101
|
Abstract
Succinate is a circulating metabolite, and the relationship between abnormal changes in the physiological concentration of succinate and inflammatory diseases caused by the overreaction of certain immune cells has become a research focus. Recent investigations have shown that succinate produced by the gut microbiota has the potential to regulate host homeostasis and treat diseases such as inflammation. Gut microbes are important for maintaining intestinal homeostasis. Microbial metabolites serve as nutrients in energy metabolism, and act as signal molecules that stimulate host cell and organ function and affect the structural balance between symbiotic gut microorganisms. This review focuses on succinate as a metabolite of both host cells and gut microbes and its involvement in regulating the gut - immune tissue axis by activating intestinal mucosal cells, including macrophages, dendritic cells, and intestinal epithelial cells. We also examined its role as the mediator of microbiota - host crosstalk and its potential function in regulating intestinal microbiota homeostasis. This review explores feasible ways to moderate succinate levels and provides new insights into succinate as a potential target for microbial therapeutics for humans.
Collapse
Affiliation(s)
- Yi-Han Wei
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiang-Chao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| |
Collapse
|
102
|
Lee JL, Zhang C, Westbrook R, Gabrawy MM, Nidadavolu L, Yang H, Marx R, Wu Y, Anders NM, Ma L, Bichara MD, Kwak MJ, Buta B, Khadeer M, Yenokyan G, Tian J, Xue QL, Siragy HM, Carey RM, de Cabo R, Ferrucci L, Moaddel R, Rudek MA, Le A, Walston JD, Abadir PM. Serum Concentrations of Losartan Metabolites Correlate With Improved Physical Function in a Pilot Study of Prefrail Older Adults. J Gerontol A Biol Sci Med Sci 2022; 77:2356-2366. [PMID: 35511890 PMCID: PMC9799219 DOI: 10.1093/gerona/glac102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 01/20/2023] Open
Abstract
Losartan is an oral antihypertensive agent that is rapidly metabolized to EXP3174 (angiotensin-subtype-1-receptor blocker) and EXP3179 (peroxisome proliferator-activated receptor gamma [PPARγ] agonist), which was shown in animal studies to reduce inflammation, enhance mitochondrial energetics, and improve muscle repair and physical performance. We conducted an exploratory pilot study evaluating losartan treatment in prefrail older adults (age 70-90 years, N = 25). Participants were randomized to control (placebo) or treatment (daily oral losartan beginning at 25 mg per day and increasing every 8 weeks) for a total of 6 months. Fatigue, hyperkalemia, and hypotension were the most observed side effects of losartan treatment. Participants in the losartan group had an estimated 89% lower odds of frailty (95% confidence interval [CI]: 18% to 99% lower odds, p = .03), with a 0.3-point lower frailty score than the placebo group (95% CI: 0.01-0.5 lower odds, p = .04). Frailty score was also negatively associated with serum losartan and EXP3179 concentrations. For every one standard deviation increase in EXP3179 (ie, 0.0011 ng/μL, based on sample values above detection limit) and EXP3174 (ie, 0.27 ng/μL, based on sample values above detection limit), there was a 0.0035 N (95% CI: 0.0019-0.0051, p < .001) and a 0.0027 N (95% CI: 0.00054-0.0043, p = .007) increase in average knee strength, respectively.
Collapse
Affiliation(s)
- Jessica L Lee
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland,USA
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Cissy Zhang
- Department of Oncology, Division of Cancer Chemical and Structural Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Reyhan Westbrook
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mariann M Gabrawy
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lolita Nidadavolu
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Huanle Yang
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruth Marx
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yuqiong Wu
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole M Anders
- Department of Oncology, Division of Cancer Chemical and Structural Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Analytical Pharmacology Core Laboratory, Clinical Pharmacology, Baltimore, MD, USA
| | - Lina Ma
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, China National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Marcela-Dávalos Bichara
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Min-Ji Kwak
- Department of Internal Medicine, Division of Geriatric and Palliative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Brian Buta
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammed Khadeer
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Gayane Yenokyan
- Johns Hopkins Biostatistics Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jing Tian
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qian-Li Xue
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Helmy M Siragy
- Department of Medicine, Division of Endocrine and Metabolism, University of Virginia, Charlottesville, Virginia, USA
| | - Robert M Carey
- Department of Medicine, Division of Endocrine and Metabolism, University of Virginia, Charlottesville, Virginia, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Ruin Moaddel
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Michelle A Rudek
- The Johns Hopkins Analytical Pharmacology Core Laboratory, Clinical Pharmacology, Baltimore, MD, USA
- Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anne Le
- The Johns Hopkins Analytical Pharmacology Core Laboratory, Clinical Pharmacology, Baltimore, MD, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy D Walston
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter M Abadir
- Department of Medicine, Division of Geriatric Medicine and Gerontology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
103
|
Dai X, Zhou Y, Han F, Li J. Succinylation and redox status in cancer cells. Front Oncol 2022; 12:1081712. [PMID: 36605449 PMCID: PMC9807787 DOI: 10.3389/fonc.2022.1081712] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Succinylation is a post-translational modification (PTM) event that associates metabolic reprogramming with various pathological disorders including cancers via transferring a succinyl group to a residue of the target protein in an enzymic or non-enzymic manner. With our incremental knowledge on the roles of PTM played in tumor initiation and progression, relatively little has been focused on succinylation and its clinical implications. By delineating the associations of succinylation with cancer hallmarks, we identify the, in general, promotive roles of succinylation in manifesting cancer hallmarks, and conceptualize two working modes of succinylation in driving oncogenic signaling, i.e., via altering the structure and charge of target proteins towards enhanced stability and activity. We also characterize succinylation as a reflection of cellular redox homeostatic status and metabolic state, and bring forth the possible use of hyper-succinylated genome for early cancer diagnosis or disease progression indication. In addition, we propose redox modulation tools such as cold atmospheric plasma as a promising intervention approach against tumor cells and cancer stemness via targeting the redox homeostatic environment cells established under a pathological condition such as hypoxia. Taken together, we emphasize the central role of succinylation in bridging the gap between cellular metabolism and redox status, and its clinical relevance as a mark for cancer diagnosis as well as a target in onco-therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China,*Correspondence: Xiaofeng Dai, ; Jitian Li,
| | - Yanyan Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fei Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China,*Correspondence: Xiaofeng Dai, ; Jitian Li,
| |
Collapse
|
104
|
Yao Z, Meng J, Long J, Li L, Qiu W, Li C, Zhang JV, Ren P. Orphan receptor GPR50 attenuates inflammation and insulin signaling in 3T3-L1 preadipocytes. FEBS Open Bio 2022; 13:89-101. [PMID: 36333974 PMCID: PMC9811602 DOI: 10.1002/2211-5463.13516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/06/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022] Open
Abstract
Type 2 diabetes (T2DM) is characterized by insulin secretion deficiencies and systemic insulin resistance (IR) in adipose tissue, skeletal muscle, and the liver. Although the mechanism of T2DM is not yet fully known, inflammation and insulin resistance play a central role in the pathogenesis of T2DM. G protein-coupled receptors (GPCRs) are involved in endocrine and metabolic processes as well as many other physiological processes. GPR50 (G protein-coupled receptor 50) is an orphan GPCR that shares the highest sequence homology with melatonin receptors. The aim of this study was to investigate the effect of GPR50 on inflammation and insulin resistance in 3T3-L1 preadipocytes. GPR50 expression was observed to be significantly increased in the adipose tissue of obese T2DM mice, while GPR50 deficiency increased inflammation in 3T3-L1 cells and induced the phosphorylation of AKT and insulin receptor substrate (IRS) 1. Furthermore, GPR50 knockout in the 3T3-L1 cell line suppressed PPAR-γ expression. These data suggest that GPR50 can attenuate inflammatory levels and regulate insulin signaling in adipocytes. Furthermore, the effects are mediated through the regulation of the IRS1/AKT signaling pathway and PPAR-γ expression.
Collapse
Affiliation(s)
- Zhenyu Yao
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Jun Meng
- Department of Pathogenic BiologyShenzhen Center for Disease Control and PreventionChina,Department of Microbiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Jing Long
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Long Li
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Weicong Qiu
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Cairong Li
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Pei‐Gen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| |
Collapse
|
105
|
Ye L, Jiang Y, Zhang M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine Growth Factor Rev 2022; 68:81-92. [PMID: 36376165 DOI: 10.1016/j.cytogfr.2022.11.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
Metabolites of glycolytic metabolism have been identified as signaling molecules and regulators of gene expression, in addition to their basic function as major energy and biosynthetic source. Immune cells reprogram metabolic pathways to cater to energy and biosynthesis demands upon activation. Most lymphocytes, including inflammatory M1 macrophages, mainly shift from oxidative phosphorylation to glycolysis, whereas regulatory T cells and M2 macrophages preferentially use the tricarboxylic acid (TCA) cycle and have reduced glycolysis. Recent studies have revealed the "non-metabolic" signaling functions of intermediates of the mitochondrial pathway and glycolysis. The roles of citrate, succinate and itaconate in immune response, including post-translational modifications of proteins and macrophages activation, have been highlighted. As an end product of glycolysis, lactate has received considerable interest from researchers. In this review, we specifically focused on studies exploring the integration of lactate into immune cell biology and associated pathologies. Lactate can act as a double-edged sword. On one hand, activated immune cells prefer to use lactate to support their function. On the other hand, accumulated lactate in the tissue microenvironment acts as a signaling molecule that restricts immune cell function. Recently, a novel epigenetic change mediated by histone lysine lactylation has been proposed. The burgeoning researches support the idea that histone lactylation participates in diverse cellular events. This review describes glycolytic metabolism, including the immunoregulation of metabolites of the TCA cycle and lactate. These latest findings strengthen our understanding on tumor and chronic inflammatory diseases and offer potential therapeutic options.
Collapse
Affiliation(s)
- Lei Ye
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China
| | - Yi Jiang
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China
| | - Mingming Zhang
- Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Shanghai 200001, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
106
|
Nunns GR, Vigneshwar N, Kelher MR, Stettler GR, Gera L, Reisz JA, D’Alessandro A, Ryon J, Hansen KC, Burke T, Gamboni F, Moore EE, Peltz ED, Cohen MJ, Jones KL, Sauaia A, Liang X, Banerjee A, Ghasabyan A, Chandler JG, Rodawig S, Jones C, Eitel A, Hom P, Silliman CC. Succinate Activation of SUCNR1 Predisposes Severely Injured Patients to Neutrophil-mediated ARDS. Ann Surg 2022; 276:e944-e954. [PMID: 33214479 PMCID: PMC8128932 DOI: 10.1097/sla.0000000000004644] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Identify the metabolites that are increased in the plasma of severely injured patients that developed ARDS versus severely injured patients that did not, and assay if these increased metabolites prime pulmonary sequestration of neutrophils (PMNs) and induce pulmonary sequestration in an animal model of ARDS. We hypothesize that metabolic derangement due to advanced shock in critically injured patients leads to the PMNs, which serves as the first event in the ARDS. Summary of Background Data: Intracellular metabolites accumulate in the plasma of severely injured patients. METHODS Untargeted metabolomics profiling of 67 critically injured patients was completed to establish a metabolic signature associated with ARDS development. Metabolites that significantly increased were assayed for PMN priming activity in vitro. The metabolites that primed PMNs were tested in a 2-event animal model of ARDS to identify a molecular link between circulating metabolites and clinical risk for ARDS. RESULTS After controlling for confounders, 4 metabolites significantly increased: creatine, dehydroascorbate, fumarate, and succinate in trauma patients who developed ARDS ( P < 0.05). Succinate alone primed the PMN oxidase in vitro at physiologically relevant levels. Intravenous succinate-induced PMN sequestration in the lung, a first event, and followed by intravenous lipopolysaccharide, a second event, resulted in ARDS in vivo requiring PMNs. SUCNR1 inhibition abrogated PMN priming, PMN sequestration, and ARDS. Conclusion: Significant increases in plasma succinate post-injury may serve as the first event in ARDS. Targeted inhibition of the SUCNR1 may decrease ARDS development from other disease states to prevent ARDS globally.
Collapse
Affiliation(s)
- Geoffrey R Nunns
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Navin Vigneshwar
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Marguerite R Kelher
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| | - Gregory R Stettler
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Lajos Gera
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Julie A. Reisz
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Angelo D’Alessandro
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Joshua Ryon
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Kirk C Hansen
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Timothy Burke
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| | - Fabia Gamboni
- Biochemistry and Molecular Genetics, School of Medicine University of Colorado, Aurora, CO
| | - Ernest E. Moore
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Department of Surgery, Denver Health Medical Center, Denver, CO
| | - Erik D Peltz
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Mitchell J Cohen
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Department of Surgery, Denver Health Medical Center, Denver, CO
| | | | - Angela Sauaia
- Department of Surgery, Denver Health Medical Center, Denver, CO
- School of Public Health, University of Colorado, Aurora, CO
| | - Xiayuan Liang
- Pathology, School of Medicine, University of Colorado, Aurora, CO
| | - Anirban Banerjee
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Arsen Ghasabyan
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - James G Chandler
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Sophia Rodawig
- Vitalant Research Institute, Vitalant Denver, Denver, CO
- College of Arts and Letters, University of Notre Dame, Notre Dame, IL
| | - Carter Jones
- Vitalant Research Institute, Vitalant Denver, Denver, CO
- College of Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Andrew Eitel
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Patrick Hom
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
| | - Christopher C Silliman
- Department of Surgery, School of Medicine University of Colorado, Aurora, CO
- Pediatrics, School of Medicine University of Colorado, CO
- Vitalant Research Institute, Vitalant Denver, Denver, CO
| |
Collapse
|
107
|
Jiang L, Shang M, Yu S, Liu Y, Zhang H, Zhou Y, Wang M, Wang T, Li H, Liu Z, Zhang X. A high-fiber diet synergizes with Prevotella copri and exacerbates rheumatoid arthritis. Cell Mol Immunol 2022; 19:1414-1424. [PMID: 36323929 PMCID: PMC9709035 DOI: 10.1038/s41423-022-00934-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Both preclinical and established rheumatoid arthritis (RA) patients display alterations in the gut microbiome. Prevotella spp. are preferentially enriched in a subset of RA patients. Here, we isolated a Prevotella strain, P. copri RA, from the feces of RA patients and showed that colonization of P. copri RA exacerbated arthritis in a collagen-induced arthritis (CIA) model. With the presence of P. copri RA colonization, a high-fiber diet exacerbated arthritis via microbial alterations and intestinal inflammation. Colonization of P. copri together with a high-fiber diet enabled the digestion of complex fiber, which led to the overproduction of organic acids, including fumarate, succinate and short-chain fatty acids. Succinate promoted proinflammatory responses in macrophages, and supplementation with succinate exacerbated arthritis in the CIA model. Our findings highlight the importance of dysbiosis when evaluating the effects of dietary interventions on RA pathogenesis and provide new insight into dietary interventions or microbiome modifications to improve RA management.
Collapse
Affiliation(s)
- Lingjuan Jiang
- Department of Medical Research Center, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengmeng Shang
- Department of Medical Research Center, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Shengnan Yu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yangzhong Zhou
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tingting Wang
- Department of Medical Research Center, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hui Li
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhihua Liu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
108
|
Mikkelsen RB, Arora T, Trošt K, Dmytriyeva O, Jensen SK, Meijnikman AS, Olofsson LE, Lappa D, Aydin Ö, Nielsen J, Gerdes V, Moritz T, van de Laar A, de Brauw M, Nieuwdorp M, Hjorth SA, Schwartz TW, Bäckhed F. Type 2 diabetes is associated with increased circulating levels of 3-hydroxydecanoate activating GPR84 and neutrophil migration. iScience 2022; 25:105683. [PMID: 36561890 PMCID: PMC9763857 DOI: 10.1016/j.isci.2022.105683] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/10/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Obesity and diabetes are associated with inflammation and altered plasma levels of several metabolites, which may be involved in disease progression. Some metabolites can activate G protein-coupled receptors (GPCRs) expressed on immune cells where they can modulate metabolic inflammation. Here, we find that 3-hydroxydecanoate is enriched in the circulation of obese individuals with type 2 diabetes (T2D) compared with nondiabetic controls. Administration of 3-hydroxydecanoate to mice promotes immune cell recruitment to adipose tissue, which was associated with adipose inflammation and increased fasting insulin levels. Furthermore, we demonstrate that 3-hydroxydecanoate stimulates migration of primary human and mouse neutrophils, but not monocytes, through GPR84 and Gαi signaling in vitro. Our findings indicate that 3-hydroxydecanoate is a T2D-associated metabolite that increases inflammatory responses and may contribute to the chronic inflammation observed in diabetes.
Collapse
Affiliation(s)
- Randi Bonke Mikkelsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kajetan Trošt
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sune Kjærsgaard Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Abraham Stijn Meijnikman
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Louise Elisabeth Olofsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dimitra Lappa
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ömrüm Aydin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Maurits de Brauw
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Siv Annegrethe Hjorth
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thue Walter Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik Bäckhed
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark,Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden,Corresponding author
| |
Collapse
|
109
|
Kuo CC, Wu JY, Wu KK. Cancer-derived extracellular succinate: a driver of cancer metastasis. J Biomed Sci 2022; 29:93. [DOI: 10.1186/s12929-022-00878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractSuccinate is a tricarboxylic acid (TCA) cycle intermediate normally confined to the mitochondrial matrix. It is a substrate of succinate dehydrogenase (SDH). Mutation of SDH subunits (SDHD and SDHB) in hereditary tumors such as paraganglioma or reduction of SDHB expression in cancer results in matrix succinate accumulation which is transported to cytoplasma and secreted into the extracellular milieu. Excessive cytosolic succinate is known to stabilize hypoxia inducible factor-1α (HIF-1α) by inhibiting prolyl hydroxylase. Recent reports indicate that cancer-secreted succinate enhances cancer cell migration and promotes cancer metastasis by activating succinate receptor-1 (SUCNR-1)-mediated signaling and transcription pathways. Cancer-derived extracellular succinate enhances cancer cell and macrophage migration through SUCNR-1 → PI-3 K → HIF-1α pathway. Extracellular succinate induces tumor angiogenesis through SUCNR-1-mediated ERK1/2 and STAT3 activation resulting in upregulation of vascular endothelial growth factor (VEGF) expression. Succinate increases SUCNR-1 expression in cancer cells which is considered as a target for developing new anti-metastasis drugs. Furthermore, serum succinate which is elevated in cancer patients may be a theranostic biomarker for selecting patients for SUCNR-1 antagonist therapy.
Collapse
|
110
|
Fan TWM, Daneshmandi S, Cassel TA, Uddin MB, Sledziona J, Thompson PT, Lin P, Higashi RM, Lane AN. Polarization and β-Glucan Reprogram Immunomodulatory Metabolism in Human Macrophages and Ex Vivo in Human Lung Cancer Tissues. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1674-1690. [PMID: 36150727 PMCID: PMC9588758 DOI: 10.4049/jimmunol.2200178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Abstract
Immunomodulatory (IM) metabolic reprogramming in macrophages (Mϕs) is fundamental to immune function. However, limited information is available for human Mϕs, particularly in response plasticity, which is critical to understanding the variable efficacy of immunotherapies in cancer patients. We carried out an in-depth analysis by combining multiplex stable isotope-resolved metabolomics with reversed phase protein array to map the dynamic changes of the IM metabolic network and key protein regulators in four human donors' Mϕs in response to differential polarization and M1 repolarizer β-glucan (whole glucan particles [WGPs]). These responses were compared with those of WGP-treated ex vivo organotypic tissue cultures (OTCs) of human non-small cell lung cancer. We found consistently enhanced tryptophan catabolism with blocked NAD+ and UTP synthesis in M1-type Mϕs (M1-Mϕs), which was associated with immune activation evidenced by increased release of IL-1β/CXCL10/IFN-γ/TNF-α and reduced phagocytosis. In M2a-Mϕs, WGP treatment of M2a-Mϕs robustly increased glucose utilization via the glycolysis/oxidative branch of the pentose phosphate pathway while enhancing UDP-N-acetyl-glucosamine turnover and glutamine-fueled gluconeogenesis, which was accompanied by the release of proinflammatory IL-1β/TNF-α to above M1-Mϕ's levels, anti-inflammatory IL-10 to above M2a-Mϕ's levels, and attenuated phagocytosis. These IM metabolic responses could underlie the opposing effects of WGP, i.e., reverting M2- to M1-type immune functions but also boosting anti-inflammation. Variable reprogrammed Krebs cycle and glutamine-fueled synthesis of UTP in WGP-treated OTCs of human non-small cell lung cancer were observed, reflecting variable M1 repolarization of tumor-associated Mϕs. This was supported by correlation with IL-1β/TNF-α release and compromised tumor status, making patient-derived OTCs unique models for studying variable immunotherapeutic efficacy in cancer patients.
Collapse
Affiliation(s)
- Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY;
- Markey Cancer Center, University of Kentucky, Lexington, KY; and
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY
| | - Saeed Daneshmandi
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - Teresa A Cassel
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - Mohammad B Uddin
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - James Sledziona
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - Patrick T Thompson
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - Penghui Lin
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY
- Markey Cancer Center, University of Kentucky, Lexington, KY; and
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY;
- Markey Cancer Center, University of Kentucky, Lexington, KY; and
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY
| |
Collapse
|
111
|
Chen K, Wu T, Wang D, Li R, Shen X, Zhao T, Ozato K, Li R. Transcriptomics and quantitative proteomics reveal changes after second stimulation of bone marrow-derived macrophages from lupus-prone MRL/lpr mice. Front Immunol 2022; 13:1004232. [PMID: 36341359 PMCID: PMC9627492 DOI: 10.3389/fimmu.2022.1004232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immune memory can cause the occurrence and exacerbation of autoimmune diseases, and it is as well as being strongly associated with the pathogenesis of systemic lupus erythematosus (SLE), however, the specific mechanism remains to be further studied. We learned that IFN-γ stimulation generated innate immune memory in bone marrow-derived macrophages (BMDMs) and activated memory interferon-stimulated genes (ISGs). This research used IFN-γ and lipopolysaccharide (LPS) to treat BMDMs with lupus-prone MRL/lpr mice and showed that particular memory ISGs were substantially elevated in prestimulated macrophages. In order to identify the differentially expressed genes (DEGs), researchers turned to RNA-seq. GO and KEGG analysis showed that up-regulated DEGs were enriched in defense and innate immune responses, and were related to the expression of pattern recognition receptors (PRRs)-related pathways in macrophages. TMT-based proteome analysis revealed differentially expressed proteins (DEPs) up-regulated in BMDMs were abundant in metabolic pathways such as glucose metabolism. Our study found that after the secondary stimulation of MRL/lpr mice, the expression of PRRs in innate immune cells was changed, and IFN-related pathways were activated to release a large number of ISGs to promote the secondary response. At the same time, related metabolic modes such as glycolysis were enhanced, and epigenetic changes may occur. Therefore, SLE is brought on, maintained, and worsened by a variety of factors that work together to produce innate immune memory.
Collapse
Affiliation(s)
- Keyue Chen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tiyun Wu
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Danyan Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rong Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangfeng Shen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Keiko Ozato, ; Rongqun Li,
| | - Rongqun Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Keiko Ozato, ; Rongqun Li,
| |
Collapse
|
112
|
Hu H, Guo L, Overholser J, Wang X. Mitochondrial VDAC1: A Potential Therapeutic Target of Inflammation-Related Diseases and Clinical Opportunities. Cells 2022; 11:cells11193174. [PMID: 36231136 PMCID: PMC9562648 DOI: 10.3390/cells11193174] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022] Open
Abstract
The multifunctional protein, voltage-dependent anion channel 1 (VDAC1), is located on the mitochondrial outer membrane. It is a pivotal protein that maintains mitochondrial function to power cellular bioactivities via energy generation. VDAC1 is involved in regulating energy production, mitochondrial oxidase stress, Ca2+ transportation, substance metabolism, apoptosis, mitochondrial autophagy (mitophagy), and many other functions. VDAC1 malfunction is associated with mitochondrial disorders that affect inflammatory responses, resulting in an up-regulation of the body’s defensive response to stress stimulation. Overresponses to inflammation may cause chronic diseases. Mitochondrial DNA (mtDNA) acts as a danger signal that can further trigger native immune system activities after its secretion. VDAC1 mediates the release of mtDNA into the cytoplasm to enhance cytokine levels by activating immune responses. VDAC1 regulates mitochondrial Ca2+ transportation, lipid metabolism and mitophagy, which are involved in inflammation-related disease pathogenesis. Many scientists have suggested approaches to deal with inflammation overresponse issues via specific targeting therapies. Due to the broad functionality of VDAC1, it may become a useful target for therapy in inflammation-related diseases. The mechanisms of VDAC1 and its role in inflammation require further exploration. We comprehensively and systematically summarized the role of VDAC1 in the inflammatory response, and hope that our research will lead to novel therapeutic strategies that target VDAC1 in order to treat inflammation-related disorders.
Collapse
Affiliation(s)
- Hang Hu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Linlin Guo
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (L.G.); (X.W.)
| | - Jay Overholser
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Correspondence: (L.G.); (X.W.)
| |
Collapse
|
113
|
Huang LY, Ma JY, Song JX, Xu JJ, Hong R, Fan HD, Cai H, Wang W, Wang YL, Hu ZL, Shen JG, Qi SH. Ischemic accumulation of succinate induces Cdc42 succinylation and inhibits neural stem cell proliferation after cerebral ischemia/reperfusion. Neural Regen Res 2022; 18:1040-1045. [PMID: 36254990 PMCID: PMC9827777 DOI: 10.4103/1673-5374.355821] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemic accumulation of succinate causes cerebral damage by excess production of reactive oxygen species. However, it is unknown whether ischemic accumulation of succinate affects neural stem cell proliferation. In this study, we established a rat model of cerebral ischemia/reperfusion injury by occlusion of the middle cerebral artery. We found that succinate levels increased in serum and brain tissue (cortex and hippocampus) after ischemia/reperfusion injury. Oxygen-glucose deprivation and reoxygenation stimulated primary neural stem cells to produce abundant succinate. Succinate can be converted into diethyl succinate in cells. Exogenous diethyl succinate inhibited the proliferation of mouse-derived C17.2 neural stem cells and increased the infarct volume in the rat model of cerebral ischemia/reperfusion injury. Exogenous diethyl succinate also increased the succinylation of the Rho family GTPase Cdc42 but repressed Cdc42 GTPase activity in C17.2 cells. Increasing Cdc42 succinylation by knockdown of the desuccinylase Sirt5 also inhibited Cdc42 GTPase activity in C17.2 cells. Our findings suggest that ischemic accumulation of succinate decreases Cdc42 GTPase activity by induction of Cdc42 succinylation, which inhibits the proliferation of neural stem cells and aggravates cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Lin-Yan Huang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ju-Yun Ma
- College of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jin-Xiu Song
- College of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jing-Jing Xu
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Rui Hong
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hai-Di Fan
- College of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Heng Cai
- College of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wan Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yan-Ling Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zhao-Li Hu
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jian-Gang Shen
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Su-Hua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,College of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Su-Hua Qi, .
| |
Collapse
|
114
|
Casey AM, Murphy MP. Uncovering the source of mitochondrial superoxide in pro-inflammatory macrophages: Insights from immunometabolism. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166481. [PMID: 35792320 PMCID: PMC7614207 DOI: 10.1016/j.bbadis.2022.166481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 11/15/2022]
Abstract
Mitochondrial-derived reactive oxygen species are important as antimicrobial agents and redox signals in pro-inflammatory macrophages. Macrophages produce superoxide in response to the TLR4 ligand LPS. However, the mechanism of LPS-induced superoxide generation is not fully understood. Superoxide is produced at complex I and complex III of the electron transport chain. Production of superoxide at either of these sites is highly dependent on the metabolic state of the cell which is dramatically altered by TLR4-induced metabolic reprogramming. This review will outline how metabolism impacts superoxide production in LPS-activated macrophages downstream of TLR4 signalling and address outstanding questions in this field.
Collapse
|
115
|
Macrophage immunometabolism in inflammatory bowel diseases: From pathogenesis to therapy. Pharmacol Ther 2022; 238:108176. [DOI: 10.1016/j.pharmthera.2022.108176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/11/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022]
|
116
|
Pulmonary neuroendocrine cells sense succinate to stimulate myoepithelial cell contraction. Dev Cell 2022; 57:2221-2236.e5. [PMID: 36108628 DOI: 10.1016/j.devcel.2022.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/19/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022]
Abstract
Pulmonary neuroendocrine cells (PNECs) are rare airway cells with potential sensory capacity linked to vagal neurons and immune cells. How PNECs sense and respond to external stimuli remains poorly understood. We discovered PNECs located within pig and human submucosal glands, a tissue that produces much of the mucus that defends the lung. These PNECs sense succinate, an inflammatory molecule in liquid lining the airway surface. The results indicate that succinate migrates down the submucosal gland duct to the acinus, where it triggers apical succinate receptors, causing PNECs to release ATP. The short-range ATP signal stimulates the contraction of myoepithelial cells wrapped tightly around the submucosal glands. Succinate-triggered gland contraction may complement the action of neurotransmitters that induce mucus release but not gland contraction to promote mucus ejection onto the airway surface. These findings identify a local circuit in which rare PNECs within submucosal glands sense an environmental cue to orchestrate the function of airway glands.
Collapse
|
117
|
Guo Y, Xu F, Thomas SC, Zhang Y, Paul B, Sakilam S, Chae S, Li P, Almeter C, Kamer AR, Arora P, Graves DT, Saxena D, Li X. Targeting the succinate receptor effectively inhibits periodontitis. Cell Rep 2022; 40:111389. [PMID: 36130514 PMCID: PMC9533417 DOI: 10.1016/j.celrep.2022.111389] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/06/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Periodontal disease (PD) is one of the most common inflammatory diseases in humans and is initiated by an oral microbial dysbiosis that stimulates inflammation and bone loss. Here, we report an abnormal elevation of succinate in the subgingival plaque of subjects with severe PD. Succinate activates succinate receptor-1 (SUCNR1) and stimulates inflammation. We detected SUCNR1 expression in the human and mouse periodontium and hypothesize that succinate activates SUCNR1 to accelerate periodontitis through the inflammatory response. Administration of exogenous succinate enhanced periodontal disease, whereas SUCNR1 knockout mice were protected from inflammation, oral dysbiosis, and subsequent periodontal bone loss in two different models of periodontitis. Therapeutic studies demonstrated that a SUCNR1 antagonist inhibited inflammatory events and osteoclastogenesis in vitro and reduced periodontal bone loss in vivo. Our study reveals succinate’s effect on periodontitis pathogenesis and provides a topical treatment for this disease. Periodontitis is the most prevalent adult oral disease. Guo et al. show elevation of succinate in periodontitis, which aggravates the disease through the succinate receptor (SUCNR1). They developed a gel formulation of a small compound specifically blocking SUCNR1 to prevent and treat periodontitis by inhibiting dysbiosis, inflammation, and bone loss.
Collapse
Affiliation(s)
- Yuqi Guo
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Yanli Zhang
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Bidisha Paul
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Satish Sakilam
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Sungpil Chae
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Patty Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Caleb Almeter
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Angela R Kamer
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York, NY 10010, USA
| | - Paramjit Arora
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Dana T Graves
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; Department of Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA.
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; Department of Urology, New York University Grossman School of Medicine, New York, NY 10016, USA; Perlmutter Cancer Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
118
|
Yeshi K, Ruscher R, Loukas A, Wangchuk P. Immunomodulatory and biological properties of helminth-derived small molecules: Potential applications in diagnostics and therapeutics. FRONTIERS IN PARASITOLOGY 2022; 1:984152. [PMID: 39816468 PMCID: PMC11731824 DOI: 10.3389/fpara.2022.984152] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2025]
Abstract
Parasitic helminths secrete and excrete a vast array of molecules known to help skew or suppress the host's immune response, thereby establishing a niche for sustained parasite maintenance. Indeed, the immunomodulatory potency of helminths is attributed mainly to excretory/secretory products (ESPs). The ESPs of helminths and the identified small molecules (SM) are reported to have diverse biological and pharmacological properties. The available literature reports only limited metabolites, and the identity of many metabolites remains unknown due to limitations in the identification protocols and helminth-specific compound libraries. Many metabolites are known to be involved in host-parasite interactions and pathogenicity. For example, fatty acids (e.g., stearic acid) detected in the infective stages of helminths are known to have a role in host interaction through facilitating successful penetration and migration inside the host. Moreover, excreted/secreted SM detected in helminth species are found to possess various biological properties, including anti-inflammatory activities, suggesting their potential in developing immunomodulatory drugs. For example, helminths-derived somatic tissue extracts and whole crude ESPs showed anti-inflammatory properties by inhibiting the secretion of proinflammatory cytokines from human peripheral blood mononuclear cells and suppressing the pathology in chemically-induced experimental mice model of colitis. Unlike bigger molecules like proteins, SM are ideal candidates for drug development since they are small structures, malleable, and lack immunogenicity. Future studies should strive toward identifying unknown SM and isolating the under-explored niche of helminth metabolites using the latest metabolomics technologies and associated software, which hold potential keys for finding new diagnostics and novel therapeutics.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia
| | | | | | | |
Collapse
|
119
|
Li S, Lin R, Chen J, Hussain R, Zhang S, Su Y, Chan Y, Ghaffar A, Shi D. Integrated gut microbiota and metabolomic analysis reveals immunomodulatory effects of Echinacea extract and Astragalus polysaccharides. Front Vet Sci 2022; 9:971058. [PMID: 36118329 PMCID: PMC9478787 DOI: 10.3389/fvets.2022.971058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Immunosuppression in different animals increases the susceptibility of various infections caused by pathogenic microorganisms leading to increase risks posed by antibiotics in different animal farming sectors. Therefore, investigation of the interactions between natural medicines and the intestinal environmental ecosystem is of vital importance and crucial. This study for the first time investigated the effects of Echinacea Extract (EE) and Astragalus polysaccharide (APS) on the gut using 16S rRNA and metabolomic analysis approaches in immunosuppressed broiler chickens. There were four groups divided into control (C), immunosuppression (IS), EE, and APS groups. Sequencing of gut microbes showed that immunosuppression decreased the relative abundance of Anaerofustis, Anaeroplasma, Anaerotroncus, and Lachnospira in the gut while increasing that of c_115 and Holdemania. However, EE and APS diminished the effects on the immunosuppression on the microbiota. The results revealed up-regulation of the relative abundance of Enterococcus in broiler chickens. In addition, EE reduced the relative abundance of Ruminococcus and Blautia. The results on metabolomic analysis revealed that immunosuppression mainly affects cyanuric acid metabolism, starch and sucrose metabolism while interconversion of pentose and glucuronide. EE and APS, on the other hand mainly impact butyrate metabolism, alanine, aspartate and glutamate metabolism while the interconversion of pentose and glucuronide, and D-glutamine and D-glutamate metabolism. Results regarding correlation analysis revealed significantly metabolic pathways including TCA cycle, butyrate metabolism, glyoxylate and dicarboxylate metabolism, propionate metabolism, alanine, aspartate and glutamate metabolism associated with Ruminococcus and Blautia. Both EE and APS can antagonize the effects of immunosuppression by modulating the disrupted gut microbiota. Nevertheless, EE might have a bidirectional regulatory functions on the intestinal health and further studies are needed to know the exact and relevant mechanisms of action regarding the effects of EE and APS.
Collapse
Affiliation(s)
- Shaochuan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Renzhao Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiaxin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Riaz Hussain
- The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Shiwei Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yalin Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yanzi Chan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Abdul Ghaffar
- The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- *Correspondence: Dayou Shi
| |
Collapse
|
120
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
121
|
Tang X, Rönnberg E, Säfholm J, Thulasingam M, Trauelsen M, Schwartz TW, Wheelock CE, Dahlén S, Nilsson G, Haeggström JZ. Activation of succinate receptor 1 boosts human mast cell reactivity and allergic bronchoconstriction. Allergy 2022; 77:2677-2687. [PMID: 35122266 PMCID: PMC9545225 DOI: 10.1111/all.15245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/31/2021] [Accepted: 01/23/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND SUCNR1 is a sensor of extracellular succinate, a Krebs cycle intermediate generated in excess during oxidative stress and has been linked to metabolic regulation and inflammation. While mast cells express SUCNR1, its role in mast cell reactivity and allergic conditions such as asthma remains to be elucidated. METHODS Cord blood-derived mast cells and human mast cell line LAD-2 challenged by SUCNR1 ligands were analyzed for the activation and mediator release. Effects on mast cell-dependent bronchoconstriction were assessed in guinea pig trachea and isolated human small bronchi challenged with antigen and anti-IgE, respectively. RESULTS SUCNR1 is abundantly expressed on human mast cells. Challenge with succinate, or the synthetic non-metabolite agonist cis-epoxysuccinate, renders mast cells hypersensitive to IgE-dependent activation, resulting in augmented degranulation and histamine release, de novo biosynthesis of eicosanoids and cytokine secretion. The succinate-potentiated mast cell reactivity was attenuated by SUCNR1 knockdown and selective SUCNR1 antagonists and could be tuned by pharmacologically targeting protein kinase C and extracellular signal-regulated kinase. Both succinate and cis-epoxysuccinate dose-dependently potentiated antigen-induced contraction in a mast cell-dependent guinea pig airway model, associated with increased generation of cysteinyl-leukotrienes and histamine in trachea. Similarly, cis-epoxysuccinate aggravated IgE-receptor-induced contraction of human bronchi, which was blocked by SUCNR1 antagonism. CONCLUSION SUCNR1 amplifies IgE-receptor-induced mast cell activation and allergic bronchoconstriction, suggesting a role for this pathway in aggravation of allergic asthma, thus linking metabolic perturbations to mast cell-dependent inflammation.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Elin Rönnberg
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet, and Karolinska University HospitalSolnaSweden
| | - Jesper Säfholm
- Unit of Experimental Asthma and Allergy ResearchInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Madhuranayaki Thulasingam
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Mette Trauelsen
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Thue W. Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Craig E. Wheelock
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Sven‐Erik Dahlén
- Unit of Experimental Asthma and Allergy ResearchInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden,Department of Respiratory MedicineKarolinska University Hospital HuddingeStockholmSweden
| | - Gunnar Nilsson
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet, and Karolinska University HospitalSolnaSweden,Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Jesper Z. Haeggström
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| |
Collapse
|
122
|
Srimadh Bhagavatham SK, Pulukool SK, Pradhan SS, R S, Ashok Naik A, V M DD, Sivaramakrishnan V. Systems biology approach delineates critical pathways associated with disease progression in rheumatoid arthritis. J Biomol Struct Dyn 2022:1-22. [PMID: 36047508 DOI: 10.1080/07391102.2022.2115555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Rheumatoid Arthritis (RA) is a chronic systemic autoimmune disease leading to inflammation, cartilage cell death, synoviocyte proliferation, and increased and impaired differentiation of osteoclasts and osteoblasts leading to joint erosions and deformities. Transcriptomics, proteomics, and metabolomics datasets were analyzed to identify the critical pathways that drive the RA pathophysiology. Single nucleotide polymorphisms (SNPs) associated with RA were analyzed for the functional implications, clinical outcomes, and blood parameters later validated by literature. SNPs associated with RA were grouped into pathways that drive the immune response and cytokine production. Further gene set enrichment analysis (GSEA) was performed on gene expression omnibus (GEO) data sets of peripheral blood mononuclear cells (PBMCs), synovial macrophages, and synovial biopsies from RA patients showed enrichment of Th1, Th2, Th17 differentiation, viral and bacterial infections, metabolic signalling and immunological pathways with potential implications for RA. The proteomics data analysis presented pathways with genes involved in immunological signaling and metabolic pathways, including vitamin B12 and folate metabolism. Metabolomics datasets analysis showed significant pathways like amino-acyl tRNA biosynthesis, metabolism of amino acids (arginine, alanine aspartate, glutamate, glutamine, phenylalanine, and tryptophan), and nucleotide metabolism. Furthermore, our commonality analysis of multi-omics datasets identified common pathways with potential implications for joint remodeling in RA. Disease-modifying anti-rheumatic drugs (DMARDs) and biologics treatments were found to modulate many of the pathways that were deregulated in RA. Overall, our analysis identified molecular signatures associated with the observed symptoms, joint erosions, potential biomarkers, and therapeutic targets in RA. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Sujith Kumar Pulukool
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Saiswaroop R
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Ashwin Ashok Naik
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Datta Darshan V M
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| |
Collapse
|
123
|
Murillo-Saich JD, Coras R, Meyer R, Llorente C, Lane NE, Guma M. Synovial tissue metabolomic profiling reveal biomarkers of synovial inflammation in patients with osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100295. [PMID: 36474936 PMCID: PMC9718344 DOI: 10.1016/j.ocarto.2022.100295] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 01/30/2023] Open
Abstract
Objective Inflammatory responses are associated with changes in tissue metabolism. Prior studies find altered metabolomic profiles in both the synovial fluid (SF) and serum of osteoarthritis subjects. Our study determined the metabolomic profile of synovial tissue (ST) and SF of individuals with osteoarthritis (OA) and its association with synovial inflammation. Design 37 OA ST samples were collected during joint replacement, 21 also had SF. ST samples were fixed in formalin for histological analysis, cultured (explants) for cytokine analysis by enzyme-linked immunosorbent assay, or snap-frozen for metabolomic analysis. ST samples were categorized by Krenn synovitis score and picrosirius red. CD68 and vimentin expression was assessed by immunohistochemistry and semi-quantified using Image J. Proton-nuclear magnetic resonance (1H NMR) was used to acquire a spectrum from ST and SF samples. Chenomx NMR suite 8.5 was used for metabolite identification and quantification. Metaboanalyst 5.0, SPSS v26, and R (v4.1.2) were used for statistical analysis. Results 42 and 29 metabolites were detected in the ST and SF respectively by 1H NMR. Only 3 metabolites, lactate, dimethylamine, and creatine positively correlated between SF and ST. ST concentrations of several metabolites (lactate, alanine, fumarate, glutamine, glycine, leucine, lysine, methionine, trimethylamine N-oxide, tryptophan and valine) were associated with synovitis score, mostly to the lining score. IL-6, acetoacetate, and tyrosine in SF predicted high Krenn synovitis scores in ST. Conclusion Metabolomic profiling of ST identified metabolic changes associated with inflammation. Further studies are needed to determine whether metabolomic profiling of synovial tissue can identify new therapeutic targets in osteoarthritis.
Collapse
Affiliation(s)
- Jessica D. Murillo-Saich
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA, 92093, USA
| | - Roxana Coras
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA, 92093, USA
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
| | - Robert Meyer
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA, 92093, USA
- San Diego VA Healthcare Service, San Diego, CA, 92161, USA
| | - Cristina Llorente
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA, 92093, USA
| | - Nancy E. Lane
- Department of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Monica Guma
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA, 92093, USA
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
- San Diego VA Healthcare Service, San Diego, CA, 92161, USA
| |
Collapse
|
124
|
Pinget GV, Tan JK, Ni D, Taitz J, Daien CI, Mielle J, Moore RJ, Stanley D, Simpson S, King NJC, Macia L. Dysbiosis in imiquimod-induced psoriasis alters gut immunity and exacerbates colitis development. Cell Rep 2022; 40:111191. [PMID: 35977500 DOI: 10.1016/j.celrep.2022.111191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 05/30/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Psoriasis has long been associated with inflammatory bowel disease (IBD); however, a causal link is yet to be established. Here, we demonstrate that imiquimod-induced psoriasis (IMQ-pso) in mice disrupts gut homeostasis, characterized by increased proportions of colonic CX3CR1hi macrophages, altered cytokine production, and bacterial dysbiosis. Gut microbiota from these mice produce higher levels of succinate, which induce de novo proliferation of CX3CR1hi macrophages ex vivo, while disrupted gut homeostasis primes IMQ-pso mice for more severe colitis with dextran sulfate sodium (DSS) challenge. These results demonstrate that changes in the gut environment in psoriasis lead to greater susceptibility to IBD in mice, suggesting a two-hit requirement, that is, psoriasis-induced altered gut homeostasis and a secondary environmental challenge. This may explain the increased prevalence of IBD in patients with psoriasis.
Collapse
Affiliation(s)
- Gabriela Veronica Pinget
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jian Kai Tan
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Duan Ni
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jemma Taitz
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Claire Immediato Daien
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; CHRU Montpellier, University of Montpellier & INSERM U1046, CNRS UMR, PhyMedExp, 9214 Montpellier, France
| | - Julie Mielle
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; CHRU Montpellier, University of Montpellier & INSERM U1046, CNRS UMR, PhyMedExp, 9214 Montpellier, France
| | | | - Dragana Stanley
- School of Health, Medical and Applied Sciences, Central Queensland University, Kawana, QLD 4701, Australia
| | - Stephen Simpson
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Nicholas Jonathan Cole King
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Laurence Macia
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Cytometry, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
125
|
Tang X, Hou Y, Schwartz TW, Haeggström JZ. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids. Biochem Pharmacol 2022; 204:115208. [PMID: 35963340 DOI: 10.1016/j.bcp.2022.115208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Eicosanoids are a family of bioactive compounds derived from arachidonic acid (AA) that play pivotal roles in physiology and disease, including inflammatory conditions of multiple organ systems. The biosynthesis of eicosanoids requires a series of catalytic steps that are controlled by designated enzymes, which can be regulated by inflammatory and stress signals via transcriptional and translational mechanisms. In the past decades, evidence have emerged indicating that G-protein coupled receptors (GPCRs) can sense extracellular metabolites, and regulate inflammatory responses including eicosanoid production. This review focuses on the recent advances of metabolite GPCRs research, their role in regulation of eicosanoid biosynthesis, and the link to pathophysiological conditions.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| | - Yaolin Hou
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Thue W Schwartz
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department for Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| |
Collapse
|
126
|
Hughey CC, Puchalska P, Crawford PA. Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159209. [DOI: 10.1016/j.bbalip.2022.159209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/25/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
|
127
|
Pérez-Hernández CA, Moreno-Altamirano MMB, López-Villegas EO, Butkeviciute E, Ali M, Kronsteiner B, Dunachie SJ, Dockrell HM, Smith SG, Sánchez-García FJ. Mitochondrial Ultrastructure and Activity Are Differentially Regulated by Glycolysis-, Krebs Cycle-, and Microbiota-Derived Metabolites in Monocytes. BIOLOGY 2022; 11:biology11081132. [PMID: 36009759 PMCID: PMC9404980 DOI: 10.3390/biology11081132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022]
Abstract
Several intermediate metabolites harbour cell-signalling properties, thus, it is likely that specific metabolites enable the communication between neighbouring cells, as well as between host cells with the microbiota, pathogens, and tumour cells. Mitochondria, a source of intermediate metabolites, participate in a wide array of biological processes beyond that of ATP production, such as intracellular calcium homeostasis, cell signalling, apoptosis, regulation of immune responses, and host cell-microbiota crosstalk. In this regard, mitochondria's plasticity allows them to adapt their bioenergetics status to intra- and extra-cellular cues, and the mechanisms driving such plasticity are currently a matter of intensive research. Here, we addressed whether mitochondrial ultrastructure and activity are differentially shaped when human monocytes are exposed to an exogenous source of lactate (derived from glycolysis), succinate, and fumarate (Krebs cycle metabolic intermediates), or butyrate and acetate (short-chain fatty acids produced by intestinal microbiota). It has previously been shown that fumarate induces mitochondrial fusion, increases the mitochondrial membrane potential (Δψm), and reshapes the mitochondrial cristae ultrastructure. Here, we provide evidence that, in contrast to fumarate, lactate, succinate, and butyrate induce mitochondrial fission, while acetate induces mitochondrial swelling. These traits, along with mitochondrial calcium influx kinetics and glycolytic vs. mitochondrial ATP-production rates, suggest that these metabolites differentially shape mitochondrial function, paving the way for the understanding of metabolite-induced metabolic reprogramming of monocytes and its possible use for immune-response intervention.
Collapse
Affiliation(s)
- C. Angélica Pérez-Hernández
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
| | - M. Maximina Bertha Moreno-Altamirano
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
| | - Edgar O. López-Villegas
- Unidad de Microscopía, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Egle Butkeviciute
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.B.); (H.M.D.)
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Susanna J. Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Hazel M. Dockrell
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.B.); (H.M.D.)
| | - Steven G. Smith
- Division of Biosciences, Brunel University London, London UB8 3PH, UK;
| | - F. Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
- Correspondence:
| |
Collapse
|
128
|
Simulated hypoxia modulates P2X7 receptor function in mice peritoneal macrophages. Int Immunopharmacol 2022; 110:109062. [PMID: 35863257 DOI: 10.1016/j.intimp.2022.109062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
The inflammatory focus is similar to the tumor microenvironment, which contains a complex milieu with immune cells and macrophages. The accumulation of cells promotes local pH and O2 tension decline (hypoxia). Local O2 tension decline activates hypoxia-inducible factor α and β (HIF-1α and HIF-1β adenosine triphosphate (ATP) release. ATP activates the P2X7 receptor and modulates ischemic/hypoxic conditions. Similarly, α1α may regulate P2X7 receptor expression in the hypoxic microenvironment. Therefore, we investigated P2X7 receptor function under simulated hypoxic conditions by pretreating peritoneal macrophages with mitochondrial electron transport chain complex inhibitors (simulated hypoxia). Treatment with mitochondrial electron transport chain complex inhibitors until three hours of exposure did not cause LDH release. Additionally, mitochondrial electron transport chain complex inhibitors increased ATP-induced P2X7 receptor function without being able to directly activate this receptor. Other P2 receptor subtypes do not appear to participate in this mechanism. Simulated hypoxia augmented HIF-1α levels and suppressed HIF-1α and P2X7 receptor antagonists. Similarly, simulated hypoxia increased ATP-induced dye uptake and inhibited HIF-1α antagonists. Another factor activated in simulated hypoxic conditions was the intracellular P2X7 receptor regulator PIP2. Treatment with HIF-1α agonists increased PIP2 levels and reversed the effects of HIF-1α and P2X7 receptor antagonists. Additionally, the improved ATP-induced dye uptake caused by the simulated hypoxia stimulus was inhibited by P2X7 receptor and PIP2 antagonists. Therefore, simulated hypoxia may augment P2X7 receptor activity for a pathway dependent on HIF-1α and PIP2 activation.
Collapse
|
129
|
Sanchez M, Hamel D, Bajon E, Duhamel F, Bhosle VK, Zhu T, Rivera JC, Dabouz R, Nadeau-Vallée M, Sitaras N, Tremblay DÉ, Omri S, Habelrih T, Rouget R, Hou X, Gobeil F, Joyal JS, Sapieha P, Mitchell G, Ribeiro-Da-Silva A, Mohammad Nezhady MA, Chemtob S. The Succinate Receptor SUCNR1 Resides at the Endoplasmic Reticulum and Relocates to the Plasma Membrane in Hypoxic Conditions. Cells 2022; 11:2185. [PMID: 35883628 PMCID: PMC9321536 DOI: 10.3390/cells11142185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022] Open
Abstract
The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.
Collapse
Affiliation(s)
- Melanie Sanchez
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - David Hamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Emmanuel Bajon
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - François Duhamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Vikrant K. Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Cell Biology Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Tang Zhu
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Jose Carlos Rivera
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Rabah Dabouz
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mathieu Nadeau-Vallée
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Nicholas Sitaras
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - David-Étienne Tremblay
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Samy Omri
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Tiffany Habelrih
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Raphael Rouget
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Xin Hou
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Fernand Gobeil
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Grant Mitchell
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Alfredo Ribeiro-Da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Program of Molecular Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
130
|
Zhao J, Wei K, Jiang P, Chang C, Xu L, Xu L, Shi Y, Guo S, He D. G-Protein-Coupled Receptors in Rheumatoid Arthritis: Recent Insights into Mechanisms and Functional Roles. Front Immunol 2022; 13:907733. [PMID: 35874704 PMCID: PMC9304905 DOI: 10.3389/fimmu.2022.907733] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint damage and even disability. Although there are various clinical therapies for RA, some patients still have poor or no response. Thus, the development of new drug targets remains a high priority. In this review, we discuss the role of G-protein-coupled receptors (GPCRs), including chemokine receptors, melanocortin receptors, lipid metabolism-related receptors, adenosine receptors, and other inflammation-related receptors, on mechanisms of RA, such as inflammation, lipid metabolism, angiogenesis, and bone destruction. Additionally, we summarize the latest clinical trials on GPCR targeting to provide a theoretical basis and guidance for the development of innovative GPCR-based clinical drugs for RA.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
131
|
Pardella E, Ippolito L, Giannoni E, Chiarugi P. Nutritional and metabolic signalling through GPCRs. FEBS Lett 2022; 596:2364-2381. [PMID: 35776088 DOI: 10.1002/1873-3468.14441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022]
Abstract
Deregulated metabolism is a well-known feature of several challenging diseases, including diabetes, obesity and cancer. Besides their important role as intracellular bioenergetic molecules, dietary nutrients and metabolic intermediates are released in the extracellular environment. As such, they may achieve unconventional roles as hormone-like molecules by activating cell-surface G-protein-coupled receptors (GPCRs) that regulate several pathophysiological processes. In this review, we provide an insight into the role of lactate, succinate, fatty acids, amino acids, ketogenesis-derived and β-oxidation-derived intermediates as extracellular signalling molecules. Moreover, the mechanisms by which their cognate metabolite-sensing GPCRs integrate nutritional and metabolic signals with specific intracellular pathways will be described. A better comprehension of these aspects is of fundamental importance to identify GPCRs as novel druggable targets.
Collapse
Affiliation(s)
- Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| |
Collapse
|
132
|
Zhang M, Liu J, Li C, Gao J, Xu C, Wu X, Xu T, Cui C, Wei H, Peng J, Zheng R. Functional Fiber Reduces Mice Obesity by Regulating Intestinal Microbiota. Nutrients 2022; 14:nu14132676. [PMID: 35807856 PMCID: PMC9268532 DOI: 10.3390/nu14132676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 12/10/2022] Open
Abstract
Obesity may cause metabolic syndrome and has become a global public health problem, and dietary fibers (DF) could alleviate obesity and metabolic syndrome by regulating intestinal microbiota. We developed a functional fiber (FF) with a synthetic mixture of polysaccharides, high viscosity, water-binding capacity, swelling capacity, and fermentability. This study aimed to investigate the effect of FF on obesity and to determine its prevention of obesity by modulating the gut microbiota. Physiological, histological, and biochemical parameters, and gut microbiota composition were investigated in the following six groups: control group (Con), high-fat diet group (HFD), low-fat diet group (LFD, conversion of HFD to LFD), high-fat +8% FF group (8% FF), high-fat +12% FF group (12% FF), and high-fat +12% FF + antibiotic group (12% FF + AB). The results demonstrated that 12% FF could promote a reduction in body weight and epididymal adipocyte area, augment insulin sensitivity, and stimulate heat production from brown adipose tissue (BAT) (p < 0.05). Compared with the HFD, 12% FF could also significantly improve the intestinal morphological integrity, attenuate systemic inflammation, promote intestinal microbiota homeostasis, and stabilize the production of short-chain fatty acids (SCFAs) (p < 0.05). Consistent with the results of 12% FF, the LFD could significantly reduce the body weight and epididymal adipocyte area relative to the HFD (p < 0.05), but the LFD and HFD showed no significant difference (p > 0.05) in the level of inflammation and SCFAs. Meanwhile, 12% FF supplementation showed an increase (p < 0.05) in the abundance of the Bifidobacterium, Lactococcus, and Coprococcus genus in the intestine, which had a negative correlation with obesity and insulin resistance. Additionally, the treatment with antibiotics (12% FF + AB) could inhibit the effect of FF in the HFD. The Kyoto Encyclopedia of Genes and Genomes (KEGG) function prediction revealed that 12% FF could significantly inhibit the cyanogenic amino acid metabolic pathway and decrease the serum succinate concentration relative to the HFD group. The overall results indicate that 12% FF has the potential to reduce obesity through the beneficial regulation of the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Mengdi Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
| | - Jianhua Liu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
| | - Chen Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
| | - Jianwei Gao
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
| | - Chuanhui Xu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (X.W.); (C.C.); (H.W.); (J.P.)
| | - Xiaoyu Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (X.W.); (C.C.); (H.W.); (J.P.)
| | - Tiesheng Xu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
| | - Chenbin Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (X.W.); (C.C.); (H.W.); (J.P.)
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (X.W.); (C.C.); (H.W.); (J.P.)
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (X.W.); (C.C.); (H.W.); (J.P.)
- The Cooperative Innovation Centre for Sustainable Pig Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Rong Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.Z.); (J.L.); (C.L.); (J.G.); (T.X.)
- Correspondence: ; Tel.: +86-134-1952-7039
| |
Collapse
|
133
|
López-Armada MJ, Fernández-Rodríguez JA, Blanco FJ. Mitochondrial Dysfunction and Oxidative Stress in Rheumatoid Arthritis. Antioxidants (Basel) 2022; 11:antiox11061151. [PMID: 35740048 PMCID: PMC9220001 DOI: 10.3390/antiox11061151] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
Control of excessive mitochondrial oxidative stress could provide new targets for both preventive and therapeutic interventions in the treatment of chronic inflammation or any pathology that develops under an inflammatory scenario, such as rheumatoid arthritis (RA). Increasing evidence has demonstrated the role of mitochondrial alterations in autoimmune diseases mainly due to the interplay between metabolism and innate immunity, but also in the modulation of inflammatory response of resident cells, such as synoviocytes. Thus, mitochondrial dysfunction derived from several danger signals could activate tricarboxylic acid (TCA) disruption, thereby favoring a vicious cycle of oxidative/mitochondrial stress. Mitochondrial dysfunction can act through modulating innate immunity via redox-sensitive inflammatory pathways or direct activation of the inflammasome. Besides, mitochondria also have a central role in regulating cell death, which is deeply altered in RA. Additionally, multiple evidence suggests that pathological processes in RA can be shaped by epigenetic mechanisms and that in turn, mitochondria are involved in epigenetic regulation. Finally, we will discuss about the involvement of some dietary components in the onset and progression of RA.
Collapse
Affiliation(s)
- María José López-Armada
- Grupo de Investigación en Envejecimiento e Inflamación (ENVEINF), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain;
- Correspondence: (M.J.L.-A.); (F.J.B.); Tel./Fax: +34-981-178272-73 (M.J.L.-A.)
| | - Jennifer Adriana Fernández-Rodríguez
- Grupo de Investigación en Envejecimiento e Inflamación (ENVEINF), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain;
| | - Francisco Javier Blanco
- Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación de Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña, 15001 A Coruña, Spain
- Correspondence: (M.J.L.-A.); (F.J.B.); Tel./Fax: +34-981-178272-73 (M.J.L.-A.)
| |
Collapse
|
134
|
Deutsch L, Debevec T, Millet GP, Osredkar D, Opara S, Šket R, Murovec B, Mramor M, Plavec J, Stres B. Urine and Fecal 1H-NMR Metabolomes Differ Significantly between Pre-Term and Full-Term Born Physically Fit Healthy Adult Males. Metabolites 2022; 12:metabo12060536. [PMID: 35736470 PMCID: PMC9228004 DOI: 10.3390/metabo12060536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/04/2022] Open
Abstract
Preterm birth (before 37 weeks gestation) accounts for ~10% of births worldwide and remains one of the leading causes of death in children under 5 years of age. Preterm born adults have been consistently shown to be at an increased risk for chronic disorders including cardiovascular, endocrine/metabolic, respiratory, renal, neurologic, and psychiatric disorders that result in increased death risk. Oxidative stress was shown to be an important risk factor for hypertension, metabolic syndrome and lung disease (reduced pulmonary function, long-term obstructive pulmonary disease, respiratory infections, and sleep disturbances). The aim of this study was to explore the differences between preterm and full-term male participants' levels of urine and fecal proton nuclear magnetic resonance (1H-NMR) metabolomes, during rest and exercise in normoxia and hypoxia and to assess general differences in human gut-microbiomes through metagenomics at the level of taxonomy, diversity, functional genes, enzymatic reactions, metabolic pathways and predicted gut metabolites. Significant differences existed between the two groups based on the analysis of 1H-NMR urine and fecal metabolomes and their respective metabolic pathways, enabling the elucidation of a complex set of microbiome related metabolic biomarkers, supporting the idea of distinct host-microbiome interactions between the two groups and enabling the efficient classification of samples; however, this could not be directed to specific taxonomic characteristics.
Collapse
Affiliation(s)
- Leon Deutsch
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
| | - Tadej Debevec
- Faculty of Sports, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
- Department of Automation, Biocybernetics and Robotics, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Gregoire P. Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland;
| | - Damjan Osredkar
- Department of Pediatric Neurology, University Children’s Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Simona Opara
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
| | - Robert Šket
- Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Boštjan Murovec
- Faculty of Electrical Engineering, University of Ljubljana, Jamova 2, SI-1000 Ljubljana, Slovenia;
| | - Minca Mramor
- Department of Infectious Diseases, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Janez Plavec
- National Institute of Chemistry, NMR Center, SI-1000 Ljubljana, Slovenia;
| | - Blaz Stres
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
- Department of Automation, Biocybernetics and Robotics, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
- Institute of Sanitary Engineering, Faculty of Civil and Geodetic Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-4156-7633
| |
Collapse
|
135
|
Kong E, Li Y, Deng M, Hua T, Yang M, Li J, Feng X, Yuan H. Glycometabolism Reprogramming of Glial Cells in Central Nervous System: Novel Target for Neuropathic Pain. Front Immunol 2022; 13:861290. [PMID: 35669777 PMCID: PMC9163495 DOI: 10.3389/fimmu.2022.861290] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain is characterized by hyperalgesia and allodynia. Inflammatory response is conducive to tissue recovery upon nerve injury, but persistent and exaggerated inflammation is detrimental and participates in neuropathic pain. Synaptic transmission in the nociceptive pathway, and particularly the balance between facilitation and inhibition, could be affected by inflammation, which in turn is regulated by glial cells. Importantly, glycometabolism exerts a vital role in the inflammatory process. Glycometabolism reprogramming of inflammatory cells in neuropathic pain is characterized by impaired oxidative phosphorylation in mitochondria and enhanced glycolysis. These changes induce phenotypic transition of inflammatory cells to promote neural inflammation and oxidative stress in peripheral and central nervous system. Accumulation of lactate in synaptic microenvironment also contributes to synaptic remodeling and central sensitization. Previous studies mainly focused on the glycometabolism reprogramming in peripheral inflammatory cells such as macrophage or lymphocyte, little attention was paid to the regulation effects of glycometabolism reprogramming on the inflammatory responses in glial cells. This review summarizes the evidences for glycometabolism reprogramming in peripheral inflammatory cells, and presents a small quantity of present studies on glycometabolism in glial cells, expecting to promote the exploration in glycometabolism in glial cells of neuropathic pain.
Collapse
Affiliation(s)
- Erliang Kong
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China.,Department of Anesthesiology, The No. 988 Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou, China
| | - Yongchang Li
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Mengqiu Deng
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Tong Hua
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Mei Yang
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jian Li
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xudong Feng
- Department of Anesthesiology, The No. 988 Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
136
|
Ding Q, Lu C, Hao Q, Zhang Q, Yang Y, Olsen RE, Ringo E, Ran C, Zhang Z, Zhou Z. Dietary Succinate Impacts the Nutritional Metabolism, Protein Succinylation and Gut Microbiota of Zebrafish. Front Nutr 2022; 9:894278. [PMID: 35685883 PMCID: PMC9171437 DOI: 10.3389/fnut.2022.894278] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Succinate is widely used in the food and feed industry as an acidulant, flavoring additive, and antimicrobial agent. This study investigated the effects of dietary succinate on growth, energy budget, nutritional metabolism, protein succinylation, and gut microbiota composition of zebrafish. Zebrafish were fed a control-check (0% succinate) or four succinate-supplemented diets (0.05, 0.10, 0.15, and 0.2%) for 4 weeks. The results showed that dietary succinate at the 0.15% additive amount (S0.15) can optimally promote weight gain and feed intake. Whole body protein, fat, and energy deposition increased in the S0.15 group. Fasting plasma glucose level decreased in fish fed the S0.15 diet, along with improved glucose tolerance. Lipid synthesis in the intestine, liver, and muscle increased with S0.15 feeding. Diet with 0.15% succinate inhibited intestinal gluconeogenesis but promoted hepatic gluconeogenesis. Glycogen synthesis increased in the liver and muscle of S0.15-fed fish. Glycolysis was increased in the muscle of S0.15-fed fish. In addition, 0.15% succinate-supplemented diet inhibited protein degradation in the intestine, liver, and muscle. Interestingly, different protein succinylation patterns in the intestine and liver were observed in fish fed the S0.15 diet. Intestinal proteins with increased succinylation levels were enriched in the tricarboxylic acid cycle while proteins with decreased succinylation levels were enriched in pathways related to fatty acid and amino acid degradation. Hepatic proteins with increased succinylation levels were enriched in oxidative phosphorylation while proteins with decreased succinylation levels were enriched in the processes of protein processing and transport in the endoplasmic reticulum. Finally, fish fed the S0.15 diet had a higher abundance of Proteobacteria but a lower abundance of Fusobacteria and Cetobacterium. In conclusion, dietary succinate could promote growth and feed intake, promote lipid anabolism, improve glucose homeostasis, and spare protein. The effects of succinate on nutritional metabolism are associated with alterations in the levels of metabolic intermediates, transcriptional regulation, and protein succinylation levels. However, hepatic fat accumulation and gut microbiota dysbiosis induced by dietary succinate suggest potential risks of succinate application as a feed additive for fish. This study would be beneficial in understanding the application of succinate as an aquatic feed additive.
Collapse
Affiliation(s)
- Qianwen Ding
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chenyao Lu
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiang Hao
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingshuang Zhang
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Rolf Erik Olsen
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Ringo
- Norwegian College of Fishery Science, Faculty of Bioscience, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Zhen Zhang,
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Zhigang Zhou,
| |
Collapse
|
137
|
Monfort-Ferré D, Caro A, Menacho M, Martí M, Espina B, Boronat-Toscano A, Nuñez-Roa C, Seco J, Bautista M, Espín E, Megía A, Vendrell J, Fernández-Veledo S, Serena C. The Gut Microbiota Metabolite Succinate Promotes Adipose Tissue Browning in Crohn's Disease. J Crohns Colitis 2022; 16:1571-1583. [PMID: 35554517 PMCID: PMC9624294 DOI: 10.1093/ecco-jcc/jjac069] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/17/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] is associated with complex microbe-host interactions, involving changes in microbial communities, and gut barrier defects, leading to the translocation of microorganisms to surrounding adipose tissue [AT]. We evaluated the presence of beige AT depots in CD and questioned whether succinate and/or bacterial translocation promotes white-to-beige transition in adipocytes. METHODS Visceral [VAT] and subcutaneous [SAT] AT biopsies, serum and plasma were obtained from patients with active [n = 21] or inactive [n = 12] CD, and from healthy controls [n = 15]. Adipose-derived stem cells [ASCs] and AT macrophages [ATMs] were isolated from VAT biopsies. RESULTS Plasma succinate levels were significantly higher in patients with active CD than in controls and were intermediate in those with inactive disease. Plasma succinate correlated with the inflammatory marker high-sensitivity C-reactive protein. Expression of the succinate receptor SUCNR1 was higher in VAT, ASCs and ATMs from the active CD group than from the inactive or control groups. Succinate treatment of ASCs elevated the expression of several beige AT markers from controls and from patients with inactive disease, including uncoupling protein-1 [UCP1]. Notably, beige AT markers were prominent in ASCs from patients with active CD. Secretome profiling revealed that ASCs from patients with active disease secrete beige AT-related proteins, and co-culture assays showed that bacteria also trigger the white-to-beige switch of ASCs from patients with CD. Finally, AT depots from patients with CD exhibited a conversion from white to beige AT together with high UCP1 expression, which was corroborated by in situ thermal imaging analysis. CONCLUSIONS Succinate and bacteria trigger white-to-beige AT transition in CD. Understanding the role of beige AT in CD might aid in the development of therapeutic or diagnostic interventions.
Collapse
Affiliation(s)
- Diandra Monfort-Ferré
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Aleidis Caro
- Colorectal Surgery Unit, Hospital Universitari Joan XXIII, Tarragona, Spain
| | | | - Marc Martí
- Colorectal Surgery Unit, General Surgery Service, Hospital Valle de Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Beatriz Espina
- Colorectal Surgery Unit, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Albert Boronat-Toscano
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Cati Nuñez-Roa
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Seco
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Michelle Bautista
- Digestive Unit, Hospital Universitari Joan XXIII, 43007, Tarragona, Spain
| | - Eloy Espín
- Colorectal Surgery Unit, General Surgery Service, Hospital Valle de Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Ana Megía
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain,Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Corresponding authors: Sonia Fernández-Veledo, PhD, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain. ;
| | - Carolina Serena
- Carolina Serena, PhD, Hospital Universitari de Tarragona Joan XXIII, Institut d’Investigació Sanitària Pere Virgili, Tarragona, Spain. ;
| |
Collapse
|
138
|
Guillon A, Brea-Diakite D, Cezard A, Wacquiez A, Baranek T, Bourgeais J, Picou F, Vasseur V, Meyer L, Chevalier C, Auvet A, Carballido JM, Nadal Desbarats L, Dingli F, Turtoi A, Le Gouellec A, Fauvelle F, Donchet A, Crépin T, Hiemstra PS, Paget C, Loew D, Herault O, Naffakh N, Le Goffic R, Si-Tahar M. Host succinate inhibits influenza virus infection through succinylation and nuclear retention of the viral nucleoprotein. EMBO J 2022; 41:e108306. [PMID: 35506364 PMCID: PMC9194747 DOI: 10.15252/embj.2021108306] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
Influenza virus infection causes considerable morbidity and mortality, but current therapies have limited efficacy. We hypothesized that investigating the metabolic signaling during infection may help to design innovative antiviral approaches. Using bronchoalveolar lavages of infected mice, we here demonstrate that influenza virus induces a major reprogramming of lung metabolism. We focused on mitochondria‐derived succinate that accumulated both in the respiratory fluids of virus‐challenged mice and of patients with influenza pneumonia. Notably, succinate displays a potent antiviral activity in vitro as it inhibits the multiplication of influenza A/H1N1 and A/H3N2 strains and strongly decreases virus‐triggered metabolic perturbations and inflammatory responses. Moreover, mice receiving succinate intranasally showed reduced viral loads in lungs and increased survival compared to control animals. The antiviral mechanism involves a succinate‐dependent posttranslational modification, that is, succinylation, of the viral nucleoprotein at the highly conserved K87 residue. Succinylation of viral nucleoprotein altered its electrostatic interactions with viral RNA and further impaired the trafficking of viral ribonucleoprotein complexes. The finding that succinate efficiently disrupts the influenza replication cycle opens up new avenues for improved treatment of influenza pneumonia.
Collapse
Affiliation(s)
- Antoine Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, CHRU de Tours, Tours, France
| | - Deborah Brea-Diakite
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Adeline Cezard
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Alan Wacquiez
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Thomas Baranek
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Jérôme Bourgeais
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Frédéric Picou
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Virginie Vasseur
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Léa Meyer
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christophe Chevalier
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Adrien Auvet
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, CHRU de Tours, Tours, France
| | | | | | - Florent Dingli
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Andrei Turtoi
- Tumor Microenvironment Laboratory, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier, France.,Institut du Cancer de Montpellier, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Audrey Le Gouellec
- CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, University Grenoble Alpes, Grenoble, France
| | - Florence Fauvelle
- UGA/INSERM U1216, Grenoble Institute of Neurosciences, Grenoble, France.,UGA/INSERM US17, Grenoble MRI Facility IRMaGe, Grenoble, France
| | - Amélie Donchet
- Institut de Biologie Structurale (IBS), CEA, CNRS, University Grenoble Alpes, Grenoble, France
| | - Thibaut Crépin
- Institut de Biologie Structurale (IBS), CEA, CNRS, University Grenoble Alpes, Grenoble, France
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Christophe Paget
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Damarys Loew
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Olivier Herault
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Nadia Naffakh
- Institut Pasteur, Unité Biologie des ARN et Virus Influenza, CNRS UMR3569, Paris, France
| | - Ronan Le Goffic
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| |
Collapse
|
139
|
Xiao Y, Chen X, Wang Z, Quan J, Zhao X, Tang H, Wu H, Di Q, Wu Z, Chen W. Succinate Is a Natural Suppressor of Antiviral Immune Response by Targeting MAVS. Front Immunol 2022; 13:816378. [PMID: 35309330 PMCID: PMC8924363 DOI: 10.3389/fimmu.2022.816378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/07/2022] [Indexed: 11/21/2022] Open
Abstract
Succinate is at the crossroads of multiple metabolic pathways and plays a role in several immune responses acting as an inflammation signal. However, whether succinate regulates antiviral immune response remains unclear. Here, we found that the production of succinate was reduced in RAW264.7 cells during vesicular stomatitis virus (VSV) infection. Using diethyl succinate to pretreat the mouse peritoneal macrophages and RAW264.7 cells before VSV infection, the production of interferon-β (IFN-β), chemokine (C–X–C motif) ligand 10 (CXCL-10), and IFN-stimulated genes 15 (ISG15) was significantly decreased, following which the VSV replication in diethyl succinate-pretreated cells was obviously increased. Moreover, succinate decreased the expression of IFN-β in serum, lung, and spleen derived from the VSV-infected mice. The overall survival rate in the VSV-infected mice with diethyl succinate pretreatment was also remarkably downregulated. Furthermore, we identified that succinate inhibited the activation of MAVS-TBK1-IRF3 signaling by suppressing the formation of MAVS aggregates. Our findings provide previously unrecognized roles of succinate in antiviral immune response and establish a novel link between metabolism and innate immune response.
Collapse
Affiliation(s)
- Yue Xiao
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Xinyi Chen
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhun Wang
- Technological Center, Changchun Customs, Changchun, China
| | - Jiazheng Quan
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Xibao Zhao
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Haimei Tang
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Han Wu
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Qianqian Di
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zherui Wu
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Weilin Chen
- Department of Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| |
Collapse
|
140
|
Xu J, Zheng Y, Zhao Y, Zhang Y, Li H, Zhang A, Wang X, Wang W, Hou Y, Wang J. Succinate/IL-1β Signaling Axis Promotes the Inflammatory Progression of Endothelial and Exacerbates Atherosclerosis. Front Immunol 2022; 13:817572. [PMID: 35273600 PMCID: PMC8901997 DOI: 10.3389/fimmu.2022.817572] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is an important driver of atherosclerosis. Succinate is a new extracellular inflammatory alarm released by activated macrophages. Succinate is sensed by succinate receptor 1 (Sucnr1) and then transferred to effector cells. It is worth exploring whether succinate is capable of facilitating the inflammatory response in atherosclerosis. In this study, we firstly found that arterial serum of Coronary Heart Disease (CHD) patients contained significantly higher succinate and interleukin (IL)-1β than Health control (HC) subjects, and succinate was positively correlated with IL-1β. As demonstrated by the in vitro study, succinate/hypoxia-inducible factor 1α (Hif)-1α/IL-1β signal axis existed and significantly facilitated the inflammatory program in human umbilical vein endothelial cells (HUVECs). Under the coculture, activated macrophages released succinate, which would be transferred to HUVECs via Sucnr1 and then activate Hif-1α to produce a greater amount of IL-1β. Likewise, the aortic sinus’s inflammatory phenotype was found to be more significant within Apoe-/- mice that were injected with succinate. Furthermore, Sucnr1 inhibitor (NF-56-EJ40) could significantly interrupt succinate/IL-1β signal in HUVECs and macrophages. As revealed by this study, glycolytic metabolism following the release of succinate could be found in atherosclerotic pathology, and succinate would drive succinate/IL-1β signal dependent on Sucnr1 and then exacerbate inflammatory responses. Sucnr1 might be a novel target for cutting off the transduction of succinate signal to prevent the inflammation of atherosclerosis.
Collapse
Affiliation(s)
- Jingwen Xu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yabing Zheng
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yaqing Zhao
- College of Second Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yujiao Zhang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Huilin Li
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - An Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuehan Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Weizong Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yinglong Hou
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiangrong Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
141
|
Fearon U, Hanlon MM, Floudas A, Veale DJ. Cellular metabolic adaptations in rheumatoid arthritis and their therapeutic implications. Nat Rev Rheumatol 2022; 18:398-414. [PMID: 35440762 DOI: 10.1038/s41584-022-00771-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Activation of endothelium and immune cells is fundamental to the initiation of autoimmune diseases such as rheumatoid arthritis (RA), and it results in trans-endothelial cell migration and synovial fibroblast proliferation, leading to joint destruction. In RA, the synovial microvasculature is highly dysregulated, resulting in inefficient oxygen perfusion to the synovium, which, along with the high metabolic demands of activated immune and stromal cells, leads to a profoundly hypoxic microenvironment. In inflamed joints, infiltrating immune cells and synovial resident cells have great requirements for energy and nutrients, and they adapt their metabolic profiles to generate sufficient energy to support their highly activated inflammatory states. This shift in metabolic capacity of synovial cells enables them to produce the essential building blocks to support their proliferation, activation and invasiveness. Furthermore, it results in the accumulation of metabolic intermediates and alteration of redox-sensitive pathways, affecting signalling pathways that further potentiate the inflammatory response. Importantly, the inflamed synovium is a multicellular tissue, with cells differing in their metabolic requirements depending on complex cell-cell interactions, nutrient supply, metabolic intermediates and transcriptional regulation. Therefore, understanding the complex interplay between metabolic and inflammatory pathways in synovial cells in RA will provide insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland. .,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland.
| | - Megan M Hanlon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
142
|
Koenig A, Buskiewicz-Koenig IA. Redox Activation of Mitochondrial DAMPs and the Metabolic Consequences for Development of Autoimmunity. Antioxid Redox Signal 2022; 36:441-461. [PMID: 35352943 PMCID: PMC8982130 DOI: 10.1089/ars.2021.0073] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Reactive oxygen species (ROS) are well known to promote innate immune responses during and in the absence of microbial infections. However, excessive or prolonged exposure to ROS provokes innate immune signaling dysfunction and contributes to the pathogenesis of many autoimmune diseases. The relatively high basal expression of pattern recognition receptors (PRRs) in innate immune cells renders them prone to activation in response to minor intrinsic or extrinsic ROS misbalances in the absence of pathogens. Critical Issues: A prominent source of ROS are mitochondria, which are also major inter-organelle hubs for innate immunity activation, since most PRRs and downstream receptor molecules are directly located either at mitochondria or at mitochondria-associated membranes. Due to their ancestral bacterial origin, mitochondria can also act as quasi-intrinsic self-microbes that mimic a pathogen invasion and become a source of danger-associated molecular patterns (DAMPs) that triggers innate immunity from within. Recent Advances: The release of mitochondrial DAMPs correlates with mitochondrial metabolism changes and increased generation of ROS, which can lead to the oxidative modification of DAMPs. Recent studies suggest that ROS-modified mitochondrial DAMPs possess increased, persistent immunogenicity. Future Directions: Herein, we discuss how mitochondrial DAMP release and oxidation activates PRRs, changes cellular metabolism, and causes innate immune response dysfunction by promoting systemic inflammation, thereby contributing to the onset or progression of autoimmune diseases. The future goal is to understand what the tipping point for DAMPs is to become oxidized, and whether this is a road without return. Antioxid. Redox Signal. 36, 441-461.
Collapse
Affiliation(s)
- Andreas Koenig
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | | |
Collapse
|
143
|
Anders CB, Lawton TM, Smith HL, Garret J, Doucette MM, Ammons MCB. Use of integrated metabolomics, transcriptomics, and signal protein profile to characterize the effector function and associated metabotype of polarized macrophage phenotypes. J Leukoc Biol 2022; 111:667-693. [PMID: 34374126 PMCID: PMC8825884 DOI: 10.1002/jlb.6a1120-744r] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 06/24/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022] Open
Abstract
MΦs display remarkable plasticity and the ability to activate diverse responses to a host of intracellular and external stimuli. Despite extensive characterization of M1 MΦs and a broad set of M2 MΦs, comprehensive characterization of functional phenotype and associated metabotype driving this diverse MΦ activation remains. Herein, an ex vivo model was utilized to produce 6 MΦ functional phenotypes. Isolated CD14+ PBMCs were differentiated into resting M0 MΦs, and then polarized into M1 (IFN-γ/LPS), M2a (IL-4/IL-13), M2b (IC/LPS), M2c (IL-10), and M2d (IL-6/LIF) MΦs. The MΦs were profiled using a bioanalyte matrix of 4 cell surface markers, ∼50 secreted proteins, ∼800 expressed myeloid genes, and ∼450 identified metabolites relative to M0 MΦs. Signal protein and expressed gene profiles grouped the MΦs into inflammatory (M1 and M2b) and wound resolution (M2a, M2c, and M2d) phenotypes; however, each had a unique metabolic profile. While both M1 and M2b MΦs shared metabotype profiles consistent with an inflammatory signature; key differences were observed in the TCA cycle, FAO, and OXPHOS. Additionally, M2a, M2c, and M2d MΦs all profiled as tissue repair MΦs; however, metabotype differences were observed in multiple pathways including hexosamine, polyamine, and fatty acid metabolism. These metabolic and other key functional distinctions suggest phagocytic and proliferative functions for M2a MΦs, and angiogenesis and ECM assembly capabilities for M2b, M2c, and M2d MΦs. By integrating metabolomics into a systems analysis of MΦ phenotypes, we provide the most comprehensive map of MΦ diversity to date, along with the global metabolic shifts that correlate to MΦ functional plasticity in these phenotypes.
Collapse
Affiliation(s)
- Catherine B. Anders
- Idaho Veteran’s Research and Education Foundation (IVREF); Boise VA Medical Center (BVAMC), Boise, ID 83702; USA
| | - Tyler M.W. Lawton
- Idaho Veteran’s Research and Education Foundation (IVREF); Boise VA Medical Center (BVAMC), Boise, ID 83702; USA
| | - Hannah L. Smith
- Idaho Veteran’s Research and Education Foundation (IVREF); Boise VA Medical Center (BVAMC), Boise, ID 83702; USA, Department of Microbiology and Immunology; Montana State University, Bozeman, MT, ZIP 59717; USA
| | - Jamie Garret
- Idaho Veteran’s Research and Education Foundation (IVREF); Boise VA Medical Center (BVAMC), Boise, ID 83702; USA,School of Medicine, University of Washington, Seattle, WA, ZIP 98195; USA
| | - Margaret M. Doucette
- Department of Physical Medicine & Rehabilitation, Boise VA Medical Center (BVAMC), Boise, ID 83702; USA
| | - Mary Cloud B. Ammons
- Idaho Veteran’s Research and Education Foundation (IVREF); Boise VA Medical Center (BVAMC), Boise, ID 83702; USA
| |
Collapse
|
144
|
Schiattarella GG, Alcaide P, Condorelli G, Gillette TG, Heymans S, Jones EAV, Kallikourdis M, Lichtman A, Marelli-Berg F, Shah S, Thorp EB, Hill JA. Immunometabolic Mechanisms of Heart Failure with Preserved Ejection Fraction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:211-222. [PMID: 35755006 PMCID: PMC9229992 DOI: 10.1038/s44161-022-00032-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing in prevalence worldwide, already accounting for at least half of all heart failure (HF). As most patients with HFpEF are obese with metabolic syndrome, metabolic stress has been implicated in syndrome pathogenesis. Recently, compelling evidence for bidirectional crosstalk between metabolic stress and chronic inflammation has emerged, and alterations in systemic and cardiac immune responses are held to participate in HFpEF pathophysiology. Indeed, based on both preclinical and clinical evidence, comorbidity-driven systemic inflammation, coupled with metabolic stress, have been implicated together in HFpEF pathogenesis. As metabolic alterations impact immune function(s) in HFpEF, major changes in immune cell metabolism are also recognized in HFpEF and in HFpEF-predisposing conditions. Both arms of immunity - innate and adaptive - are implicated in the cardiomyocyte response in HFpEF. Indeed, we submit that crosstalk among adipose tissue, the immune system, and the heart represents a critical component of HFpEF pathobiology. Here, we review recent evidence in support of immunometabolic mechanisms as drivers of HFpEF pathogenesis, discuss pivotal biological mechanisms underlying the syndrome, and highlight questions requiring additional inquiry.
Collapse
Affiliation(s)
- Gabriele G. Schiattarella
- Center for Cardiovascular Research (CCR), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gianluigi Condorelli
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Cardio Center, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Elizabeth A. V. Jones
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marinos Kallikourdis
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Andrew Lichtman
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sanjiv Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Edward B. Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
145
|
SUCNR1 Mediates the Priming Step of the Inflammasome in Intestinal Epithelial Cells: Relevance in Ulcerative Colitis. Biomedicines 2022; 10:biomedicines10030532. [PMID: 35327334 PMCID: PMC8945150 DOI: 10.3390/biomedicines10030532] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 12/10/2022] Open
Abstract
Intestinal epithelial cells (IECs) constitute a defensive physical barrier in mucosal tissues and their disruption is involved in the etiopathogenesis of several inflammatory pathologies, such as Ulcerative Colitis (UC). Recently, the succinate receptor SUCNR1 was associated with the activation of inflammatory pathways in several cell types, but little is known about its role in IECs. We aimed to analyze the role of SUCNR1 in the inflammasome priming and its relevance in UC. Inflammatory and inflammasome markers and SUCNR1 were analyzed in HT29 cells treated with succinate and/or an inflammatory cocktail and transfected with SUCNR1 siRNA in a murine DSS model, and in intestinal resections from 15 UC and non-IBD patients. Results showed that this receptor mediated the inflammasome, priming both in vitro in HT29 cells and in vivo in a murine chronic DSS-colitis model. Moreover, SUNCR1 was also found to be involved in the activation of the inflammatory pathways NFкB and ERK pathways, even in basal conditions, since the transient knock-down of this receptor significantly reduced the constitutive levels of pERK-1/2 and pNFкB and impaired LPS-induced inflammation. Finally, UC patients showed a significant increase in the expression of SUCNR1 and several inflammasome components which correlated positively and significantly. Therefore, our results demonstrated a role for SUCNR1 in basal and stimulated inflammatory pathways in intestinal epithelial cells and suggested a pivotal role for this receptor in inflammasome activation in UC.
Collapse
|
146
|
Gauthier T, Chen W. Modulation of Macrophage Immunometabolism: A New Approach to Fight Infections. Front Immunol 2022; 13:780839. [PMID: 35154105 PMCID: PMC8825490 DOI: 10.3389/fimmu.2022.780839] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Macrophages are essential innate immune cells that contribute to host defense during infection. An important feature of macrophages is their ability to respond to extracellular cues and to adopt different phenotypes and functions in response to these stimuli. The evidence accumulated in the last decade has highlighted the crucial role of metabolic reprogramming during macrophage activation in infectious context. Thus, understanding and manipulation of macrophage immunometabolism during infection could be of interest to develop therapeutic strategies. In this review, we focus on 5 major metabolic pathways including glycolysis, pentose phosphate pathway, fatty acid oxidation and synthesis, tricarboxylic acid cycle and amino acid metabolism and discuss how they sustain and regulate macrophage immune function in response to parasitic, bacterial and viral infections as well as trained immunity. At the end, we assess whether some drugs including those used in clinic and in development can target macrophage immunometabolism for potential therapy during infection with an emphasis on SARS-CoV2 infection.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wanjun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
147
|
Alves RW, da Silva EM, Doretto-Silva L, Andrade-Oliveira V. Metabolic Pathways in Immune Cells Commitment and Fate. ESSENTIAL ASPECTS OF IMMUNOMETABOLISM IN HEALTH AND DISEASE 2022:53-82. [DOI: 10.1007/978-3-030-86684-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
148
|
Wangchuk P, Anderson D, Yeshi K, Loukas A. Identification of Small Molecules of the Infective Stage of Human Hookworm Using LCMS-Based Metabolomics and Lipidomics Protocols. ACS Infect Dis 2021; 7:3264-3276. [PMID: 34767348 DOI: 10.1021/acsinfecdis.1c00428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hookworm infections affect millions of people worldwide and are responsible for impaired mental and physical growth in children, and anemias. There is no vaccine, and increasing anthelmintic drug resistance in nematodes of domestic animals, and reduced drug cure rates in nematode infections of humans is alarming. Despite this looming health problem, there is a significant knowledge gap in terms of nonproteinaceous "excretory/secretory products" (ESPs) and how they orchestrate a parasitic existence. In the current study, we have conducted the first metabolomic and lipidomic analysis of the infective third-stage filariform larvae (L3) of the predominant human hookworm Necator americanus using liquid chromatography-mass spectrometry. Altogether, we have identified a total of 645 small molecules that were mainly produced through amino acid and glycerophospholipid metabolism. Putatively, 495 metabolites were unique to the somatic tissue extract, and 34 metabolites were present only in the ESP component. More than 21 novel mass features with nitrogen and sulfur functional groups were detected in the ESP component for the first time from helminths. While this study could not establish the biological functions of the metabolites identified, literature searches revealed that these metabolites possess various biological properties, including anti-inflammatory activities. These metabolites are likely used by the parasite upon exposure to a host to facilitate skin penetration, passage through different tissues, and immune regulation in the small bowel. Overall, the results presented herein offer significant insight into the metabolome of N. americanus L3 and have the potential to instigate future work to establish biomarkers of infection. This area urgently needs attention, given the lack of sensitive point-of-care diagnostic tools.
Collapse
Affiliation(s)
- Phurpa Wangchuk
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Road, Smithfield, Cairns, Queensland 4878, Australia
| |
Collapse
|
149
|
Diethyl Succinate Modulates Microglial Polarization and Activation by Reducing Mitochondrial Fission and Cellular ROS. Metabolites 2021; 11:metabo11120854. [PMID: 34940612 PMCID: PMC8705220 DOI: 10.3390/metabo11120854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/14/2022] Open
Abstract
Succinate is a metabolite in the tricarboxylic acid cycle (TCA) which plays a central role in mitochondrial activity. Excess succinate is known to be transported out of the cytosol, where it activates a succinate receptor (SUCNR1) to enhance inflammation through macrophages in various contexts. In addition, the intracellular role of succinate beyond an intermediate metabolite and prior to its extracellular release is also important to the polarization of macrophages. However, the role of succinate in microglial cells has not been characterized. Lipopolysaccharide (LPS) stimulates the elevation of intracellular succinate levels. To reveal the function of intracellular succinate associated with LPS-stimulated inflammatory response in microglial cells, we assessed the levels of ROS, cytokine production and mitochondrial fission in the primary microglia pretreated with cell-permeable diethyl succinate mimicking increased intracellular succinate. Our results suggest that elevated intracellular succinate exerts a protective role in the primary microglia by preventing their conversion into the pro-inflammatory M1 phenotype induced by LPS. This protective effect is SUCNR1-independent and mediated by reduced mitochondrial fission and cellular ROS production.
Collapse
|
150
|
Fajar JK, Ilmawan M, Mamada SS, Mutiawati E, Husnah M, Yusuf H, Nainu F, Sirinam S, Keam S, Ophinni Y, Rosiello F, Fahriani M, Rosa SGV. Global prevalence of persistent neuromuscular symptoms and the possible pathomechanisms in COVID-19 recovered individuals: A systematic review and meta-analysis. NARRA J 2021; 1:e48. [PMID: 38450213 PMCID: PMC10914045 DOI: 10.52225/narra.v1i3.48] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/26/2021] [Indexed: 02/05/2023]
Abstract
This study was conducted to determine the prevalence of prolonged neuromuscular symptoms, including fatigue, anosmia, headache, myalgia, and joint pain in COVID-19 survivors hospitalized with mild, moderate, or severe infections worldwide. The search was conducted up to January 30th, 2021 using three databases (PubMed, Scopus, and Web of Science) to identify potentially eligible studies. Data on study characteristics, follow-up characteristics, and severity of COVID-19 during hospitalization were collected in accordance with PRISMA guidelines. The Newcastle-Ottawa scale was used to assess the quality of relevant articles. The estimated prevalence of specific prolonged neuromuscular symptoms and the association between COVID-19 severity and occurrence of prolonged neuromuscular symptoms was analyzed wherever appropriate. Database search yielded 4,050 articles and 22 articles were included for meta-analysis. The estimated prevalence of prolonged fatigue was recorded in 21.2% (95%CI: 11.9%- 34.8%) of 3,730 COVID-19 survivors. Persistent anosmia was recorded in 239 of 2,600 COVID-19 survivors (9.7%, 95%CI: 6.1%-15.2%). In 84 out of 2,412 COVID-19 survivors (8.9%, 95%CI: 3.2%-22.6%), prolonged headache was observed. A total of 53 out of 1,125 COVID-19 patients (5.6%, 95%CI: 2.1%-14.2%) complained of persistent myalgia even after being discharged from the hospital. The prevalence of prolonged joint pain was in 15.4% (95%CI: 8.2%-27.2%) of subjects. Due to data scarcity on COVID-19 severity and prolonged neuromuscular symptoms, association analysis could not be conducted. Widespread concern regarding long-term impacts of COVID-19 was raised after several studies reported prolonged symptoms in COVID-19 survivors. Numerous theories have been proposed to address this concern; however, as the research on this pandemic is still ongoing, no explanation is definitive yet. Therefore, follow-up studies in COVID-19 survivors after recovery from COVID-19 are warranted to determine the pathogenesis of prolonged symptoms. PROSPERO registration: CRD42021242332.
Collapse
Affiliation(s)
- Jonny K Fajar
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | | | - Sukamto S Mamada
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar, Indonesia
| | - Endang Mutiawati
- Department of Neurology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Milda Husnah
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Hanifah Yusuf
- Department of Pharmacology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar, Indonesia
| | - Salin Sirinam
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Synat Keam
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Youdiil Ophinni
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Harvard University, Cambridge, MA, USA
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Francesco Rosiello
- Department of Public Health and Infectious Disease, Sapienza-University of Rome, Rome, Italy
| | - Marhami Fahriani
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Sandro G V Rosa
- Diretoria de Patentes, Divisão De Farmácia - Instituto Nacional Da Propriedade Industrial, Rio de Janeiro, Brasil
- Programa De Pós-Graduação Em Ciências Aplicadas a Produtos Para Saúde, Faculdade De Farmácia, Universidade Federal Fluminense, Brasil
| |
Collapse
|