.
Collapse
Affiliation(s)
- Sarah MacPherson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Marisa Kilgour
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Canada
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Canada
| |
Collapse
|
241
|
Shakespear MR, Iyer A, Cheng CY, Das Gupta K, Singhal A, Fairlie DP, Sweet MJ. Lysine Deacetylases and Regulated Glycolysis in Macrophages. Trends Immunol 2018; 39:473-488. [PMID: 29567326 DOI: 10.1016/j.it.2018.02.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 12/27/2022]
Abstract
Regulated cellular metabolism has emerged as a fundamental process controlling macrophage functions, but there is still much to uncover about the precise signaling mechanisms involved. Lysine acetylation regulates the activity, stability, and/or localization of metabolic enzymes, as well as inflammatory responses, in macrophages. Two protein families, the classical zinc-dependent histone deacetylases (HDACs) and the NAD-dependent HDACs (sirtuins, SIRTs), mediate lysine deacetylation. We describe here mechanisms by which classical HDACs and SIRTs directly regulate specific glycolytic enzymes, as well as evidence that links these protein deacetylases to the regulation of glycolysis-related genes. In these contexts, we discuss HDACs and SIRTs as key control points for regulating immunometabolism and inflammatory outputs from macrophages.
Collapse
Affiliation(s)
- Melanie R Shakespear
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Abishek Iyer
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia; Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Catherine Youting Cheng
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Kaustav Das Gupta
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Amit Singhal
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; Vaccine and Infectious Disease Research Centre (VIDRC), Translational Health Science and Technology Institute (THSTI), National Capital Region (NCR) Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - David P Fairlie
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia; Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
242
|
Mitochondria-Endoplasmic Reticulum Contact Sites Function as Immunometabolic Hubs that Orchestrate the Rapid Recall Response of Memory CD8 + T Cells. Immunity 2018. [PMID: 29523440 DOI: 10.1016/j.immuni.2018.02.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Glycolysis is linked to the rapid response of memory CD8+ T cells, but the molecular and subcellular structural elements enabling enhanced glucose metabolism in nascent activated memory CD8+ T cells are unknown. We found that rapid activation of protein kinase B (PKB or AKT) by mammalian target of rapamycin complex 2 (mTORC2) led to inhibition of glycogen synthase kinase 3β (GSK3β) at mitochondria-endoplasmic reticulum (ER) junctions. This enabled recruitment of hexokinase I (HK-I) to the voltage-dependent anion channel (VDAC) on mitochondria. Binding of HK-I to VDAC promoted respiration by facilitating metabolite flux into mitochondria. Glucose tracing pinpointed pyruvate oxidation in mitochondria, which was the metabolic requirement for rapid generation of interferon-γ (IFN-γ) in memory T cells. Subcellular organization of mTORC2-AKT-GSK3β at mitochondria-ER contact sites, promoting HK-I recruitment to VDAC, thus underpins the metabolic reprogramming needed for memory CD8+ T cells to rapidly acquire effector function.
Collapse
|
243
|
Xiong J, Kawagishi H, Yan Y, Liu J, Wells QS, Edmunds LR, Fergusson MM, Yu ZX, Rovira II, Brittain EL, Wolfgang MJ, Jurczak MJ, Fessel JP, Finkel T. A Metabolic Basis for Endothelial-to-Mesenchymal Transition. Mol Cell 2018; 69:689-698.e7. [PMID: 29429925 DOI: 10.1016/j.molcel.2018.01.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/12/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
Endothelial-to-mesenchymal transition (EndoMT) is a cellular process often initiated by the transforming growth factor β (TGF-β) family of ligands. Although required for normal heart valve development, deregulated EndoMT is linked to a wide range of pathological conditions. Here, we demonstrate that endothelial fatty acid oxidation (FAO) is a critical in vitro and in vivo regulator of EndoMT. We further show that this FAO-dependent metabolic regulation of EndoMT occurs through alterations in intracellular acetyl-CoA levels. Disruption of FAO via conditional deletion of endothelial carnitine palmitoyltransferase II (Cpt2E-KO) augments the magnitude of embryonic EndoMT, resulting in thickening of cardiac valves. Consistent with the known pathological effects of EndoMT, adult Cpt2E-KO mice demonstrate increased permeability in multiple vascular beds. Taken together, these results demonstrate that endothelial FAO is required to maintain endothelial cell fate and that therapeutic manipulation of endothelial metabolism could provide the basis for treating a growing number of EndoMT-linked pathological conditions.
Collapse
Affiliation(s)
- Jianhua Xiong
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Hiroyuki Kawagishi
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Ye Yan
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Jie Liu
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA; Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Quinn S Wells
- Department of Medicine, Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lia R Edmunds
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maria M Fergusson
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Ilsa I Rovira
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Evan L Brittain
- Department of Medicine, Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J Jurczak
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Joshua P Fessel
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Toren Finkel
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA; Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
244
|
Williams NC, O'Neill LAJ. A Role for the Krebs Cycle Intermediate Citrate in Metabolic Reprogramming in Innate Immunity and Inflammation. Front Immunol 2018; 9:141. [PMID: 29459863 PMCID: PMC5807345 DOI: 10.3389/fimmu.2018.00141] [Citation(s) in RCA: 406] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/16/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolism in immune cells is no longer thought of as merely a process for adenosine triphosphate (ATP) production, biosynthesis, and catabolism. The reprogramming of metabolic pathways upon activation is also for the production of metabolites that can act as immune signaling molecules. Activated dendritic cells (DCs) and macrophages have an altered Krebs cycle, one consequence of which is the accumulation of both citrate and succinate. Citrate is exported from the mitochondria via the mitochondrial citrate- carrier. Cytosolic metabolism of citrate to acetyl-coenzyme A (acetyl-CoA) is important for both fatty-acid synthesis and protein acetylation, both of which have been linked to macrophage and DC activation. Citrate-derived itaconate has a direct antibacterial effect and also has been shown to act as an anti-inflammatory agent, inhibiting succinate dehydrogenase. These findings identify citrate as an important metabolite for macrophage and DC effector function.
Collapse
Affiliation(s)
- Niamh C Williams
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
245
|
Zhang L, Romero P. Metabolic Control of CD8+ T Cell Fate Decisions and Antitumor Immunity. Trends Mol Med 2018; 24:30-48. [DOI: 10.1016/j.molmed.2017.11.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 01/20/2023]
|
246
|
Abstract
There is a growing appreciation that metabolic processes and individual metabolites can shape the function of immune cells and thereby play important roles in the outcome of immune responses. In this respect, the use of MS- and NMR spectroscopy-based platforms to characterize and quantify metabolites in biological samples has recently yielded important novel insights into how our immune system functions and has contributed to the identification of biomarkers for immune-mediated diseases. Here, these recent immunological studies in which metabolomics has been used and made significant contributions to these fields will be discussed. In particular the role of metabolomics to the rapidly advancing field of cellular immunometabolism will be highlighted as well as the future prospects of such metabolomic tools in immunology.
Collapse
Affiliation(s)
- Bart Everts
- Department of Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| |
Collapse
|
247
|
Sivanand S, Viney I, Wellen KE. Spatiotemporal Control of Acetyl-CoA Metabolism in Chromatin Regulation. Trends Biochem Sci 2018; 43:61-74. [PMID: 29174173 PMCID: PMC5741483 DOI: 10.1016/j.tibs.2017.11.004] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023]
Abstract
The epigenome is sensitive to the availability of metabolites that serve as substrates of chromatin-modifying enzymes. Links between acetyl-CoA metabolism, histone acetylation, and gene regulation have been documented, although how specificity in gene regulation is achieved by a metabolite has been challenging to answer. Recent studies suggest that acetyl-CoA metabolism is tightly regulated both spatially and temporally to elicit responses to nutrient availability and signaling cues. Here we discuss evidence that acetyl-CoA production is differentially regulated in the nucleus and cytosol of mammalian cells. Recent findings indicate that acetyl-CoA availability for site-specific histone acetylation is influenced through post-translational modification of acetyl-CoA-producing enzymes, as well as through dynamic regulation of the nuclear localization and chromatin recruitment of these enzymes.
Collapse
Affiliation(s)
- Sharanya Sivanand
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Isabella Viney
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
248
|
Ibitokou SA, Dillon BE, Sinha M, Szczesny B, Delgadillo A, Reda Abdelrahman D, Szabo C, Abu-Elheiga L, Porter C, Tuvdendorj D, Stephens R. Early Inhibition of Fatty Acid Synthesis Reduces Generation of Memory Precursor Effector T Cells in Chronic Infection. THE JOURNAL OF IMMUNOLOGY 2017; 200:643-656. [PMID: 29237780 DOI: 10.4049/jimmunol.1602110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 11/09/2017] [Indexed: 01/13/2023]
Abstract
Understanding the mechanisms of CD4 memory T cell (Tmem) differentiation in malaria is critical for vaccine development. However, the metabolic regulation of CD4 Tmem differentiation is not clear, particularly in persistent infections. In this study, we investigated the role of fatty acid synthesis (FAS) in Tmem development in Plasmodium chabaudi chronic mouse malaria infection. We show that T cell-specific deletion and early pharmaceutical inhibition of acetyl CoA carboxylase 1, the rate limiting step of FAS, inhibit generation of early memory precursor effector T cells (MPEC). To compare the role of FAS during early differentiation or survival of Tmem in chronic infection, a specific inhibitor of acetyl CoA carboxylase 1, 5-(tetradecyloxy)-2-furoic acid, was administered at different times postinfection. Strikingly, the number of Tmem was only reduced when FAS was inhibited during T cell priming and not during the Tmem survival phase. FAS inhibition during priming increased effector T cell (Teff) proliferation and strongly decreased peak parasitemia, which is consistent with improved Teff function. Conversely, MPEC were decreased, in a T cell-intrinsic manner, upon early FAS inhibition in chronic, but not acute, infection. Early cure of infection also increased mitochondrial volume in Tmem compared with Teff, supporting previous reports in acute infection. We demonstrate that the MPEC-specific effect was due to the higher fatty acid content and synthesis in MPEC compared with terminally differentiated Teff. In conclusion, FAS in CD4 T cells regulates the early divergence of Tmem from Teff in chronic infection.
Collapse
Affiliation(s)
- Samad A Ibitokou
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Brian E Dillon
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Mala Sinha
- Biomedical Informatics, Institute for Translational Science, University of Texas Medical Branch, Galveston, TX 77555
| | - Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555
| | | | | | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555
| | - Lutfi Abu-Elheiga
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Craig Porter
- Shriners Hospital for Children, Galveston, TX 77550
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Robin Stephens
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; .,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
249
|
de Groot PF, Belzer C, Aydin Ö, Levin E, Levels JH, Aalvink S, Boot F, Holleman F, van Raalte DH, Scheithauer TP, Simsek S, Schaap FG, Olde Damink SWM, Roep BO, Hoekstra JB, de Vos WM, Nieuwdorp M. Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One 2017; 12:e0188475. [PMID: 29211757 PMCID: PMC5718513 DOI: 10.1371/journal.pone.0188475] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Environmental factors driving the development of type 1 diabetes (T1D) are still largely unknown. Both animal and human studies have shown an association between altered fecal microbiota composition, impaired production of short-chain fatty acids (SCFA) and T1D onset. However, observational evidence on SCFA and fecal and oral microbiota in adults with longstanding T1D vs healthy controls (HC) is lacking. RESEARCH DESIGN AND METHODS We included 53 T1D patients without complications or medication and 50 HC matched for age, sex and BMI. Oral and fecal microbiota, fecal and plasma SCFA levels, markers of intestinal inflammation (fecal IgA and calprotectin) and markers of low-grade systemic inflammation were measured. RESULTS Oral microbiota were markedly different in T1D (eg abundance of Streptococci) compared to HC. Fecal analysis showed decreased butyrate producing species in T1D and less butyryl-CoA transferase genes. Also, plasma levels of acetate and propionate were lower in T1D, with similar fecal SCFA. Finally, fecal strains Christensenella and Subdoligranulum correlated with glycemic control, inflammatory parameters and SCFA. CONCLUSIONS We conclude that T1D patients harbor a different amount of intestinal SCFA (butyrate) producers and different plasma acetate and propionate levels. Future research should disentangle cause and effect and whether supplementation of SCFA-producing bacteria or SCFA alone can have disease-modifying effects in T1D.
Collapse
Affiliation(s)
- Pieter F. de Groot
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Ömrüm Aydin
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Johannes H. Levels
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Fransje Boot
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Frits Holleman
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Daniël H. van Raalte
- Department of Internal medicine, VU University Medical Center, Amsterdam, The Netherlands
- ICAR, VU University Medical Center, Amsterdam, The Netherlands
| | - Torsten P. Scheithauer
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
- Department of Internal medicine, VU University Medical Center, Amsterdam, The Netherlands
- ICAR, VU University Medical Center, Amsterdam, The Netherlands
| | - Suat Simsek
- Department of Internal Medicine, Medisch Centrum Alkmaar, Alkmaar, the Netherlands
| | - Frank G. Schaap
- Department of Surgery, Maastricht University, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, the Netherlands
- Department of General, Visceral and Transplantation Surgery, RWTH University Hospital Aachen, Aachen, Germany
| | | | - Bart O. Roep
- Department of Immunohaematology & Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
- Beckman Research Institute, DMRI, City of Hope, Duarte, CA, United States of America
| | - Joost B. Hoekstra
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
- RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Academic Medical Center–University of Amsterdam, Amsterdam, the Netherlands
- Department of Internal medicine, VU University Medical Center, Amsterdam, The Netherlands
- ICAR, VU University Medical Center, Amsterdam, The Netherlands
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
250
|
Zhao S, Torres A, Henry RA, Trefely S, Wallace M, Lee JV, Carrer A, Sengupta A, Campbell SL, Kuo YM, Frey AJ, Meurs N, Viola JM, Blair IA, Weljie AM, Metallo CM, Snyder NW, Andrews AJ, Wellen KE. ATP-Citrate Lyase Controls a Glucose-to-Acetate Metabolic Switch. Cell Rep 2017; 17:1037-1052. [PMID: 27760311 DOI: 10.1016/j.celrep.2016.09.069] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/09/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022] Open
Abstract
Mechanisms of metabolic flexibility enable cells to survive under stressful conditions and can thwart therapeutic responses. Acetyl-coenzyme A (CoA) plays central roles in energy production, lipid metabolism, and epigenomic modifications. Here, we show that, upon genetic deletion of Acly, the gene coding for ATP-citrate lyase (ACLY), cells remain viable and proliferate, although at an impaired rate. In the absence of ACLY, cells upregulate ACSS2 and utilize exogenous acetate to provide acetyl-CoA for de novo lipogenesis (DNL) and histone acetylation. A physiological level of acetate is sufficient for cell viability and abundant acetyl-CoA production, although histone acetylation levels remain low in ACLY-deficient cells unless supplemented with high levels of acetate. ACLY-deficient adipocytes accumulate lipid in vivo, exhibit increased acetyl-CoA and malonyl-CoA production from acetate, and display some differences in fatty acid content and synthesis. Together, these data indicate that engagement of acetate metabolism is a crucial, although partial, mechanism of compensation for ACLY deficiency.
Collapse
Affiliation(s)
- Steven Zhao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - AnnMarie Torres
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan A Henry
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sophie Trefely
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Martina Wallace
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joyce V Lee
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessandro Carrer
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sydney L Campbell
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yin-Ming Kuo
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Alexander J Frey
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Noah Meurs
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - John M Viola
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christian M Metallo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Andrew J Andrews
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|