201
|
Yeshi K, Creek DJ, Anderson D, Ritmejerytė E, Becker L, Loukas A, Wangchuk P. Metabolomes and Lipidomes of the Infective Stages of the Gastrointestinal nematodes, Nippostrongylus brasiliensis and Trichuris muris. Metabolites 2020; 10:metabo10110446. [PMID: 33171998 PMCID: PMC7694664 DOI: 10.3390/metabo10110446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
Soil-transmitted helminths, including hookworms and whipworms, infect billions of people worldwide. Their capacity to penetrate and migrate through their hosts’ tissues is influenced by the suite of molecules produced by the infective developmental stages. To facilitate a better understanding of the immunobiology and pathogenicity of human hookworms and whipworms, we investigated the metabolomes of the infective stage of Nippostrongylus brasiliensis third-stage larvae (L3) which penetrate the skin and Trichuris muris eggs which are orally ingested, using untargeted liquid chromatography-mass spectrometry (LC-MS). We identified 55 polar metabolites through Metabolomics Standard Initiative level-1 (MSI-I) identification from N. brasiliensis and T. muris infective stages, out of which seven were unique to excretory/secretory products (ESPs) of N. brasiliensis L3. Amino acids were a principal constituent (33 amino acids). Additionally, we identified 350 putative lipids, out of which 28 (all known lipids) were unique to N. brasiliensis L3 somatic extract and four to T. muris embryonated egg somatic extract. Glycerophospholipids and glycerolipids were the major lipid groups. The catalogue of metabolites identified in this study shed light on the biology, and possible therapeutic and diagnostic targets for the treatment of these critical infectious pathogens. Moreover, with the growing body of literature on the therapeutic utility of helminth ESPs for treating inflammatory diseases, a role for metabolites is likely but has received little attention thus far.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
- Correspondence: (K.Y.); (P.W.)
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; (D.J.C.); (D.A.)
| | - Dovile Anderson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; (D.J.C.); (D.A.)
| | - Edita Ritmejerytė
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Luke Becker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
| | - Phurpa Wangchuk
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; (E.R.); (L.B.); (A.L.)
- Correspondence: (K.Y.); (P.W.)
| |
Collapse
|
202
|
Moreno C, Santos RM, Burns R, Zhang WC. Succinate Dehydrogenase and Ribonucleic Acid Networks in Cancer and Other Diseases. Cancers (Basel) 2020; 12:cancers12113237. [PMID: 33153035 PMCID: PMC7693138 DOI: 10.3390/cancers12113237] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Although the dysfunction of the succinate dehydrogenase complex in mitochondria leads to cancer and other diseases due to aberrant metabolic reactions and signaling pathways, it is not well known how the succinate dehydrogenase complex is regulated. Our review highlights that non-coding ribonucleic acids (RNAs), RNA editing enzymes, and RNA modifying enzymes regulate expressions and functions of the succinate dehydrogenase complex. This research will provide new strategies for treating succinate dehydrogenase-relevant diseases in a clinic. Abstract Succinate dehydrogenase (SDH) complex connects both the tricarboxylic acid (TCA) cycle and the electron transport chain (ETC) in the mitochondria. However, SDH mutation or dysfunction-induced succinate accumulation results in multiple cancers and non-cancer diseases. The mechanistic studies show that succinate activates hypoxia response and other signal pathways via binding to 2-oxoglutarate-dependent oxygenases and succinate receptors. Recently, the increasing knowledge of ribonucleic acid (RNA) networks, including non-coding RNAs, RNA editors, and RNA modifiers has expanded our understanding of the interplay between SDH and RNA networks in cancer and other diseases. Here, we summarize recent discoveries in the RNA networks and their connections to SDH. Additionally, we discuss current therapeutics targeting SDH in both pre-clinical and clinical trials. Thus, we propose a new model of SDH–RNA network interaction and bring promising RNA therapeutics against SDH-relevant cancer and other diseases.
Collapse
|
203
|
Henderson J, Duffy L, Stratton R, Ford D, O'Reilly S. Metabolic reprogramming of glycolysis and glutamine metabolism are key events in myofibroblast transition in systemic sclerosis pathogenesis. J Cell Mol Med 2020; 24:14026-14038. [PMID: 33140521 PMCID: PMC7754020 DOI: 10.1111/jcmm.16013] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022] Open
Abstract
Systemic Sclerosis (SSc) is a rare fibrotic autoimmune disorder for which no curative treatments currently exist. Metabolic remodelling has recently been implicated in other autoimmune diseases; however, its potential role in SSc has received little attention. Here, we aimed to determine whether changes to glycolysis and glutaminolysis are important features of skin fibrosis. TGF‐β1, the quintessential pro‐fibrotic stimulus, was used to activate fibrotic pathways in NHDFs in vitro. Dermal fibroblasts derived from lesions of SSc patients were also used for in vitro experiments. Parameters of glycolytic function were assessed using by measuring extracellular acidification in response to glycolytic activators/inhibitors, whilst markers of fibrosis were measured by Western blotting following the use of the glycolysis inhibitors 2‐dg and 3PO and the glutaminolysis inhibitor G968. Succinate was also measured after TGF‐β1 stimulation. Itaconate was added to SSc fibroblasts and collagen examined. TGF‐β1 up‐regulates glycolysis in dermal fibroblasts, and inhibition of glycolysis attenuates its pro‐fibrotic effects. Furthermore, inhibition of glutamine metabolism also reverses TGF‐β1‐induced fibrosis, whilst glutaminase expression is up‐regulated in dermal fibroblasts derived from SSc patient skin lesions, suggesting that enhanced glutamine metabolism is another aspect of the pro‐fibrotic metabolic phenotype in skin fibrosis. TGF‐β1 was also able to enhance succinate production, with increased succinate shown to be associated with increased collagen expression. Incubation of SSc cells with itaconate, an important metabolite, reduced collagen expression. TGF‐β1 activation of glycolysis is a key feature of the fibrotic phenotype induced by TGF‐B1 in skin cells, whilst increased glutaminolysis is also evident in SSc fibroblasts.
Collapse
Affiliation(s)
- John Henderson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Laura Duffy
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Richard Stratton
- Centre for Rheumatic and Connective Tissue Diseases, University College London, London, UK
| | - Dianne Ford
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Steven O'Reilly
- School of Biological and Biomedical Sciences, Durham University, Durham, UK
| |
Collapse
|
204
|
|
205
|
Targeting interleukin-4 to the arthritic joint. J Control Release 2020; 326:172-180. [DOI: 10.1016/j.jconrel.2020.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 01/08/2023]
|
206
|
Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim Biophys Acta Rev Cancer 2020; 1874:188427. [PMID: 32961257 DOI: 10.1016/j.bbcan.2020.188427] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023]
Abstract
Macrophages are innate phagocytic leukocytes that are highly present in solid tumors, where they are referred to as tumor-associated macrophages (TAMs). In solid tumors, the microenvironment is often immunosuppressive and hypoxic regions are prevalent. These hypoxic conditions impose tumor cells to reprogram their metabolism, shifting from oxidative phosphorylation to anaerobic glycolysis. This so-called glycolytic switch enables hypoxic tumor cells to survive, proliferate, and eventually to outcompete untransformed cells. The hypoxia-induced change in tumor cell metabolism leads to the production of oncometabolites, among which are the glycolytic end-metabolite lactate and the tricarboxylic acid cycle intermediate succinate. TAMs can react to these oncometabolites, resulting in an altered maturation and the adoption of pro-angiogenic features. These angiogenesis-promoting TAMs have been reported to cooperate with tumor cells in the formation of new vessels, and even have been considered an important cause of resistance against anti-angiogenic therapies. For a long time, the mechanisms by which lactate and succinate activated pro-angiogenic TAMs were not understood. Researchers now start to unravel and understand some of the underlying mechanisms. Here, the importance of microenvironmental cues in inducing different macrophage activation states is discussed, as well as the role of hypoxia in the recruitment and activation of pro-angiogenic macrophages. In addition, the latest findings on the oncometabolites lactate and succinate in the activation of angiogenesis supporting macrophages are reviewed. Finally, various oncometabolite-targeting therapeutic strategies are proposed that could improve the response to anti-angiogenic therapies. SIGNIFICANCE STATEMENT: Tumor-associated macrophages (TAMs) are known promotors of tumor neovascularization, and significantly contribute to the emergence of resistance to anti-angiogenic therapies. Recent evidence suggests that the angiogenesis promoting phenotype of TAMs can be activated by hypoxic tumor cell-derived oncometabolites, including lactate and succinate. Here, the latest findings into the lactate- and succinate-mediated mechanistic activation of pro-angiogenic TAMs are reviewed, and therapeutic strategies that interfere with this mechanism and may delay or even prevent acquired resistance to anti-angiogenic agents are discussed.
Collapse
|
207
|
Reddy A, Bozi LHM, Yaghi OK, Mills EL, Xiao H, Nicholson HE, Paschini M, Paulo JA, Garrity R, Laznik-Bogoslavski D, Ferreira JCB, Carl CS, Sjøberg KA, Wojtaszewski JFP, Jeppesen JF, Kiens B, Gygi SP, Richter EA, Mathis D, Chouchani ET. pH-Gated Succinate Secretion Regulates Muscle Remodeling in Response to Exercise. Cell 2020; 183:62-75.e17. [PMID: 32946811 DOI: 10.1016/j.cell.2020.08.039] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/05/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022]
Abstract
In response to skeletal muscle contraction during exercise, paracrine factors coordinate tissue remodeling, which underlies this healthy adaptation. Here we describe a pH-sensing metabolite signal that initiates muscle remodeling upon exercise. In mice and humans, exercising skeletal muscle releases the mitochondrial metabolite succinate into the local interstitium and circulation. Selective secretion of succinate is facilitated by its transient protonation, which occurs upon muscle cell acidification. In the protonated monocarboxylic form, succinate is rendered a transport substrate for monocarboxylate transporter 1, which facilitates pH-gated release. Upon secretion, succinate signals via its cognate receptor SUCNR1 in non-myofibrillar cells in muscle tissue to control muscle-remodeling transcriptional programs. This succinate-SUCNR1 signaling is required for paracrine regulation of muscle innervation, muscle matrix remodeling, and muscle strength in response to exercise training. In sum, we define a bioenergetic sensor in muscle that utilizes intracellular pH and succinate to coordinate tissue adaptation to exercise.
Collapse
Affiliation(s)
- Anita Reddy
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Luiz H M Bozi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hilary E Nicholson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Margherita Paschini
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ryan Garrity
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Christian S Carl
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
208
|
Harber KJ, de Goede KE, Verberk SGS, Meinster E, de Vries HE, van Weeghel M, de Winther MPJ, Van den Bossche J. Succinate Is an Inflammation-Induced Immunoregulatory Metabolite in Macrophages. Metabolites 2020; 10:metabo10090372. [PMID: 32942769 PMCID: PMC7569821 DOI: 10.3390/metabo10090372] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/28/2020] [Accepted: 09/15/2020] [Indexed: 01/09/2023] Open
Abstract
Immunometabolism revealed the crucial role of cellular metabolism in controlling immune cell phenotype and functions. Macrophages, key immune cells that support progression of numerous inflammatory diseases, have been well described as undergoing vast metabolic rewiring upon activation. The immunometabolite succinate particularly gained a lot of attention and emerged as a crucial regulator of macrophage responses and inflammation. Succinate was originally described as a metabolite that supports inflammation via distinct routes. Recently, studies have indicated that succinate and its receptor SUCNR1 can suppress immune responses as well. These apparent contradictory effects might be due to specific experimental settings and particularly the use of distinct succinate forms. We therefore compared the phenotypic and functional effects of distinct succinate forms and receptor mouse models that were previously used for studying succinate immunomodulation. Here, we show that succinate can suppress secretion of inflammatory mediators IL-6, tumor necrosis factor (TNF) and nitric oxide (NO), as well as inhibit Il1b mRNA expression of inflammatory macrophages in a SUCNR1-independent manner. We also observed that macrophage SUCNR1 deficiency led to an enhanced inflammatory response without addition of exogenous succinate. While our study does not reveal new mechanistic insights into how succinate elicits different inflammatory responses, it does indicate that the inflammatory effects of succinate and its receptor SUCNR1 in macrophages are clearly context dependent.
Collapse
Affiliation(s)
- Karl J. Harber
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Experimental Vascular Biology, 1105 AZ Amsterdam, The Netherlands;
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Kyra E. de Goede
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
| | - Sanne G. S. Verberk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
| | - Elisa Meinster
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Menno P. J. de Winther
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Experimental Vascular Biology, 1105 AZ Amsterdam, The Netherlands;
| | - Jan Van den Bossche
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (K.J.H.); (K.E.d.G.); (S.G.S.V.); (E.M.); (H.E.d.V.)
- Correspondence:
| |
Collapse
|
209
|
Flerin NC, Pinioti S, Menga A, Castegna A, Mazzone M. Impact of Immunometabolism on Cancer Metastasis: A Focus on T Cells and Macrophages. Cold Spring Harb Perspect Med 2020; 10:a037044. [PMID: 31615868 PMCID: PMC7461771 DOI: 10.1101/cshperspect.a037044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite improved treatment options, cancer remains the leading cause of morbidity and mortality worldwide, with 90% of this mortality correlated to the development of metastasis. Since metastasis has such an impact on treatment success, disease outcome, and global health, it is important to understand the different steps and factors playing key roles in this process, how these factors relate to immune cell function and how we can target metabolic processes at different steps of metastasis in order to improve cancer treatment and patient prognosis. Recent insights in immunometabolism direct to promising therapeutic targets for cancer treatment, however, the specific contribution of metabolism on antitumor immunity in different metastatic niches warrant further investigation. Here, we provide an overview of what is so far known in the field of immunometabolism at different steps of the metastatic cascade, and what may represent the next steps forward. Focusing on metabolic checkpoints in order to translate these findings from in vitro and mouse studies to the clinic has the potential to revolutionize cancer immunotherapy and greatly improve patient prognosis.
Collapse
Affiliation(s)
- Nina C Flerin
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Sotiria Pinioti
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Alessio Menga
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari 70125, Italy
- IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| |
Collapse
|
210
|
Zhang J, Zhang Q, Yang Y, Wang Q. Association Between Succinate Receptor SUCNR1 Expression and Immune Infiltrates in Ovarian Cancer. Front Mol Biosci 2020; 7:150. [PMID: 33062639 PMCID: PMC7488939 DOI: 10.3389/fmolb.2020.00150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/18/2020] [Indexed: 12/23/2022] Open
Abstract
Background The activation of succinate receptor 1 (SUCNR1) by extracellular succinate has been found to regulate immune cell function. However, the clinical significance of SUCNR1 in ovarian cancer and its correlation with tumor-infiltrating lymphocytes remain unclear. Methods The genetic alteration and expression patterns of SUCNR1 were analyzed by using cBioPortal and Gene Expression Omnibus (GEO) datasets. Kaplan-Meier Plotter was used to assess the prognostic value of SUCNR1 in patients with ovarian cancer. The correlations between SUCNR1 expression and immune infiltration, gene markers of immune cells, cytokines, chemokines, or T cell exhaustion were explored by using TIMER and TISIDB platforms. We also performed Gene Set Enrichment Analysis (GSEA) to reveal biological function of SUCNR1 in ovarian cancer. Results The expression of SUCNR1 was closely related to tumor infiltrating lymphocytes, multiple gene markers of immune cells, and T cell exhaustion in ovarian cancer. The expression of SUCNR1 was also associated with the expression of cytokine- or chemokine-related genes. Moreover, GSEA revealed that various immune-related pathways might be regulated by SUCNR1. In addition, we found that SUCNR1 was amplified in ovarian cancer, and the high expression of SUCNR1 predicted worse progression-free survival (p = 0.0073, HR = 1.49, 95% CI = 1.11–2). Conclusion These results highlight the role of SUCNR1 in regulating tumor immunity in ovarian cancer.
Collapse
Affiliation(s)
- Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinyi Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongbin Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
211
|
Velcicky J, Wilcken R, Cotesta S, Janser P, Schlapbach A, Wagner T, Piechon P, Villard F, Bouhelal R, Piller F, Harlfinger S, Stringer R, Fehlmann D, Kaupmann K, Littlewood-Evans A, Haffke M, Gommermann N. Discovery and Optimization of Novel SUCNR1 Inhibitors: Design of Zwitterionic Derivatives with a Salt Bridge for the Improvement of Oral Exposure. J Med Chem 2020; 63:9856-9875. [DOI: 10.1021/acs.jmedchem.0c01020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Juraj Velcicky
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Rainer Wilcken
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Simona Cotesta
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Philipp Janser
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Achim Schlapbach
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Trixie Wagner
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Philippe Piechon
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Frederic Villard
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Rochdi Bouhelal
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Fabian Piller
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Rowan Stringer
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Klemens Kaupmann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Matthias Haffke
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Nina Gommermann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
212
|
Hackett EE, Sheedy FJ. An Army Marches on Its Stomach: Metabolic Intermediates as Antimicrobial Mediators in Mycobacterium tuberculosis Infection. Front Cell Infect Microbiol 2020; 10:446. [PMID: 32984072 PMCID: PMC7477320 DOI: 10.3389/fcimb.2020.00446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
The cells of the immune system are reliant on their metabolic state to launch effective responses to combat mycobacterial infections. The bioenergetic profile of the cell determines the molecular fuels and metabolites available to the host, as well as to the bacterial invader. How cells utilize the nutrients in their microenvironment—including glucose, lipids and amino acids—to sustain their functions and produce antimicrobial metabolites, and how mycobacteria exploit this to evade the immune system is of great interest. Changes in flux through metabolic pathways alters the intermediate metabolites present. These intermediates are beginning to be recognized as key modulators of immune signaling as well as direct antimicrobial effectors, and their impact on tuberculosis infection is becoming apparent. A better understanding of how metabolism impacts immunity to Mycobacterium tuberculosis and how it is regulated and thus can be manipulated will open the potential for novel therapeutic interventions and vaccination strategies.
Collapse
Affiliation(s)
- Emer E Hackett
- Macrophage Homeostasis, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Frederick J Sheedy
- Macrophage Homeostasis, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
213
|
Sabapathy V, Venkatadri R, Dogan M, Sharma R. The Yin and Yang of Alarmins in Regulation of Acute Kidney Injury. Front Med (Lausanne) 2020; 7:441. [PMID: 32974364 PMCID: PMC7472534 DOI: 10.3389/fmed.2020.00441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a major clinical burden affecting 20 to 50% of hospitalized and intensive care patients. Irrespective of the initiating factors, the immune system plays a major role in amplifying the disease pathogenesis with certain immune cells contributing to renal damage, whereas others offer protection and facilitate recovery. Alarmins are small molecules and proteins that include granulysins, high-mobility group box 1 protein, interleukin (IL)-1α, IL-16, IL-33, heat shock proteins, the Ca++ binding S100 proteins, adenosine triphosphate, and uric acid. Alarmins are mostly intracellular molecules, and their release to the extracellular milieu signals cellular stress or damage, generally leading to the recruitment of the cells of the immune system. Early studies indicated a pro-inflammatory role for the alarmins by contributing to immune-system dysregulation and worsening of AKI. However, recent developments demonstrate anti-inflammatory mechanisms of certain alarmins or alarmin-sensing receptors, which may participate in the prevention, resolution, and repair of AKI. This dual function of alarmins is intriguing and has confounded the role of alarmins in AKI. In this study, we review the contribution of various alarmins to the pathogenesis of AKI in experimental and clinical studies. We also analyze the approaches for the therapeutic utilization of alarmins for AKI.
Collapse
Affiliation(s)
| | | | | | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
214
|
Rodgers LC, Cole J, Rattigan KM, Barrett MP, Kurian N, McInnes IB, Goodyear CS. The rheumatoid synovial environment alters fatty acid metabolism in human monocytes and enhances CCL20 secretion. Rheumatology (Oxford) 2020; 59:869-878. [PMID: 31497857 DOI: 10.1093/rheumatology/kez378] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/05/2019] [Accepted: 08/02/2019] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES Fatty acid oxidation (FAO) and glycolysis have been implicated in immune regulation and activation of macrophages. However, investigation of human monocyte intracellular metabolism in the context of the hypoxic and inflammatory rheumatoid arthritis (RA) synovium is lacking. We hypothesized that exposure of monocytes to the hypoxic and inflammatory RA environment would have a profound impact on their metabolic state, and potential to contribute to disease pathology. METHODS Human monocytes were isolated from buffy coats and exposed to hypoxia. Metabolic profiling of monocytes was carried out by LC-MS metabolomics. Inflammatory mediator release after LPS or RA-synovial fluid (RA-SF) stimulation was analysed by ELISA. FAO was inhibited by etomoxir or enhanced with exogenous carnitine supplementation. Transcriptomics of RA blood monocytes and RA-SF macrophages was carried out by microarray. RESULTS Hypoxia exacerbated monocyte-derived CCL20 and IL-1β release in response to LPS, and increased glycolytic intermediates at the expense of carnitines. Modulation of carnitine identified a novel role for FAO in the production of CCL20 in response to LPS. Transcriptional analysis of RA blood monocytes and RA-SF macrophages revealed that fatty acid metabolism was altered and CCL20 increased when monocytes enter the synovial environment. In vitro analysis of monocytes showed that RA-SF increases carnitine abundance and CCL20 production in hypoxia, which was exacerbated by exogenous carnitine. CONCLUSION This work has revealed a novel inflammatory mechanism in RA that links FAO to CCL20 production in human monocytes, which could subsequently contribute to RA disease pathogenesis by promoting the recruitment of Th17 cells and osteoclastogenesis.
Collapse
Affiliation(s)
- Lewis C Rodgers
- Centre of Immunobiology, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Glasgow, UK
| | - John Cole
- GLAZgo Discovery Centre, Glasgow, UK
| | - Kevin M Rattigan
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Glasgow, UK
| | - Michael P Barrett
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Glasgow, UK.,Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nisha Kurian
- Respiratory Inflammation and Autoimmune (RIA) Precision Medicine Unit, Precision Medicine, Oncology R&D, AstraZeneca, Gothenburg, Sweden
| | - Iain B McInnes
- Centre of Immunobiology, University of Glasgow, Glasgow, UK
| | - Carl S Goodyear
- Centre of Immunobiology, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Glasgow, UK
| |
Collapse
|
215
|
Li X, Xie L, Qu X, Zhao B, Fu W, Wu B, Wu J. GPR91, a critical signaling mechanism in modulating pathophysiologic processes in chronic illnesses. FASEB J 2020; 34:13091-13105. [PMID: 32812686 DOI: 10.1096/fj.202001037r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022]
Abstract
Succinate receptor GPR91 is one of G protein-coupled receptors (GPCRs), and is expressed in a variety of cell types and tissues. Succinate is its natural ligand, and its activation represents that an intrinsic metabolic intermediate exerts a regulatory role on many critical life processes involving pathophysiologic mechanisms, such as innate immunity, inflammation, tissue repair, and oncogenesis. With the illustration of 3-dimensional crystal structure of the receptor and discovery of its antagonists, it is possible to dissect the succinate-GPR91-G protein signaling pathways in different cell types under pathophysiological conditions. Deep understanding of the GPR91-ligand binding mode with various agonists and antagonists would aid in elucidating the molecular basis of a spectrum of chronic illnesses, such as hypertension, diabetes, and their renal and retina complications, metabolic-associated fatty liver diseases, such as nonalcoholic steatohepatitis and its fibrotic progression, inflammatory bowel diseases (Crohn's disease and ulcerative colitis), age-related macular degeneration, rheumatoid arthritis, and progressive behaviors of malignancies. With better delineation of critical regulatory role of the succinate-GPR91 axis in these illnesses, therapeutic intervention may be developed by specifically targeting this signaling pathway with small molecular antagonists or other strategies.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Medical Microbiology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Xie
- Department of Medical Microbiology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiangli Qu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bangyi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Beili Wu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jian Wu
- Department of Medical Microbiology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Gastroenterology & Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
216
|
Metabolic and Molecular Mechanisms of Macrophage Polarisation and Adipose Tissue Insulin Resistance. Int J Mol Sci 2020; 21:ijms21165731. [PMID: 32785109 PMCID: PMC7460862 DOI: 10.3390/ijms21165731] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/26/2020] [Accepted: 08/08/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammation plays a key role in the development and progression of type-2 diabetes (T2D), a disease characterised by peripheral insulin resistance and systemic glucolipotoxicity. Visceral adipose tissue (AT) is the main source of inflammation early in the disease course. Macrophages are innate immune cells that populate all peripheral tissues, including AT. Dysregulated AT macrophage (ATM) responses to microenvironmental changes are at the root of aberrant inflammation and development of insulin resistance, locally and systemically. The inflammatory activation of macrophages is regulated at multiple levels: cell surface receptor stimulation, intracellular signalling, transcriptional and metabolic levels. This review will cover the main mechanisms involved in AT inflammation and insulin resistance in T2D. First, we will describe the physiological and pathological changes in AT that lead to inflammation and insulin resistance. We will next focus on the transcriptional and metabolic mechanisms described that lead to the activation of ATMs. We will discuss more novel metabolic mechanisms that influence macrophage polarisation in other disease or tissue contexts that may be relevant to future work in insulin resistance and T2D.
Collapse
|
217
|
Soto‐Heredero G, Gómez de las Heras MM, Gabandé‐Rodríguez E, Oller J, Mittelbrunn M. Glycolysis - a key player in the inflammatory response. FEBS J 2020; 287:3350-3369. [PMID: 32255251 PMCID: PMC7496292 DOI: 10.1111/febs.15327] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/06/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
The inflammatory response involves the activation of several cell types to fight insults caused by a plethora of agents, and to maintain the tissue homoeostasis. On the one hand, cells involved in the pro-inflammatory response, such as inflammatory M1 macrophages, Th1 and Th17 lymphocytes or activated microglia, must rapidly provide energy to fuel inflammation, which is essentially accomplished by glycolysis and high lactate production. On the other hand, regulatory T cells or M2 macrophages, which are involved in immune regulation and resolution of inflammation, preferentially use fatty acid oxidation through the TCA cycle as a main source for energy production. Here, we discuss the impact of glycolytic metabolism at the different steps of the inflammatory response. Finally, we review a wide variety of molecular mechanisms which could explain the relationship between glycolytic metabolites and the pro-inflammatory phenotype, including signalling events, epigenetic remodelling, post-transcriptional regulation and post-translational modifications. Inflammatory processes are a common feature of many age-associated diseases, such as cardiovascular and neurodegenerative disorders. The finding that immunometabolism could be a master regulator of inflammation broadens the avenue for treating inflammation-related pathologies through the manipulation of the vascular and immune cell metabolism.
Collapse
Affiliation(s)
- Gonzalo Soto‐Heredero
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - Manuel M. Gómez de las Heras
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
| | - Enrique Gabandé‐Rodríguez
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - Jorge Oller
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - María Mittelbrunn
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| |
Collapse
|
218
|
Riquelme SA, Lozano C, Moustafa AM, Liimatta K, Tomlinson KL, Britto C, Khanal S, Gill SK, Narechania A, Azcona-Gutiérrez JM, DiMango E, Saénz Y, Planet P, Prince A. CFTR-PTEN-dependent mitochondrial metabolic dysfunction promotes Pseudomonas aeruginosa airway infection. Sci Transl Med 2020; 11:11/499/eaav4634. [PMID: 31270271 DOI: 10.1126/scitranslmed.aav4634] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/12/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor best known for regulating cell proliferation and metabolism. PTEN forms a complex with the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) at the plasma membrane, and this complex is known to be functionally impaired in CF. Here, we demonstrated that the combined effect of PTEN and CFTR dysfunction stimulates mitochondrial activity, resulting in excessive release of succinate and reactive oxygen species. This environment promoted the colonization of the airway by Pseudomonas aeruginosa, bacteria that preferentially metabolize succinate, and stimulated an anti-inflammatory host response dominated by immune-responsive gene 1 (IRG1) and itaconate. The recruitment of myeloid cells induced by these strains was inefficient in clearing the infection and increased numbers of phagocytes accumulated under CFTR-PTEN axis dysfunction. This central metabolic defect in mitochondrial function due to impaired PTEN activity contributes to P. aeruginosa infection in CF.
Collapse
Affiliation(s)
| | - Carmen Lozano
- Area de Microbiología Molecular, Centro de Investigación Biomédica de la Rioja (CIBIR), Microbiología Molecular, Logroño, LG 26006, Spain
| | - Ahmed M Moustafa
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kalle Liimatta
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Kira L Tomlinson
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Clemente Britto
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sara Khanal
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Simren K Gill
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | | - Jose M Azcona-Gutiérrez
- Departamento de Diagnóstico Biomédico. Laboratorio de Microbiología, Hospital San Pedro, Logroño, LG 26006, Spain
| | - Emily DiMango
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Yolanda Saénz
- Area de Microbiología Molecular, Centro de Investigación Biomédica de la Rioja (CIBIR), Microbiología Molecular, Logroño, LG 26006, Spain
| | - Paul Planet
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alice Prince
- Department of Pediatrics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
219
|
Ketelhuth DFJ, Lutgens E, Bäck M, Binder CJ, Van den Bossche J, Daniel C, Dumitriu IE, Hoefer I, Libby P, O'Neill L, Weber C, Evans PC. Immunometabolism and atherosclerosis: perspectives and clinical significance: a position paper from the Working Group on Atherosclerosis and Vascular Biology of the European Society of Cardiology. Cardiovasc Res 2020; 115:1385-1392. [PMID: 31228191 DOI: 10.1093/cvr/cvz166] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/19/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Inflammation is an important driver of atherosclerosis, and the favourable outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial revealed the large potential of anti-inflammatory drugs for the treatment of cardiovascular disease, especially in patients with a pro-inflammatory constitution. However, the complex immune reactions driving inflammation in the vascular wall in response to an atherosclerotic microenvironment are still being unravelled. Novel insights into the cellular processes driving immunity and inflammation revealed that alterations in intracellular metabolic pathways are strong drivers of survival, growth, and function of immune cells. Therefore, this position paper presents a brief overview of the recent developments in the immunometabolism field, focusing on its role in atherosclerosis. We will also highlight the potential impact of immunometabolic markers and targets in clinical cardiovascular medicine.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Esther Lutgens
- Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Magnus Bäck
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria and CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jan Van den Bossche
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Carolin Daniel
- Division of Clinical Pharmacology, Department of Medicine IV, Ludwig Maximilians University of Munich, Munich, Germany
| | - Ingrid E Dumitriu
- Molecular and Clinical Sciences Research Institute & Cardiology Clinical Academic Group, St. George's Hospital, University of London, Cranmer Terrace, London, UK
| | - Imo Hoefer
- Laboratory of Clinical Chemistry and Hematology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luke O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute of In Silico Medicine and the Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
220
|
Yang X, Chang Y, Wei W. Emerging role of targeting macrophages in rheumatoid arthritis: Focus on polarization, metabolism and apoptosis. Cell Prolif 2020; 53:e12854. [PMID: 32530555 PMCID: PMC7377929 DOI: 10.1111/cpr.12854] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/09/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages maintain a dynamic balance in physiology. Various known or unknown microenvironmental signals influence the polarization, activation and death of macrophages, which creates an imbalance that leads to disease. Rheumatoid arthritis (RA) is characterized by the massive infiltration of a variety of chronic inflammatory cells in synovia. Abundant activated macrophages found in RA synovia are an early hallmark of RA, and the number of these macrophages can be decreased after effective treatment. In RA, the proportion of M1 (pro‐inflammatory macrophages) is higher than that of M2 (anti‐inflammatory macrophages). The increased pro‐inflammatory ability of macrophages is related to their excessive activation and proliferation as well as an enhanced anti‐apoptosis ability. At present, there are no clinical therapies specific to macrophages in RA. Understanding the mechanisms and functional consequences of the heterogeneity of macrophages will aid in confirming their potential role in inflammation development. This review will outline RA‐related macrophage properties (focus on polarization, metabolism and apoptosis) as well as the origin of macrophages. The molecular mechanisms that drive macrophage properties also be elucidated to identify novel therapeutic targets for RA and other autoimmune disease.
Collapse
Affiliation(s)
- Xuezhi Yang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
221
|
Liu X, Zhu C, Zha H, Tang J, Rong F, Chen X, Fan S, Xu C, Du J, Zhu J, Wang J, Ouyang G, Yu G, Cai X, Chen Z, Xiao W. SIRT5 impairs aggregation and activation of the signaling adaptor MAVS through catalyzing lysine desuccinylation. EMBO J 2020; 39:e103285. [PMID: 32301534 PMCID: PMC7265249 DOI: 10.15252/embj.2019103285] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
RLR-mediated type I IFN production plays a pivotal role in innate antiviral immune responses, where the signaling adaptor MAVS is a critical determinant. Here, we show that MAVS is a physiological substrate of SIRT5. Moreover, MAVS is succinylated upon viral challenge, and SIRT5 catalyzes desuccinylation of MAVS. Mass spectrometric analysis indicated that Lysine 7 of MAVS is succinylated. SIRT5-catalyzed desuccinylation of MAVS at Lysine 7 diminishes the formation of MAVS aggregation after viral infection, resulting in the inhibition of MAVS activation and leading to the impairment of type I IFN production and antiviral gene expression. However, the enzyme-deficient mutant of SIRT5 (SIRT5-H158Y) loses its suppressive role on MAVS activation. Furthermore, we show that Sirt5-deficient mice are resistant to viral infection. Our study reveals the critical role of SIRT5 in limiting RLR signaling through desuccinylating MAVS.
Collapse
Affiliation(s)
- Xing Liu
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Chunchun Zhu
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Huangyuan Zha
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Jinhua Tang
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fangjing Rong
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaoyun Chen
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Sijia Fan
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chenxi Xu
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Juan Du
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Junji Zhu
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Gang Ouyang
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Guangqing Yu
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Zhu Chen
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and BiotechnologyInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
- The Key Laboratory of Aquaculture Disease ControlMinistry of AgricultureWuhanChina
- The Key Laboratory of Aquatic Biodiversity and ConservationInstitute of HydrobiologyChinese Academy of SciencesWuhanChina
- The Innovation Academy of Seed DesignChinese Academy of SciencesWuhanChina
| |
Collapse
|
222
|
Cytokine-like Roles for Metabolites in Immunity. Mol Cell 2020; 78:814-823. [DOI: 10.1016/j.molcel.2020.04.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/17/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
|
223
|
Liu P, Wang J, Wen W, Pan T, Chen H, Fu Y, Wang F, Huang JH, Xu S. Cinnamaldehyde suppresses NLRP3 derived IL-1β via activating succinate/HIF-1 in rheumatoid arthritis rats. Int Immunopharmacol 2020; 84:106570. [PMID: 32413739 DOI: 10.1016/j.intimp.2020.106570] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/18/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Cinnamaldehyde (CA) is an essential component of cinnamon (Cinnamomum cassia Presland), which is often used as a flavoring condiment in beverages, pastries, perfumes, etc. Cinnamon is also used as herbal medicine in China and Southeast Asia to treat rheumatoid arthritis. However, the molecular mechanism is unclear. In this study, we aim to investigate its anti-inflammatory effects against Rheumatoid arthritis (RA) using activated macrophages (Raw246.7) in vitro and adjuvant arthritis rats (AA) in vivo. The results demonstrated that CA significantly reduced synovial inflammation in AA rats, possibly due to suppression of the expressions of pro-inflammatory cytokines, especially the IL-1β. Further investigation found that CA also suppressed the activity of HIF-1α by inhibiting the accumulation of succinate in cytoplasm. As we know, the reduction of HIF-1α nucleation slows down IL-1β production, because HIF-1α activates the expression of NLRP3, which is involved in the assembly of inflammasome and processing of IL-1β. In addition, CA also inhibited the expression of the succinate receptor GPR91, which in turn inhibited the activation of HIF-1α. In conclusions, our results suggested that CA might be a potential therapeutic compound to relieve rheumatoid arthritis progress by suppressing IL-1β through modulating succinate/HIF-1α axis and inhibition of NLRP3.
Collapse
Affiliation(s)
- Panwang Liu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jie Wang
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ting Pan
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Huan Chen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ying Fu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Fushun Wang
- Institute of Brain and Psyhological Sciences, Sichuan Normal University, Chengdu, Sichuan 610000, China
| | - Jason H Huang
- Department of Neurosurgery, Balor Scott & White Health Science Center, Temple, TX, United States; Department of Surgery, Texas A&M University College of Medicine, Temple, TX 79409, United States
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| |
Collapse
|
224
|
Inhibition of GPR91 Reduces Inflammatory Mediators Involved in Active Labor in Myometrium. Mediators Inflamm 2020; 2020:6454282. [PMID: 32377163 PMCID: PMC7180404 DOI: 10.1155/2020/6454282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/25/2020] [Accepted: 03/12/2020] [Indexed: 12/29/2022] Open
Abstract
Results GPR91 mRNA expression was significantly higher in myometrium from women during term spontaneous labor compared to no labor. Likewise, in mice, GPR91 mRNA expression was significantly upregulated in myometrium during inflammation-induced preterm labor compared to preterm no labor. In myometrial cells, IL1B and TNF significantly increased GPR91 mRNA expression. Knockdown of GPR91 by siRNA in myometrial cells significantly suppressed the secretion and/or expression of IL1B- and TNF-induced proinflammatory cytokines (GM-CSF, IL1A, IL1B, and IL6) and chemokines (CXCL8 and CCL2), myometrial contractility (expression of the contraction-associated proteins PTGFR and CX43, secretion of the uterotonic PGF2α, and in situ collagen gel contraction), and the transcription factor NF-κB. Conclusion Our findings demonstrate that GPR91 is involved in the genesis of proinflammatory and prolabor mediators induced by IL1B or TNF and collectively suggest that GPR91 may contribute to augmentation of the labor processes.
Collapse
|
225
|
Burja B, Mertelj T, Frank-Bertoncelj M. Hi- JAKi-ng Synovial Fibroblasts in Inflammatory Arthritis With JAK Inhibitors. Front Med (Lausanne) 2020; 7:124. [PMID: 32432116 PMCID: PMC7214667 DOI: 10.3389/fmed.2020.00124] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022] Open
Abstract
The Janus kinase (JAK)-Signal transducer and activator of transcription (STAT) pathway is one of the central signaling hubs in inflammatory, immune and cancer cells. Inhibiting the JAK-STAT pathway with JAK inhibitors (jakinibs) constitutes an important therapeutic strategy in cancer and chronic inflammatory diseases like rheumatoid arthritis (RA). FDA has approved different jakinibs for the treatment of RA, including tofacitinib, baricitinib and upadacitinib, and several jakinibs are being tested in clinical trials. Here, we reviewed published studies of jakinib effects on resolving synovial pathology in inflammatory arthritis. We discussed the results of jakinibs on structural joint damage in clinical trials and explored the effects of jakinibs across different in vitro, ex vivo, and in vivo synovial experimental models. We delved rigorously into experimental designs of in vitro fibroblast studies, deconvoluted jakinib efficacy in synovial fibroblasts across diverse experimental conditions and discussed their translatability in vivo. Synovial fibroblasts can readily activate the JAK-STAT signaling pathway in response to cytokine stimulation. We highlighted rather limited effects of jakinibs on the in vitro cultured synovial fibroblasts and inferred that direct and indirect (immune cell-dependent) actions of jakinibs are required to curb the fibroblast pathology in vivo. These actions have not been mimicked optimally in current in vitro experimental designs, where inflammatory stimuli do not naturally clear out with treatment as they do in vivo. While summarizing the broad knowledge of synovial jakinib effects, our review uniquely challenges future study designs to better mimick the jakinib actions in broader cell communities, as occurring in vivo in the inflamed synovium. This can deepen our understanding of collective synovial activities of jakinibs and their therapeutic limitations, thereby fostering jakinib development in arthritis.
Collapse
Affiliation(s)
- Blaž Burja
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland.,Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tonja Mertelj
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland
| | | |
Collapse
|
226
|
Succinate Activates EMT in Intestinal Epithelial Cells through SUCNR1: A Novel Protagonist in Fistula Development. Cells 2020; 9:cells9051104. [PMID: 32365557 PMCID: PMC7290938 DOI: 10.3390/cells9051104] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/15/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of Crohn’s disease-associated fibrostenosis and fistulas imply the epithelial-to-mesenchymal transition (EMT) process. As succinate and its receptor (SUCNR1) are involved in intestinal inflammation and fibrosis, we investigated their relevance in EMT and Crohn’s disease (CD) fistulas. Succinate levels and SUCNR1-expression were analyzed in intestinal resections from non-Inflammatory Bowel Disease (non-IBD) subjects and CD patients with stenosing-B2 or penetrating-B3 complications and in a murine heterotopic-transplant model of intestinal fibrosis. EMT, as increased expression of Snail1, Snail2 and vimentin and reduction in E-cadherin, was analyzed in tissues and succinate-treated HT29 cells. The role played by SUCNR1 was studied by silencing its gene. Succinate levels and SUCNR1 expression are increased in B3-CD patients and correlate with EMT markers. SUCNR1 is detected in transitional cells lining the fistula tract and in surrounding mesenchymal cells. Grafts from wild type (WT) mice present increased succinate levels, SUCNR1 up-regulation and EMT activation, effects not observed in SUCNR1−/− tissues. SUCNR1 activation induces the expression of Wnt ligands, activates WNT signaling and induces a WNT-mediated EMT in HT29 cells. In conclusion, succinate and its receptor are up-regulated around CD-fistulas and activate Wnt signaling and EMT in intestinal epithelial cells. These results point to SUCNR1 as a novel pharmacological target for fistula prevention.
Collapse
|
227
|
Molecular mechanisms and systemic targeting of NRF2 dysregulation in cancer. Biochem Pharmacol 2020; 177:114002. [PMID: 32360363 DOI: 10.1016/j.bcp.2020.114002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
NF-E2-related factor 2 (NRF2) is a master regulator of redox homeostasis and provides cellular protection against oxidants and electrophiles by inducing the expression of a wide array of phase II cytoprotective genes. Until now, a number of NRF2 activators have been developed for treatment of chronic diseases and some are under evaluation in the clinical studies. On the other hand, accumulating evidence indicates that NRF2 confers chemoresistance and radioresistance, and its expression is correlated with poor prognosis in cancer patients. Studies in the last decade demonstrate that diverse mechanisms such as somatic mutations, accumulation of KEAP1 binding proteins, transcriptional dysregulation, oncogene activation, and accumulation of reactive metabolites contribute to NRF2 activation in cancer. In the present review, we illustrate the molecular mechanisms governing the function of NRF2 and explain how they are hijacked in cancer. We also provide some examples of NRF2 inhibitors together with a brief explanation of their mechanisms of action.
Collapse
|
228
|
Martin K, Touil R, Cvijetic G, Israel L, Kolb Y, Sarret S, Valeaux S, Degl'Innocenti E, Le Meur T, Caesar N, Bardet M, Beerli C, Zerwes HG, Kovarik J, Beltz K, Schlapbach A, Quancard J, Régnier CH, Bigaud M, Junt T, Wieczorek G, Isnardi I, Littlewood-Evans A, Bornancin F, Calzascia T. Requirement of Mucosa-Associated Lymphoid Tissue Lymphoma Translocation Protein 1 Protease Activity for Fcγ Receptor-Induced Arthritis, but Not Fcγ Receptor-Mediated Platelet Elimination, in Mice. Arthritis Rheumatol 2020; 72:919-930. [PMID: 31943941 DOI: 10.1002/art.41204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases. METHODS Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP). RESULTS MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP. CONCLUSION Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders.
Collapse
Affiliation(s)
- Kea Martin
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ratiba Touil
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Laura Israel
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Yeter Kolb
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Sophie Sarret
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Thomas Le Meur
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nadja Caesar
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Maureen Bardet
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Jiri Kovarik
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Karen Beltz
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Jean Quancard
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Marc Bigaud
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
229
|
Abstract
A striking change has happened in the field of immunology whereby specific metabolic processes have been shown to be a critical determinant of immune cell activation. Multiple immune receptor types rewire metabolic pathways as a key part of how they promote effector functions. Perhaps surprisingly for immunologists, the Krebs cycle has emerged as the central immunometabolic hub of the macrophage. During proinflammatory macrophage activation, there is an accumulation of the Krebs cycle intermediates succinate and citrate, and the Krebs cycle–derived metabolite itaconate. These metabolites have distinct nonmetabolic signaling roles that influence inflammatory gene expression. A key bioenergetic target for the Krebs cycle, the electron transport chain, also becomes altered, generating reactive oxygen species from Complexes I and III. Similarly, alternatively activated macrophages require α-ketoglutarate-dependent epigenetic reprogramming to elicit anti-inflammatory gene expression. In this review, we discuss these advances and speculate on the possibility of targeting these events therapeutically for inflammatory diseases.
Collapse
Affiliation(s)
- Dylan G. Ryan
- School of Biochemistry and Immunology and Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Luke A.J. O'Neill
- School of Biochemistry and Immunology and Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| |
Collapse
|
230
|
Stathopoulou C, Nikoleri D, Bertsias G. Immunometabolism: an overview and therapeutic prospects in autoimmune diseases. Immunotherapy 2020; 11:813-829. [PMID: 31120393 DOI: 10.2217/imt-2019-0002] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metabolism is a critical immune regulator under physiologic and pathologic conditions. Culminating evidence has disentangled the contribution of distinct metabolic pathways, namely glucolysis, pentose phosphate, fatty acid oxidation, glutaminolysis, Krebs cycle and oxidative phosphorylation, in modulating innate and adaptive immune cells based on their activation/differentiation state. Metabolic aberrations and changes in the intracellular levels of specific metabolites are linked to the inflammatory phenotype of immune cells implicated in autoimmune disorders such as systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis and diabetes. Notably, targeting metabolism such as the mTOR by rapamycin, hexokinase by 2-deoxy-D-glucose, AMP-activated protein kinase by metformin, may be used to ameliorate autoimmune inflammation. Accordingly, research in immunometabolism is expected to offer novel opportunities for monitoring and treating immune-mediated diseases.
Collapse
Affiliation(s)
- Chrysoula Stathopoulou
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - Dimitra Nikoleri
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - George Bertsias
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| |
Collapse
|
231
|
Scagliola A, Mainini F, Cardaci S. The Tricarboxylic Acid Cycle at the Crossroad Between Cancer and Immunity. Antioxid Redox Signal 2020; 32:834-852. [PMID: 31847530 DOI: 10.1089/ars.2019.7974] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: The tricarboxylic acid (TCA) cycle is a housekeeping metabolic pathway essential for generation of energy and biosynthetic intermediates. Alterations of the TCA cycle play a pivotal role in oncogenesis and inflammation. As such, some metabolic vulnerabilities, imposed by TCA cycle dysfunction in cancer, have been identified. Similarly, the TCA cycle appeared as an actionable pathway in immunopathologies. Recent Advances: Metabolic changes accompanying cell transformation have been usually considered as adaptive mechanisms to malignant transformation. The identification of oncogenic mutations in some TCA cycle enzymes changed this view, indicating altered mitochondrial metabolism as an instrumental mechanism for cancer initiation. Similarly, the observation that TCA cycle-derived metabolites have multiple signaling roles in immune cells supports the idea of this pathway as a metabolic rheostat of immune responses. Critical Issues: This review summarizes the crucial role of the TCA cycle in pathophysiology describing the post-translational and epigenetic impact of oncometabolites accumulation in cancer and immune cells. Future Directions: Additional studies will be necessary to further explore the role of oncometabolites in paracrine signaling and to identify genuine metabolic and nutritional liabilities imposed by TCA cycle dysfunction in cancer, hardly to be escaped by resistance mechanisms.
Collapse
Affiliation(s)
- Alessandra Scagliola
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Mainini
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simone Cardaci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
232
|
Pucino V, Certo M, Varricchi G, Marone G, Ursini F, Rossi FW, De Paulis A, Mauro C, Raza K, Buckley CD. Metabolic Checkpoints in Rheumatoid Arthritis. Front Physiol 2020; 11:347. [PMID: 32362840 PMCID: PMC7180190 DOI: 10.3389/fphys.2020.00347] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
Several studies have highlighted the interplay between metabolism, immunity and inflammation. Both tissue resident and infiltrating immune cells play a major role in the inflammatory process of rheumatoid arthritis (RA) via the production of cytokines, adipo-cytokines and metabolic intermediates. These functions are metabolically demanding and require the most efficient use of bioenergetic pathways. The synovial membrane is the primary site of inflammation in RA and exhibits distinctive histological patterns characterized by different metabolism, prognosis and response to treatment. In the RA synovium, the high energy demand by stromal and infiltrating immune cells, causes the accumulation of metabolites, and adipo-cytokines, which carry out signaling functions, as well as activating transcription factors which act as metabolic sensors. These events drive immune and joint-resident cells to acquire pro-inflammatory effector functions which in turn perpetuate chronic inflammation. Whether metabolic changes are a consequence of the disease or one of the causes of RA pathogenesis is still under investigation. This review covers our current knowledge of cell metabolism in RA. Understanding the intricate interactions between metabolic pathways and the inflammatory and immune responses will provide more awareness of the mechanisms underlying RA pathogenesis and will identify novel therapeutic options to treat this disease.
Collapse
Affiliation(s)
- Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy
- Ospedale dei Colli, Hospital Pharmacy, Naples, Italy
| | - Francesco Ursini
- Section of Rheumatology, Department of Biomedical and Neuromotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Amato De Paulis
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Karim Raza
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Research into Inflammatory Arthritis Centre Versus Arthritis, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, United Kingdom
- MRC and Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Christopher Dominic Buckley
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
233
|
Coras R, Murillo-Saich JD, Guma M. Circulating Pro- and Anti-Inflammatory Metabolites and Its Potential Role in Rheumatoid Arthritis Pathogenesis. Cells 2020; 9:E827. [PMID: 32235564 PMCID: PMC7226773 DOI: 10.3390/cells9040827] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that affects synovial joints, leading to inflammation, joint destruction, loss of function, and disability. Although recent pharmaceutical advances have improved the treatment of RA, patients often inquire about dietary interventions to improve RA symptoms, as they perceive pain and/or swelling after the consumption or avoidance of certain foods. There is evidence that some foods have pro- or anti-inflammatory effects mediated by diet-related metabolites. In addition, recent literature has shown a link between diet-related metabolites and microbiome changes, since the gut microbiome is involved in the metabolism of some dietary ingredients. But diet and the gut microbiome are not the only factors linked to circulating pro- and anti-inflammatory metabolites. Other factors including smoking, associated comorbidities, and therapeutic drugs might also modify the circulating metabolomic profile and play a role in RA pathogenesis. This article summarizes what is known about circulating pro- and anti-inflammatory metabolites in RA. It also emphasizes factors that might be involved in their circulating concentrations and diet-related metabolites with a beneficial effect in RA.
Collapse
Affiliation(s)
- Roxana Coras
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; (R.C.); (J.D.M.-S.)
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
| | - Jessica D. Murillo-Saich
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; (R.C.); (J.D.M.-S.)
| | - Monica Guma
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; (R.C.); (J.D.M.-S.)
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
234
|
Comito G, Ippolito L, Chiarugi P, Cirri P. Nutritional Exchanges Within Tumor Microenvironment: Impact for Cancer Aggressiveness. Front Oncol 2020; 10:396. [PMID: 32266157 PMCID: PMC7105815 DOI: 10.3389/fonc.2020.00396] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Neoplastic tissues are composed not only by tumor cells but also by several non-transformed stromal cells, such as cancer-associated fibroblasts, endothelial and immune cells, that actively participate to tumor progression. Starting from the very beginning of carcinogenesis, tumor cells, through the release of paracrine soluble factors and vesicles, i.e., exosomes, modify the behavior of the neighboring cells, so that they can give efficient support for cancer cell proliferation and spreading. A mandatory role in tumor progression has been recently acknowledged to metabolic deregulation. Beside undergoing a metabolic reprogramming coherent to their high proliferation rate, tumor cells also rewire the metabolic assets of their stromal cells, educating them to serve as nutrient donors. Hence, an alteration in the composition and in the flow rate of many nutrients within tumor microenvironment has been associated with malignancy progression. This review is focused on metabolic remodeling of the different cell populations within tumor microenvironment, dealing with reciprocal re-education through the symbiotic sharing of metabolites, behaving both as nutrients and as transcriptional regulators, describing their impact on tumor growth and metastasis.
Collapse
Affiliation(s)
- Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.,Excellence Center for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| |
Collapse
|
235
|
Riquelme SA, Wong Fok Lung T, Prince A. Pulmonary Pathogens Adapt to Immune Signaling Metabolites in the Airway. Front Immunol 2020; 11:385. [PMID: 32231665 PMCID: PMC7082326 DOI: 10.3389/fimmu.2020.00385] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
A limited number of pulmonary pathogens are able to evade normal mucosal defenses to establish acute infection and then adapt to cause chronic pneumonias. Pathogens, such as Pseudomonas aeruginosa or Staphylococcus aureus, are typically associated with infection in patients with underlying pulmonary disease or damage, such as cystic fibrosis (CF) or chronic obstructive pulmonary disease (COPD). To establish infection, bacteria express a well-defined set of so-called virulence factors that facilitate colonization and activate an immune response, gene products that have been identified in murine models. Less well-understood are the adaptive changes that occur over time in vivo, enabling the organisms to evade innate and adaptive immune clearance mechanisms. These colonizers proliferate, generating a population sufficient to provide selection for mutants, such as small colony variants and mucoid variants, that are optimized for long term infection. Such host-adapted strains have evolved in response to selective pressure such as antibiotics and the recruitment of phagocytes at sites of infection and their release of signaling metabolites (e.g., succinate). These metabolites can potentially function as substrates for bacterial growth and but also generate oxidant stress. Whole genome sequencing and quantified expression of selected genes have helped to explain how P. aeruginosa and S. aureus adapt to the presence of these metabolites over the course of in vivo infection. The serial isolation of clonally related strains from patients with cystic fibrosis has provided the opportunity to identify bacterial metabolic pathways that are altered under this immune pressure, such as the anti-oxidant glyoxylate and pentose phosphate pathways, routes contributing to the generation of biofilms. These metabolic pathways and biofilm itself enable the organisms to dissipate oxidant stress, while providing protection from phagocytosis. Stimulation of host immune signaling metabolites by these pathogens drives bacterial adaptation and promotes their persistence in the airways. The inherent metabolic flexibility of P. aeruginosa and S. aureus is a major factor in their success as pulmonary pathogens.
Collapse
Affiliation(s)
- Sebastián A Riquelme
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Tania Wong Fok Lung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Alice Prince
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
236
|
Succinate Supplement Elicited "Pseudohypoxia" Condition to Promote Proliferation, Migration, and Osteogenesis of Periodontal Ligament Cells. Stem Cells Int 2020; 2020:2016809. [PMID: 32215014 PMCID: PMC7085835 DOI: 10.1155/2020/2016809] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/01/2020] [Accepted: 02/25/2020] [Indexed: 01/03/2023] Open
Abstract
Most mesenchymal stem cells reside in a niche of low oxygen tension. Iron-chelating agents such as CoCl2 and deferoxamine have been utilized to mimic hypoxia and promote cell growth. The purpose of the present study was to explore whether a supplement of succinate, a natural metabolite of the tricarboxylic acid (TCA) cycle, can mimic hypoxia condition to promote human periodontal ligament cells (hPDLCs). Culturing hPDLCs in hypoxia condition promoted cell proliferation, migration, and osteogenic differentiation; moreover, hypoxia shifted cell metabolism from oxidative phosphorylation to glycolysis with accumulation of succinate in the cytosol and its release into culture supernatants. The succinate supplement enhanced hPDLC proliferation, migration, and osteogenesis with decreased succinate dehydrogenase (SDH) expression and activity, as well as increased hexokinase 2 (HK2) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), suggesting metabolic reprogramming from oxidative phosphorylation to glycolysis in a normal oxygen condition. The succinate supplement in cell cultures promoted intracellular succinate accumulation while stabilizing hypoxia inducible factor-1α (HIF-1α), leading to a state of pseudohypoxia. Moreover, we demonstrate that hypoxia-induced proliferation was G-protein-coupled receptor 91- (GPR91-) dependent, while exogenous succinate-elicited proliferation involved the GPR91-dependent and GPR91-independent pathway. In conclusion, the succinate supplement altered cell metabolism in hPDLCs, induced a pseudohypoxia condition, and enhanced proliferation, migration, and osteogenesis of mesenchymal stem cells in vitro.
Collapse
|
237
|
Castegna A, Gissi R, Menga A, Montopoli M, Favia M, Viola A, Canton M. Pharmacological targets of metabolism in disease: Opportunities from macrophages. Pharmacol Ther 2020; 210:107521. [PMID: 32151665 DOI: 10.1016/j.pharmthera.2020.107521] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.
Collapse
Affiliation(s)
- Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| |
Collapse
|
238
|
Høgh RI, Droujinine A, Møller SH, Jepsen SD, Mellergaard M, Andresen L, Skov S. Fumarate Upregulates Surface Expression of ULBP2/ULBP5 by Scavenging Glutathione Antioxidant Capacity. THE JOURNAL OF IMMUNOLOGY 2020; 204:1746-1759. [PMID: 32144161 DOI: 10.4049/jimmunol.1900740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/26/2020] [Indexed: 12/15/2022]
Abstract
Fumarate is a tricarboxylic acid cycle metabolite whose intracellular accumulation is linked to inflammatory signaling and development of cancer. In this study, we demonstrate that endogenous fumarate accumulation upregulates surface expression of the immune stimulatory NK group 2, member D (NKG2D) ligands ULBP2 and ULBP5. In agreement with this, accumulation of fumarate by the therapeutic drug dimethyl fumarate (DMF) also promotes ULBP2/5 surface expression. Mechanistically, we found that the increased ULBP2/5 expression was dependent on oxidative stress and the antioxidants N-acetylcysteine and glutathione (GSH) abrogated ULBP2/5 upregulated by DMF. Fumarate can complex with GSH and thereby exhaust cells of functional GSH capacity. In line with this, inhibition of GSH reductase (GR), the enzyme responsible for GSH recycling, promoted ULBP2/5 surface expression. Loss of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) associates with a malignant form of renal cancer characterized by fumarate accumulation and increased production of reactive oxygen species, highlighting fumarate as an oncometabolite. Interestingly, FH-deficient renal cancer cells had low surface expression of ULBP2/5 and were unresponsive to DMF treatment, suggesting that the fumarate-stimulating ULBP2/5 pathway is abrogated in these cells as an immune-evasive strategy. Together, our data show that ULBP2/5 expression can be upregulated by accumulation of fumarate, likely by depleting cells of GSH antioxidant capacity. Given that DMF is an approved human therapeutic drug, our findings support a broader use of DMF in treatment of cancers and inflammatory conditions.
Collapse
Affiliation(s)
- Rikke Illum Høgh
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alec Droujinine
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sofie Hedlund Møller
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stine Dam Jepsen
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Mellergaard
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lars Andresen
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Søren Skov
- Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
239
|
Targeting immunometabolism as an anti-inflammatory strategy. Cell Res 2020; 30:300-314. [PMID: 32132672 PMCID: PMC7118080 DOI: 10.1038/s41422-020-0291-z] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
The growing field of immunometabolism has taught us how metabolic cellular reactions and processes not only provide a means to generate ATP and biosynthetic precursors, but are also a way of controlling immunity and inflammation. Metabolic reprogramming of immune cells is essential for both inflammatory as well as anti-inflammatory responses. Four anti-inflammatory therapies, DMF, Metformin, Methotrexate and Rapamycin all work by affecting metabolism and/or regulating or mimicking endogenous metabolites with anti-inflammatory effects. Evidence is emerging for the targeting of specific metabolic events as a strategy to limit inflammation in different contexts. Here we discuss these recent developments and speculate on the prospect of targeting immunometabolism in the effort to develop novel anti-inflammatory therapeutics. As accumulating evidence for roles of an intricate and elaborate network of metabolic processes, including lipid, amino acid and nucleotide metabolism provides key focal points for developing new therapies, we here turn our attention to glycolysis and the TCA cycle to provide examples of how metabolic intermediates and enzymes can provide potential novel therapeutic targets.
Collapse
|
240
|
Guo Y, Cho SW, Saxena D, Li X. Multifaceted Actions of Succinate as a Signaling Transmitter Vary with Its Cellular Locations. Endocrinol Metab (Seoul) 2020; 35:36-43. [PMID: 32207262 PMCID: PMC7090288 DOI: 10.3803/enm.2020.35.1.36] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 01/05/2023] Open
Abstract
Since the identification of succinate's receptor in 2004, studies supporting the involvement of succinate signaling through its receptor in various diseases have accumulated and most of these investigations have highlighted succinate's pro-inflammatory role. Taken with the fact that succinate is an intermediate metabolite in the center of mitochondrial activity, and considering its potential regulation of protein succinylation through succinyl-coenzyme A, a review on the overall multifaceted actions of succinate to discuss whether and how these actions relate to the cellular locations of succinate is much warranted. Mechanistically, it is important to consider the sources of succinate, which include somatic cellular released succinate and those produced by the microbiome, especially the gut microbiota, which is an equivalent, if not greater contributor of succinate levels in the body. Continue learning the critical roles of succinate signaling, known and unknown, in many pathophysiological conditions is important. Furthermore, studies to delineate the regulation of succinate levels and to determine how succinate elicits various types of signaling in a temporal and spatial manner are also required.
Collapse
Affiliation(s)
- Yuqi Guo
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
- Perlmutter Cancer Institute, New York, NY, USA
| | - Xin Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
- Perlmutter Cancer Institute, New York, NY, USA
- Department of Urology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
241
|
Zhao H, Raines LN, Huang SCC. Carbohydrate and Amino Acid Metabolism as Hallmarks for Innate Immune Cell Activation and Function. Cells 2020; 9:cells9030562. [PMID: 32121028 PMCID: PMC7140477 DOI: 10.3390/cells9030562] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Immune activation is now understood to be fundamentally linked to intrinsic and/or extrinsic metabolic processes which are essential for immune cells to survive, proliferate, and perform their effector functions. Moreover, disruption or dysregulation of these pathways can result in detrimental outcomes and underly a number of pathologies in both communicable and non-communicable diseases. In this review, we discuss how the metabolism of carbohydrates and amino acids in particular can modulate innate immunity and how perturbations in these pathways can result in failure of these immune cells to properly function or induce unfavorable phenotypes.
Collapse
Affiliation(s)
- Haoxin Zhao
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (H.Z.); (L.N.R.)
| | - Lydia N. Raines
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (H.Z.); (L.N.R.)
| | - Stanley Ching-Cheng Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (H.Z.); (L.N.R.)
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-368-3909
| |
Collapse
|
242
|
Wentzel AS, Janssen JJE, de Boer VCJ, van Veen WG, Forlenza M, Wiegertjes GF. Fish Macrophages Show Distinct Metabolic Signatures Upon Polarization. Front Immunol 2020; 11:152. [PMID: 32158446 PMCID: PMC7052297 DOI: 10.3389/fimmu.2020.00152] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/21/2020] [Indexed: 01/09/2023] Open
Abstract
Macrophages play important roles in conditions ranging from host immune defense to tissue regeneration and polarize their functional phenotype accordingly. Next to differences in the use of L-arginine and the production of different cytokines, inflammatory M1 macrophages and anti-inflammatory M2 macrophages are also metabolically distinct. In mammals, M1 macrophages show metabolic reprogramming toward glycolysis, while M2 macrophages rely on oxidative phosphorylation to generate energy. The presence of polarized functional immune phenotypes conserved from mammals to fish led us to hypothesize that a similar metabolic reprogramming in polarized macrophages exists in carp. We studied mitochondrial function of M1 and M2 carp macrophages under basal and stressed conditions to determine oxidative capacity by real-time measurements of oxygen consumption and glycolytic capacity by measuring lactate-based acidification. In M1 macrophages, we found increased nitric oxide production and irg1 expression in addition to altered oxidative phosphorylation and glycolysis. In M2 macrophages, we found increased arginase activity, and both oxidative phosphorylation and glycolysis were similar to control macrophages. These results indicate that M1 and M2 carp macrophages show distinct metabolic signatures and indicate that metabolic reprogramming may occur in carp M1 macrophages. This immunometabolic reprogramming likely supports the inflammatory phenotype of polarized macrophages in teleost fish such as carp, similar to what has been shown in mammals.
Collapse
Affiliation(s)
- Annelieke S Wentzel
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Joëlle J E Janssen
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands.,Human and Animal Physiology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Vincent C J de Boer
- Human and Animal Physiology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Wouter G van Veen
- Experimental Zoology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Maria Forlenza
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Geert F Wiegertjes
- Aquaculture and Fisheries Group, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
243
|
Zezina E, Sercan‐Alp O, Herrmann M, Biesemann N. Glucose transporter 1 in rheumatoid arthritis and autoimmunity. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1483. [DOI: 10.1002/wsbm.1483] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Ekaterina Zezina
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Oezen Sercan‐Alp
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Matthias Herrmann
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Nadine Biesemann
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| |
Collapse
|
244
|
|
245
|
Koenis DS, Medzikovic L, van Loenen PB, van Weeghel M, Huveneers S, Vos M, Evers-van Gogh IJ, Van den Bossche J, Speijer D, Kim Y, Wessels L, Zelcer N, Zwart W, Kalkhoven E, de Vries CJ. Nuclear Receptor Nur77 Limits the Macrophage Inflammatory Response through Transcriptional Reprogramming of Mitochondrial Metabolism. Cell Rep 2020; 24:2127-2140.e7. [PMID: 30134173 PMCID: PMC6113932 DOI: 10.1016/j.celrep.2018.07.065] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 04/20/2018] [Accepted: 07/18/2018] [Indexed: 11/25/2022] Open
Abstract
Activation of macrophages by inflammatory stimuli induces reprogramming of mitochondrial metabolism to support the production of pro-inflammatory cytokines and nitric oxide. Hallmarks of this metabolic rewiring are downregulation of α-ketoglutarate formation by isocitrate dehydrogenase (IDH) and accumulation of glutamine-derived succinate, which enhances the inflammatory response via the activity of succinate dehydrogenase (SDH). Here, we identify the nuclear receptor Nur77 (Nr4a1) as a key upstream transcriptional regulator of this pro-inflammatory metabolic switch in macrophages. Nur77-deficient macrophages fail to downregulate IDH expression and accumulate higher levels of succinate and other TCA cycle-derived metabolites in response to inflammatory stimulation in a glutamine-independent manner. Consequently, these macrophages produce more nitric oxide and pro-inflammatory cytokines in an SDH-dependent manner. In vivo, bone marrow Nur77 deficiency exacerbates atherosclerosis development and leads to increased circulating succinate levels. In summary, Nur77 induces an anti-inflammatory metabolic state in macrophages that protects against chronic inflammatory diseases such as atherosclerosis. Genome-wide profiling indicates that Nur77 regulates macrophage mitochondrial metabolism Nur77 inhibits IDH expression and TCA cycle activity in inflammatory macrophages Nur77-deficient macrophages produce more nitric oxide and cytokines via SDH Nur77 deficiency increases circulating succinate levels and atherosclerosis in vivo
Collapse
Affiliation(s)
- Duco Steven Koenis
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Lejla Medzikovic
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Pieter Bas van Loenen
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Michel van Weeghel
- Amsterdam UMC, University of Amsterdam, Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Mariska Vos
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Ingrid Johanna Evers-van Gogh
- Molecular Cancer Research and Center for Molecular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Dave Speijer
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Yongsoo Kim
- Division of Molecular Pathology, the Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Noam Zelcer
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Wilbert Zwart
- Division of Molecular Pathology, the Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Eric Kalkhoven
- Molecular Cancer Research and Center for Molecular Medicine, University Medical Centre Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | - Carlie Jacoba de Vries
- Amsterdam UMC, University of Amsterdam, Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
246
|
Daemen S, Schilling JD. The Interplay Between Tissue Niche and Macrophage Cellular Metabolism in Obesity. Front Immunol 2020; 10:3133. [PMID: 32038642 PMCID: PMC6987434 DOI: 10.3389/fimmu.2019.03133] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is associated with the development of metabolic diseases such as type 2 diabetes and non-alcoholic fatty liver disease. The presence of chronic, low-grade inflammation appears to be an important mechanistic link between excess nutrients and clinical disease. The onset of these metabolic disorders coincides with changes in the number and phenotype of macrophages in peripheral organs, particularly in the liver and adipose tissue. Macrophage accumulation in these tissues has been implicated in tissue inflammation and fibrosis, contributing to metabolic disease progression. Recently, the concept has emerged that changes in macrophage metabolism affects their functional phenotype, possibly triggered by distinct environmental metabolic cues. This may be of particular importance in the setting of obesity, where both liver and adipose tissue are faced with a high metabolic burden. In the first part of this review we will discuss current knowledge regarding macrophage dynamics in both adipose tissue and liver in obesity. Then in the second part, we will highlight data linking macrophage metabolism to functional phenotype with an emphasis on macrophage activation in metabolic disease. The importance of understanding how tissue niche influences macrophage function in obesity will be highlighted. In addition, we will identify important knowledge gaps and outstanding questions that are relevant for future research in this area and will facilitate the identification of novel targets for therapeutic intervention in associated metabolic diseases.
Collapse
Affiliation(s)
- Sabine Daemen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Joel D Schilling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
247
|
Jones AE, Divakaruni AS. Macrophage activation as an archetype of mitochondrial repurposing. Mol Aspects Med 2020; 71:100838. [PMID: 31954522 DOI: 10.1016/j.mam.2019.100838] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
Mitochondria are metabolic organelles essential not only for energy transduction, but also a range of other functions such as biosynthesis, ion and metal homeostasis, maintenance of redox balance, and cell signaling. A hallmark example of how mitochondria can rebalance these processes to adjust cell function is observed in macrophages. These innate immune cells are responsible for a remarkable breadth of processes including pathogen elimination, antigen presentation, debris clearance, and wound healing. These diverse, polarized functions often include similarly disparate alterations in the metabolic phenotype associated with their execution. In this chapter, mitochondrial bioenergetics and signaling are viewed through the lens of macrophage polarization: both classical, pro-inflammatory activation and alternative, anti-inflammatory activation are associated with substantive changes to mitochondrial metabolism. Emphasis is placed on recent evidence that aims to clarify the essential - rather than associative - mitochondrial alterations, as well as accumulating data suggesting a degree of plasticity within the metabolic phenotypes that can support pro- and anti-inflammatory functions.
Collapse
Affiliation(s)
- Anthony E Jones
- UCLA Department of Molecular and Medical Pharmacology, 650 Charles E. Young Drive, Los Angeles, CA, 90095, USA
| | - Ajit S Divakaruni
- UCLA Department of Molecular and Medical Pharmacology, 650 Charles E. Young Drive, Los Angeles, CA, 90095, USA.
| |
Collapse
|
248
|
Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun 2020; 11:102. [PMID: 31900386 PMCID: PMC6941980 DOI: 10.1038/s41467-019-13668-3] [Citation(s) in RCA: 1541] [Impact Index Per Article: 308.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are signaling organelles that regulate a wide variety of cellular functions and can dictate cell fate. Multiple mechanisms contribute to communicate mitochondrial fitness to the rest of the cell. Recent evidence confers a new role for TCA cycle intermediates, generally thought to be important for biosynthetic purposes, as signaling molecules with functions controlling chromatin modifications, DNA methylation, the hypoxic response, and immunity. This review summarizes the mechanisms by which the abundance of different TCA cycle metabolites controls cellular function and fate in different contexts. We will focus on how these metabolites mediated signaling can affect physiology and disease. Mitochondrial metabolites contribute to more than biosynthesis, and it is clear that they influence multiple cellular functions in a variety of ways. Here, Martínez-Reyes and Chandel review key metabolites and describe their effects on processes involved in physiology and disease including chromatin dynamics, immunity, and hypoxia.
Collapse
|
249
|
Baihu Jia Guizhi Decoction Improves Rheumatoid Arthritis Inflammation by Regulating Succinate/SUCNR1 Metabolic Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3258572. [PMID: 31949465 PMCID: PMC6948314 DOI: 10.1155/2019/3258572] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis. Succinate is an inflammatory metabolic signal that exacerbates RA synovitis by activating succinate receptor 1 (SUCNR1) to amplify the release of IL-1β. Thus, inhibition of succinate activation of SUCRN1 could be an effective method to inhibit the inflammation of RA. Baihu Jia Guizhi decoction (BHGZ), which is composed of Gypsum Fibrosum, Anemarrhena asphodeloides Bge., Cinnamomum cassia Presl., Glycyrrhiza uralensis Fisch., and Oryza sativa L., is a Traditional Chinese Medicine (TCM) prescription used to treat RA in clinic. In addition, TCM believes that damp and heat environment is one of the causes of RA. In this study, we tested the role of damp and heat environments in exacerbating RA inflammation and the anti-inflammatory effect of BHGZ, based on succinate/SUCNR1/IL-1β pathway in the adjuvant arthritis (AA) model with damp and heat environment (AA + DHE). Results showed that paw swelling and synovial pathology were significantly increased in AA rats, and these results were aggravated by stimulation in damp and heat environment. BHGZ improved AA + DHE rats' paw swelling, synovial hyperplasia, and inflammatory cell infiltration and reduced IL-1β. In addition, AA rats significantly increased the expression of SUCNR1, and the stimulation of damp and heat environment not only increased the expression of SUCNR1 but also promoted the accumulation of succinate. BHGZ simultaneously reduced the concentration of succinate and the expression of SUCNR1. Finally, SDH activity was decreased in AA rats and AA + DHE rats, while BHGZ increased SDH activity and then reduced succinate concentration. Therefore, we prove that damp and heat environment deteriorated the inflammation of RA which is the activation of succinate/SUCNR1 pathway, while BHGZ regulates SDH activity to reduce the accumulation of succinate and inhibit the activation of SUCNR1 that is the underlying mechanism of its treatment of RA.
Collapse
|
250
|
Interplay between the Adaptive Immune System and Insulin Resistance in Weight Loss Induced by Bariatric Surgery. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3940739. [PMID: 31885787 PMCID: PMC6925764 DOI: 10.1155/2019/3940739] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 11/05/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Low-grade chronic inflammation plays a pivotal role among other pathophysiological mechanisms involved in obesity. Innate and adaptive immune cells undergo systemic proinflammatory polarization that gives rise to an increased secretion of proinflammatory cytokines, which in turn leads to insulin resistance. Bariatric surgery is currently the most effective treatment for obesity, as it brings on significant weight loss, glucose metabolism improvement, and a decrease in systemic inflammation biomarkers. After bariatric surgery, several changes have been reported to occur in adaptive immunity, including reduction in CD4+ and CD8+ T cell counts, a decrease in the Th1/Th2 ratio, an increase in B regulatory cells, and reduction in proinflammatory cytokine secretion. Overall, there seems to be a major shift in several lymphocyte populations from a proinflammatory to an anti-inflammatory phenotype. Furthermore, increased antioxidant activity and reduced lipid and DNA oxidation products have been reported after bariatric surgery in circulating mononuclear cells. This paper highlights the shift in the adaptive immune system in response to weight loss and improved insulin sensitivity, as well as the interplay between immunological and metabolic adaptations as a result of bariatric surgery. Finally, based on data from research, we propose several mechanisms such as changes in adaptive immune cell phenotypes and their by-products, recruitment in adipose tissue, reduced oxidative stress, and modification in metabolic substrate availability as drivers to reduce low-grade chronic inflammation after bariatric surgery in severe obesity.
Collapse
|