1
|
Cheung ST, Do Y, Kim E, Rella A, Goyarts E, Pernodet N, Wong YH. G Protein-Coupled Receptors in Skin Aging. J Invest Dermatol 2025; 145:749-765.e8. [PMID: 39186022 DOI: 10.1016/j.jid.2024.06.1288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 08/27/2024]
Abstract
Skin aging is a complex biological process affected by a plethora of intrinsic and extrinsic factors that alter cutaneous functions through the modulations of signaling pathways and responses. Expressed in various cell types and skin tissue layers, G protein-coupled receptors (GPCRs) play a vital role in regulating skin aging. We have cataloged 156 GPCRs expressed in the skin and reviewed their roles in skin aging, such as pigmentation, loss of elasticity, wrinkles, rough texture, and aging-associated skin disorders. By exploring the GPCRs found in the skin, it may be possible to develop new treatment regimens for aging-associated skin conditions using GPCR ligands.
Collapse
Affiliation(s)
- Suet Ting Cheung
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yelim Do
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Eunah Kim
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Antonella Rella
- Research and Development, The Estée Lauder Companies, New York, New York, USA
| | - Earl Goyarts
- Research and Development, The Estée Lauder Companies, New York, New York, USA
| | - Nadine Pernodet
- Research and Development, The Estée Lauder Companies, New York, New York, USA; Estée Lauder Research Laboratories, Melville, New York, USA
| | - Yung Hou Wong
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China; Molecular Neuroscience Center, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
2
|
Su W, Zhang D, Wang Y, Lei L, Li H. G protein-coupled receptor 91 activations suppressed mineralization in Porphyromonas gingivalis-infected osteoblasts. Sci Rep 2024; 14:27606. [PMID: 39528607 PMCID: PMC11554824 DOI: 10.1038/s41598-024-78944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Succinate receptor GPR91 is one of the G protein-coupled receptors (GPCRs) that interacts with various proteins to regulate diverse cellular functions such as cell morphology, apoptosis, and differentiation. In this study, we investigated whether the GPR91-mediated signaling pathway regulates mineralization in Porphyromonas gingivalis (P. gingivalis)-treated osteoblasts and to determine its potential role in osteoclast differentiation. Primary mouse osteoblasts from wild-type (WT) and GPR91 knockout (GPR91-/-) mice infected with P. gingivalis were used for in vitro experiments. The results showed that inhibition by 4C, a specific inhibitor, and GPR91 knockout promoted mineralization in P. gingivalis-infected osteoblasts. Surprisingly, GPR91 knockdown decreased the migration ability of osteoblasts. Moreover, compared with P. gingivalis-infected WT osteoblasts, GPR91-/- osteoblasts exhibited decreased RANKL production, and conditioned media (CM) from bacteria-infected GPR91-/- osteoblasts suppressed the formation of osteoclast precursors. Moreover, P. gingivalis mediated the role of GPR91 in osteoblast mineralization by activating the NF-κB pathway. These findings suggest that GPR91 activation reduces mineralization of P. gingivalis-infected osteoblasts and promotes osteoclastogenesis in macrophages. Therefore, targeting GPR91 may mitigate the loss of alveolar bone during bacterial infection.
Collapse
Affiliation(s)
- Wenqi Su
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Dandan Zhang
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Yujia Wang
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Lang Lei
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Houxuan Li
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China.
| |
Collapse
|
3
|
Ben-Baruch Morgenstern N, Rochman M, Kotliar M, Dunn JLM, Mack L, Besse J, Natale MA, Klingler AM, Felton JM, Caldwell JM, Barski A, Rothenberg ME. Single-cell RNA-sequencing of human eosinophils in allergic inflammation in the esophagus. J Allergy Clin Immunol 2024; 154:974-987. [PMID: 38871184 PMCID: PMC11456386 DOI: 10.1016/j.jaci.2024.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/08/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Eosinophils are elusive cells involved in allergic inflammation. Single-cell RNA-sequencing (scRNA-seq) is an emerging approach to deeply characterize cellular properties, heterogeneity, and functionality. OBJECTIVES We sought to comprehensively characterize the transcriptome and biological functions of human eosinophils at a site of severe allergic inflammation in the esophagus (ie, eosinophilic esophagitis [EoE]). METHODS We employed a gravity-based scRNA-seq methodology to sequence blood eosinophils from patients with EoE and control individuals compared to a reanalyzed public scRNA-seq dataset of human esophageal eosinophils of EoE patients. We used flow cytometry, immunostaining, and a stimulation assay to verify mRNA findings. RESULTS In total, scRNA-seq was obtained from 586 eosinophils (188 from blood [n = 6 individuals] and 398 from esophagus [n = 6 individuals]). The esophageal eosinophils were composed of a population of activated eosinophils (enriched in 659 genes compared with peripheral blood-associated eosinophils) and a small population of eosinophils resembling peripheral blood eosinophils (enriched in 62 genes compared with esophageal eosinophils). Esophageal eosinophils expressed genes involved in sensing and responding to diverse stimuli, most notably IFN-γ, IL-10, histamine and leukotrienes, and succinate. Esophageal eosinophils were most distinguished from other esophageal populations by gene expression of the receptors CCR3, HRH4, SUCNR1, and VSTM1; transcription factors CEBPE, OLIG1, and OLIG2; protease PRSS33; and the hallmark eosinophil gene CLC. A web of bidirectional eosinophil interactions with other esophageal populations was derived. Comparing esophageal eosinophils and mast cells revealed that esophageal eosinophils expressed genes involved in DNAX-activation protein-12 (also known as TYROBP) interactions, IgG receptor-triggered events, immunoregulation, and IL-10 signaling. CONCLUSIONS In EoE, esophageal eosinophils exist as 2 populations, a minority population resembling blood eosinophils and the other population characterized by high de novo transcription of diverse sensing receptors and inflammatory mediators readying them to potentially intersect with diverse cell types.
Collapse
Affiliation(s)
- Netali Ben-Baruch Morgenstern
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mark Rochman
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael Kotliar
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julia L M Dunn
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lydia Mack
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - John Besse
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mia A Natale
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Andrea M Klingler
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jennifer M Felton
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julie M Caldwell
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Artem Barski
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
4
|
Huang H, Li G, He Y, Chen J, Yan J, Zhang Q, Li L, Cai X. Cellular succinate metabolism and signaling in inflammation: implications for therapeutic intervention. Front Immunol 2024; 15:1404441. [PMID: 38933270 PMCID: PMC11200920 DOI: 10.3389/fimmu.2024.1404441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Succinate, traditionally viewed as a mere intermediate of the tricarboxylic acid (TCA) cycle, has emerged as a critical mediator in inflammation. Disruptions within the TCA cycle lead to an accumulation of succinate in the mitochondrial matrix. This excess succinate subsequently diffuses into the cytosol and is released into the extracellular space. Elevated cytosolic succinate levels stabilize hypoxia-inducible factor-1α by inhibiting prolyl hydroxylases, which enhances inflammatory responses. Notably, succinate also acts extracellularly as a signaling molecule by engaging succinate receptor 1 on immune cells, thus modulating their pro-inflammatory or anti-inflammatory activities. Alterations in succinate levels have been associated with various inflammatory disorders, including rheumatoid arthritis, inflammatory bowel disease, obesity, and atherosclerosis. These associations are primarily due to exaggerated immune cell responses. Given its central role in inflammation, targeting succinate pathways offers promising therapeutic avenues for these diseases. This paper provides an extensive review of succinate's involvement in inflammatory processes and highlights potential targets for future research and therapeutic possibilities development.
Collapse
Affiliation(s)
- Hong Huang
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gejing Li
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yini He
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Chen
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jianye Yan
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qin Zhang
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liqing Li
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Central Research Laboratory, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| | - Xiong Cai
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
5
|
Griepke S, Trauelsen M, Nilsson MD, Hansen J, Steffensen LB, Schwartz TW, Ketelhuth DFJ. G-Protein-Coupled Receptor 91-Dependent Signalling Does Not Influence Vascular Inflammation and Atherosclerosis in Hyperlipidaemic Mice. Cells 2023; 12:2580. [PMID: 37947659 PMCID: PMC10647868 DOI: 10.3390/cells12212580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
The TCA cycle intermediate metabolite 'succinate' has been proposed as an inflammatory mediator, influencing autoimmunity and allergic reactions, through ligation to its sensing receptor SUCNR1/GPR91. Whether GPR91-mediated signalling influences the chronic inflammatory process of atherosclerosis has never been investigated. The examination of publicly available datasets revealed that the SUCNR1 gene is expressed in human atherosclerotic plaques, especially in vascular smooth muscle cells. Using GPR91 knockout (Gpr91-/-) and wildtype (WT) littermates, made hyperlipidaemic with the overexpression of the gain-of-function mutated Pcsk9 and Western diet feeding, we showed that the full ablation of GPR91 did not accelerate atherosclerosis-lesions in the aortic arch 2.18 ± 0.48% vs. 1.64 ± 0.31%, and in the aortic roots 10.06 ± 0.91% vs. 10.67 ± 1.53% for Gpr91-/- and WT mice, respectively. In line with this, no differences between groups were observed for macrophage and T-cell infiltration in the plaque, as well as the polarization towards M1- or M2-like macrophages in the aorta, spleen and liver of Gpr91-/- and WT control mice. In conclusion, our study indicates that the global ablation of GPR91 signalling does not influence vascular inflammation or atherogenesis.
Collapse
Affiliation(s)
- Silke Griepke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.G.); (M.D.N.); (J.H.); (L.B.S.)
| | - Mette Trauelsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (M.T.); (T.W.S.)
| | - Michelle D. Nilsson
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.G.); (M.D.N.); (J.H.); (L.B.S.)
| | - Jakob Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.G.); (M.D.N.); (J.H.); (L.B.S.)
| | - Lasse B. Steffensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.G.); (M.D.N.); (J.H.); (L.B.S.)
| | - Thue W. Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (M.T.); (T.W.S.)
| | - Daniel F. J. Ketelhuth
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.G.); (M.D.N.); (J.H.); (L.B.S.)
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Bioclinicum, Solna, 171 64 Stockholm, Sweden
| |
Collapse
|
6
|
Höper T, Karkossa I, Dumit VI, von Bergen M, Schubert K, Haase A. A comparative proteomics analysis of four contact allergens in THP-1 cells shows distinct alterations in key metabolic pathways. Toxicol Appl Pharmacol 2023; 475:116650. [PMID: 37541627 DOI: 10.1016/j.taap.2023.116650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Allergic contact dermatitis (ACD) is the predominant form of immunotoxicity in humans. The sensitizing potential of chemicals can be assessed in vitro. However, a better mechanistic understanding could improve the current OECD-validated test battery. The aim of this study was to get insights into toxicity mechanisms of four contact allergens, p-benzoquinone (BQ), 2,4-dinitrochlorobenzene (DNCB), p-nitrobenzyl bromide (NBB) and NiSO4, by analyzing differential proteome alterations in THP-1 cells using two common proteomics workflows, stable isotope labeling by amino acids in cell culture (SILAC) and label-free quantification (LFQ). Here, SILAC was found to deliver more robust results. Overall, the four allergens induced similar responses in THP-1 cells, which underwent profound metabolic reprogramming, including a striking upregulation of the TCA cycle accompanied by pronounced induction of the Nrf2 oxidative stress response pathway. The magnitude of induction varied between the allergens with DNCB and NBB being most potent. A considerable overlap between transcriptome-based signatures of the GARD assay and the proteins identified in our study was found. When comparing the results of this study to a previous proteomics study in human primary monocyte-derived dendritic cells, we found a rather low share in regulated proteins. However, on pathway level, the overlap was high, indicating that affected pathways rather than single proteins are more eligible to investigate proteomic changes induced by contact allergens. Overall, this study confirms the potential of proteomics to obtain a profound mechanistic understanding, which may help improving existing in vitro assays for skin sensitization.
Collapse
Affiliation(s)
- Tessa Höper
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany; Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Leipzig, Germany
| | - Verónica I Dumit
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Leipzig, Germany; Institute of Biochemistry, Leipzig University, Leipzig, Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Leipzig, Germany
| | - Andrea Haase
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany.
| |
Collapse
|
7
|
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: 5] [Impact Index Per Article: 1.7] [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
|
8
|
Forde B, Yao L, Shaha R, Murphy S, Lunjani N, O'Mahony L. Immunomodulation by foods and microbes: Unravelling the molecular tango. Allergy 2022; 77:3513-3526. [PMID: 35892227 PMCID: PMC10087875 DOI: 10.1111/all.15455] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Metabolic health and immune function are intimately connected via diet and the microbiota. Nearly 90% of all immune cells in the body are associated with the gastrointestinal tract and these immune cells are continuously exposed to a wide range of microbes and microbial-derived compounds, with important systemic ramifications. Microbial dysbiosis has consistently been observed in patients with atopic dermatitis, food allergy and asthma and the molecular mechanisms linking changes in microbial populations with disease risk and disease endotypes are being intensively investigated. The discovery of novel bacterial metabolites that impact immune function is at the forefront of host-microbe research. Co-evolution of microbial communities within their hosts has resulted in intertwined metabolic pathways that affect physiological and pathological processes. However, recent dietary and lifestyle changes are thought to negatively influence interactions between microbes and their host. This review provides an overview of some of the critical metabolite-receptor interactions that have been recently described, which may underpin the immunomodulatory effects of the microbiota, and are of relevance for allergy, asthma and infectious diseases.
Collapse
Affiliation(s)
- Brian Forde
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | - Lu Yao
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | - Rupin Shaha
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | | | - Nonhlanhla Lunjani
- APC Microbiome Ireland, UCC, Cork, Ireland.,University of Cape Town, Cape Town, South Africa
| | - Liam O'Mahony
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland.,Department of Medicine, UCC, Cork, Ireland
| |
Collapse
|
9
|
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: 19] [Impact Index Per Article: 6.3] [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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Trauelsen M, Hiron TK, Lin D, Petersen JE, Breton B, Husted AS, Hjorth SA, Inoue A, Frimurer TM, Bouvier M, O'Callaghan CA, Schwartz TW. Extracellular succinate hyperpolarizes M2 macrophages through SUCNR1/GPR91-mediated Gq signaling. Cell Rep 2021; 35:109246. [PMID: 34133934 DOI: 10.1016/j.celrep.2021.109246] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/31/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Succinate functions both as a classical TCA cycle metabolite and an extracellular metabolic stress signal sensed by the mainly Gi-coupled succinate receptor SUCNR1. In the present study, we characterize and compare effects and signaling pathways activated by succinate and both classes of non-metabolite SUCNR1 agonists. By use of specific receptor and pathway inhibitors, rescue in G-protein-depleted cells and monitoring of receptor G protein activation by BRET, we identify Gq rather than Gi signaling to be responsible for SUCNR1-mediated effects on basic transcriptional regulation. Importantly, in primary human M2 macrophages, in which SUCNR1 is highly expressed, we demonstrate that physiological concentrations of extracellular succinate act through SUCNR1-activated Gq signaling to efficiently regulate transcription of immune function genes in a manner that hyperpolarizes their M2 versus M1 phenotype. Thus, sensing of stress-induced extracellular succinate by SUCNR1 is an important transcriptional regulator in human M2 macrophages through Gq signaling.
Collapse
Affiliation(s)
- Mette Trauelsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark
| | - Thomas K Hiron
- Wellcome Trust Centre for Human Genetics and NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Da Lin
- Wellcome Trust Centre for Human Genetics and NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Jacob E Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark
| | - Billy Breton
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Anna Sofie Husted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark
| | - Siv A Hjorth
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Thomas M Frimurer
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Chris A O'Callaghan
- Wellcome Trust Centre for Human Genetics and NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Maersk Tower, 2200 Copenhagen, Denmark.
| |
Collapse
|
14
|
Prag HA, Gruszczyk AV, Huang MM, Beach TE, Young T, Tronci L, Nikitopoulou E, Mulvey JF, Ascione R, Hadjihambi A, Shattock MJ, Pellerin L, Saeb-Parsy K, Frezza C, James AM, Krieg T, Murphy MP, Aksentijević D. Mechanism of succinate efflux upon reperfusion of the ischaemic heart. Cardiovasc Res 2021; 117:1188-1201. [PMID: 32766828 PMCID: PMC7983001 DOI: 10.1093/cvr/cvaa148] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS Succinate accumulates several-fold in the ischaemic heart and is then rapidly oxidized upon reperfusion, contributing to reactive oxygen species production by mitochondria. In addition, a significant amount of the accumulated succinate is released from the heart into the circulation at reperfusion, potentially activating the G-protein-coupled succinate receptor (SUCNR1). However, the factors that determine the proportion of succinate oxidation or release, and the mechanism of this release, are not known. METHODS AND RESULTS To address these questions, we assessed the fate of accumulated succinate upon reperfusion of anoxic cardiomyocytes, and of the ischaemic heart both ex vivo and in vivo. The release of accumulated succinate was selective and was enhanced by acidification of the intracellular milieu. Furthermore, pharmacological inhibition, or haploinsufficiency of the monocarboxylate transporter 1 (MCT1) significantly decreased succinate efflux from the reperfused heart. CONCLUSION Succinate release upon reperfusion of the ischaemic heart is mediated by MCT1 and is facilitated by the acidification of the myocardium during ischaemia. These findings will allow the signalling interaction between succinate released from reperfused ischaemic myocardium and SUCNR1 to be explored.
Collapse
Affiliation(s)
- Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Anja V Gruszczyk
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Margaret M Huang
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Timothy E Beach
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Timothy Young
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Laura Tronci
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Efterpi Nikitopoulou
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Raimondo Ascione
- Bristol Medical School and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Anna Hadjihambi
- Département de Physiologie, Université de Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland
| | - Michael J Shattock
- King’s College London, British Heart Foundation Centre of Excellence, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, 146 Rue Leo Saignat, Bordeaux 33076, France
- Inserm U1082, Université de Poitiers, 2 Rue de la Miletrie, Poitiers 86021, France
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Andrew M James
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Dunja Aksentijević
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
15
|
Succinate Receptor 1: An Emerging Regulator of Myeloid Cell Function in Inflammation. Trends Immunol 2020; 42:45-58. [PMID: 33279412 DOI: 10.1016/j.it.2020.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
The rapidly evolving area of immunometabolism has shed new light on the fundamental properties of products and intermediates of cellular metabolism (metabolites), highlighting their key signaling roles in cell-to-cell communication. Recent evidence identifies the succinate-succinate receptor 1 (SUCNR1) axis as an essential regulator of tissue homeostasis. Succinate signaling via SUCNR1 guides divergent responses in immune cells, which are tissue and context dependent. Herein, we explore the main cellular pathways regulated by the succinate-SUCNR1 axis and focus on the biology of SUCNR1 and its roles influencing the function of myeloid cells. Hence, we identify new therapeutic targets and putative therapeutic approaches aimed at resolving detrimental myeloid cell responses in tissues, including those occurring in the persistently inflamed central nervous system (CNS).
Collapse
|
16
|
Saeb-Parsy K, Martin JL, Summers DM, Watson CJE, Krieg T, Murphy MP. Mitochondria as Therapeutic Targets in Transplantation. Trends Mol Med 2020; 27:185-198. [PMID: 32952044 DOI: 10.1016/j.molmed.2020.08.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022]
Abstract
Advances in surgical procedures, technology, and immune suppression have transformed organ transplantation. However, the metabolic changes that occur during organ retrieval, storage, and implantation have been relatively neglected since the developments many decades ago of cold storage organ preservation solutions. In this review we discuss how the metabolic changes that occur within the organ during transplantation, particularly those associated with mitochondria, may contribute to the outcome. We show how a better understanding of these processes can lead to changes in surgical practice and the development of new drug classes to improve the function and longevity of transplanted grafts, while increasing the pool of organs available for transplantation.
Collapse
Affiliation(s)
- Kourosh Saeb-Parsy
- Department of Surgery and Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, CB2 2QQ, UK; NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge Biomedical Campus, Cambridge, UK
| | - Jack L Martin
- Department of Surgery and Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, CB2 2QQ, UK; NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge Biomedical Campus, Cambridge, UK
| | - Dominic M Summers
- Department of Surgery and Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, CB2 2QQ, UK; NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge Biomedical Campus, Cambridge, UK
| | - Christopher J E Watson
- Department of Surgery and Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, CB2 2QQ, UK; NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge Biomedical Campus, Cambridge, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK; Medical Research Council (MRC) Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, UK.
| |
Collapse
|
17
|
Zhang J, Zhang Q, Yang Y, Wang Q. Association Between Succinate Receptor SUCNR1 Expression and Immune Infiltrates in Ovarian Cancer. Front Mol Biosci 2020; 7:150. [PMID: 33062639 PMCID: PMC7488939 DOI: 10.3389/fmolb.2020.00150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/18/2020] [Indexed: 12/23/2022] Open
Abstract
Background The activation of succinate receptor 1 (SUCNR1) by extracellular succinate has been found to regulate immune cell function. However, the clinical significance of SUCNR1 in ovarian cancer and its correlation with tumor-infiltrating lymphocytes remain unclear. Methods The genetic alteration and expression patterns of SUCNR1 were analyzed by using cBioPortal and Gene Expression Omnibus (GEO) datasets. Kaplan-Meier Plotter was used to assess the prognostic value of SUCNR1 in patients with ovarian cancer. The correlations between SUCNR1 expression and immune infiltration, gene markers of immune cells, cytokines, chemokines, or T cell exhaustion were explored by using TIMER and TISIDB platforms. We also performed Gene Set Enrichment Analysis (GSEA) to reveal biological function of SUCNR1 in ovarian cancer. Results The expression of SUCNR1 was closely related to tumor infiltrating lymphocytes, multiple gene markers of immune cells, and T cell exhaustion in ovarian cancer. The expression of SUCNR1 was also associated with the expression of cytokine- or chemokine-related genes. Moreover, GSEA revealed that various immune-related pathways might be regulated by SUCNR1. In addition, we found that SUCNR1 was amplified in ovarian cancer, and the high expression of SUCNR1 predicted worse progression-free survival (p = 0.0073, HR = 1.49, 95% CI = 1.11–2). Conclusion These results highlight the role of SUCNR1 in regulating tumor immunity in ovarian cancer.
Collapse
Affiliation(s)
- Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinyi Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongbin Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Inhibition of GPR91 Reduces Inflammatory Mediators Involved in Active Labor in Myometrium. Mediators Inflamm 2020; 2020:6454282. [PMID: 32377163 PMCID: PMC7180404 DOI: 10.1155/2020/6454282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/25/2020] [Accepted: 03/12/2020] [Indexed: 12/29/2022] Open
Abstract
Results GPR91 mRNA expression was significantly higher in myometrium from women during term spontaneous labor compared to no labor. Likewise, in mice, GPR91 mRNA expression was significantly upregulated in myometrium during inflammation-induced preterm labor compared to preterm no labor. In myometrial cells, IL1B and TNF significantly increased GPR91 mRNA expression. Knockdown of GPR91 by siRNA in myometrial cells significantly suppressed the secretion and/or expression of IL1B- and TNF-induced proinflammatory cytokines (GM-CSF, IL1A, IL1B, and IL6) and chemokines (CXCL8 and CCL2), myometrial contractility (expression of the contraction-associated proteins PTGFR and CX43, secretion of the uterotonic PGF2α, and in situ collagen gel contraction), and the transcription factor NF-κB. Conclusion Our findings demonstrate that GPR91 is involved in the genesis of proinflammatory and prolabor mediators induced by IL1B or TNF and collectively suggest that GPR91 may contribute to augmentation of the labor processes.
Collapse
|
19
|
Sugita K, Akdis CA. Recent developments and advances in atopic dermatitis and food allergy. Allergol Int 2020; 69:204-214. [PMID: 31648922 DOI: 10.1016/j.alit.2019.08.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 02/07/2023] Open
Abstract
This review highlights recent advances in atopic dermatitis (AD) and food allergy (FA), particularly on molecular mechanisms and disease endotypes, recent developments in global strategies for the management of patients, pipeline for future treatments, primary and secondary prevention and psychosocial aspects. During the recent years, there has been major advances in personalized/precision medicine linked to better understanding of disease pathophysiology and precision treatment options of AD. A greater understanding of the molecular and cellular mechanisms of AD through substantial progress in epidemiology, genetics, skin immunology and psychological aspects resulted in advancements in the precision management of AD. However, the implementation of precision medicine in the management of AD still requires the validation of reliable biomarkers, which will provide more tailored management, starting from prevention strategies towards targeted therapies for more severe diseases. Cutaneous exposure to food via defective barriers is an important route of sensitization to food allergens. Studies on the role of the skin barrier genes demonstrated their association with the development of IgE-mediated FA, and suggest novel prevention and treatment strategies for type 2 diseases in general because of their link to barrier defects not only in AD and FA, but also in asthma, chronic rhinosinusitis, allergic rhinitis and inflammatory bowel disease. The development of more accurate diagnostic tools, biomarkers for early prediction, and innovative solutions require a better understanding of molecular mechanisms and the pathophysiology of FA. Based on these developments, this review provides an overview of novel developments and advances in AD and FA, which are reported particularly during the last two years.
Collapse
|
20
|
Guo Y, Cho SW, Saxena D, Li X. Multifaceted Actions of Succinate as a Signaling Transmitter Vary with Its Cellular Locations. Endocrinol Metab (Seoul) 2020; 35:36-43. [PMID: 32207262 PMCID: PMC7090288 DOI: 10.3803/enm.2020.35.1.36] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 01/05/2023] Open
Abstract
Since the identification of succinate's receptor in 2004, studies supporting the involvement of succinate signaling through its receptor in various diseases have accumulated and most of these investigations have highlighted succinate's pro-inflammatory role. Taken with the fact that succinate is an intermediate metabolite in the center of mitochondrial activity, and considering its potential regulation of protein succinylation through succinyl-coenzyme A, a review on the overall multifaceted actions of succinate to discuss whether and how these actions relate to the cellular locations of succinate is much warranted. Mechanistically, it is important to consider the sources of succinate, which include somatic cellular released succinate and those produced by the microbiome, especially the gut microbiota, which is an equivalent, if not greater contributor of succinate levels in the body. Continue learning the critical roles of succinate signaling, known and unknown, in many pathophysiological conditions is important. Furthermore, studies to delineate the regulation of succinate levels and to determine how succinate elicits various types of signaling in a temporal and spatial manner are also required.
Collapse
Affiliation(s)
- Yuqi Guo
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
- Perlmutter Cancer Institute, New York, NY, USA
| | - Xin Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
- Perlmutter Cancer Institute, New York, NY, USA
- Department of Urology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
21
|
Abstract
There is now a wealth of evidence showing that communication between microbiota and the host is critical to sustain the vital functions of the healthy host, and disruptions of this homeostatic coexistence are known to be associated with a range of diseases including obesity and type 2 diabetes. Microbiota-derived metabolites act both as nutrients and as messenger molecules and can signal to distant organs in the body to shape host pathophysiology. In this review, we provide a new perspective on succinate as a gut microbiota-derived metabolite with a key role governing intestinal homeostasis and energy metabolism. Thus, succinate is not merely a major intermediary of the TCA traditionally considered as an extracellular danger signal in the host, but also a by-product of some bacteria and a primary cross-feeding metabolite between gut resident microbes. In addition to maintain a healthy microbiome, specific functions of microbiota-derived succinate in peripheral tissues regulating host nutrient metabolism should not be rule out. Indeed, recent research point to some probiotic interventions directed to modulate succinate levels in the intestinal lumen, as a new microbiota-based therapies to treat obesity and related co-morbidities. While further research is essential, a large body of evidence point to succinate as a new strategic mediator in the microbiota-host cross-talk, which might provide the basis for new therapeutically approaches in a near future.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Departament of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch, 4, 43007, Tarragona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.
| | - Joan Vendrell
- Departament of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch, 4, 43007, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Rovira i Virgili University, Tarragona, Spain
| |
Collapse
|
22
|
Mantuano NR, Oliveira-Nunes MC, Alisson-Silva F, Dias WB, Todeschini AR. Emerging role of glycosylation in the polarization of tumor-associated macrophages. Pharmacol Res 2019; 146:104285. [PMID: 31132403 DOI: 10.1016/j.phrs.2019.104285] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/02/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022]
Abstract
Tumors are formed by several cell types interacting in a complex environment of soluble and matrix molecules. The crosstalk between the cells and extracellular components control tumor fate. Macrophages are highly plastic and diverse immune cells that are known to be key regulators of this complex network, which is mostly because they can adjust their metabolism and reprogram their phenotype and effector function. Here, we review the studies that disclose the central role of metabolism and tumor microenvironment in shaping the phenotype and function of macrophages, highlighting the importance of the hexosamine biosynthetic pathway. We further discuss growing evidence of nutrient-sensitive protein modifications such as O-GlcNAcylation and extracellular glycosylation in the function and polarization of tumor-associated macrophages.
Collapse
Affiliation(s)
- Natalia Rodrigues Mantuano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Bloco D sala 03 CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Maria Cecilia Oliveira-Nunes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Bloco D sala 03 CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Frederico Alisson-Silva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Bloco D sala 03 CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Wagner Barbosa Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Bloco D sala 03 CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil.
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Bloco D sala 03 CCS, UFRJ, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
23
|
Keiran N, Ceperuelo-Mallafré V, Calvo E, Hernández-Alvarez MI, Ejarque M, Núñez-Roa C, Horrillo D, Maymó-Masip E, Rodríguez MM, Fradera R, de la Rosa JV, Jorba R, Megia A, Zorzano A, Medina-Gómez G, Serena C, Castrillo A, Vendrell J, Fernández-Veledo S. SUCNR1 controls an anti-inflammatory program in macrophages to regulate the metabolic response to obesity. Nat Immunol 2019; 20:581-592. [PMID: 30962591 DOI: 10.1038/s41590-019-0372-7] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/05/2019] [Indexed: 12/14/2022]
Abstract
Succinate is a signaling metabolite sensed extracellularly by succinate receptor 1 (SUNCR1). The accumulation of succinate in macrophages is known to activate a pro-inflammatory program; however, the contribution of SUCNR1 to macrophage phenotype and function has remained unclear. Here we found that activation of SUCNR1 had a critical role in the anti-inflammatory responses in macrophages. Myeloid-specific deficiency in SUCNR1 promoted a local pro-inflammatory phenotype, disrupted glucose homeostasis in mice fed a normal chow diet, exacerbated the metabolic consequences of diet-induced obesity and impaired adipose-tissue browning in response to cold exposure. Activation of SUCNR1 promoted an anti-inflammatory phenotype in macrophages and boosted the response of these cells to type 2 cytokines, including interleukin-4. Succinate decreased the expression of inflammatory markers in adipose tissue from lean human subjects but not that from obese subjects, who had lower expression of SUCNR1 in adipose-tissue-resident macrophages. Our findings highlight the importance of succinate-SUCNR1 signaling in determining macrophage polarization and assign a role to succinate in limiting inflammation.
Collapse
Affiliation(s)
- Noelia Keiran
- Unitat de Recerca, 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
| | - Victoria Ceperuelo-Mallafré
- Unitat de Recerca, 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
| | - Enrique Calvo
- Unitat de Recerca, 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
| | - Maria Isabel Hernández-Alvarez
- Unitat de Recerca, 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.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Miriam Ejarque
- Unitat de Recerca, 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
| | - Catalina Núñez-Roa
- Unitat de Recerca, 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
| | - Daniel Horrillo
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Biología Molecular, Universidad Rey Juan Carlos, Madrid, Spain
| | - Elsa Maymó-Masip
- Unitat de Recerca, 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
| | - M Mar Rodríguez
- Unitat de Recerca, 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
| | - Rosa Fradera
- General and Digestive Surgery Service, Hospital St. Pau i Sta Tecla, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Juan Vladimir de la Rosa
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitaria (IUBIS), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Rosa Jorba
- General and Digestive Surgery Service, Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili, Tarragona, Spain
| | - Ana Megia
- Unitat de Recerca, 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
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Barcelona, Spain
| | - Gema Medina-Gómez
- Departamento de Ciencias Básicas de la Salud, Área de Bioquímica y Biología Molecular, Universidad Rey Juan Carlos, Madrid, Spain
| | - Carolina Serena
- Unitat de Recerca, 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
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitaria (IUBIS), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Joan Vendrell
- Unitat de Recerca, 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
- Unitat de Recerca, 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.
| |
Collapse
|
24
|
Nadjsombati MS, McGinty JW, Lyons-Cohen MR, Jaffe JB, DiPeso L, Schneider C, Miller CN, Pollack JL, Nagana Gowda GA, Fontana MF, Erle DJ, Anderson MS, Locksley RM, Raftery D, von Moltke J. Detection of Succinate by Intestinal Tuft Cells Triggers a Type 2 Innate Immune Circuit. Immunity 2019; 49:33-41.e7. [PMID: 30021144 DOI: 10.1016/j.immuni.2018.06.016] [Citation(s) in RCA: 388] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/25/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
In the small intestine, type 2 responses are regulated by a signaling circuit that involves tuft cells and group 2 innate lymphoid cells (ILC2s). Here, we identified the microbial metabolite succinate as an activating ligand for small intestinal (SI) tuft cells. Sequencing analyses of tuft cells isolated from the small intestine, gall bladder, colon, thymus, and trachea revealed that expression of tuft cell chemosensory receptors is tissue specific. SI tuft cells expressed the succinate receptor (SUCNR1), and providing succinate in drinking water was sufficient to induce a multifaceted type 2 immune response via the tuft-ILC2 circuit. The helminth Nippostrongylus brasiliensis and a tritrichomonad protist both secreted succinate as a metabolite. In vivo sensing of the tritrichomonad required SUCNR1, whereas N. brasiliensis was SUCNR1 independent. These findings define a paradigm wherein tuft cells monitor microbial metabolites to initiate type 2 immunity and suggest the existence of other sensing pathways triggering the response to helminths.
Collapse
Affiliation(s)
- Marija S Nadjsombati
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Miranda R Lyons-Cohen
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - James B Jaffe
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Lucian DiPeso
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Christoph Schneider
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Corey N Miller
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joshua L Pollack
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - G A Nagana Gowda
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA
| | - Mary F Fontana
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - David J Erle
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark S Anderson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
25
|
Murphy MP, Hartley RC. Mitochondria as a therapeutic target for common pathologies. Nat Rev Drug Discov 2018; 17:865-886. [PMID: 30393373 DOI: 10.1038/nrd.2018.174] [Citation(s) in RCA: 533] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although the development of mitochondrial therapies has largely focused on diseases caused by mutations in mitochondrial DNA or in nuclear genes encoding mitochondrial proteins, it has been found that mitochondrial dysfunction also contributes to the pathology of many common disorders, including neurodegeneration, metabolic disease, heart failure, ischaemia-reperfusion injury and protozoal infections. Mitochondria therefore represent an important drug target for these highly prevalent diseases. Several strategies aimed at therapeutically restoring mitochondrial function are emerging, and a small number of agents have entered clinical trials. This Review discusses the opportunities and challenges faced for the further development of mitochondrial pharmacology for common pathologies.
Collapse
Affiliation(s)
- Michael P Murphy
- Medical Research Council (MRC) Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
26
|
Peruzzotti-Jametti L, Pluchino S. Targeting Mitochondrial Metabolism in Neuroinflammation: Towards a Therapy for Progressive Multiple Sclerosis. Trends Mol Med 2018; 24:838-855. [DOI: 10.1016/j.molmed.2018.07.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
|
27
|
Grimolizzi F, Arranz L. Multiple faces of succinate beyond metabolism in blood. Haematologica 2018; 103:1586-1592. [PMID: 29954939 PMCID: PMC6165802 DOI: 10.3324/haematol.2018.196097] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/27/2018] [Indexed: 11/13/2022] Open
Abstract
Succinate is an essential intermediate of the tricarboxylic acid cycle that exerts pleiotropic roles beyond metabolism in both physiological and pathological conditions. Recent evidence obtained in mouse models shows its essential role regulating blood cell function through various mechanisms that include pseudohypoxia responses by hypoxia-inducible factor-1α activation, post-translational modifications like succinylation, and communication mediated by succinate receptor 1. Hence, succinate links metabolism to processes like gene expression and intercellular communication. Interestingly, succinate plays key dual roles during inflammatory responses, leading to net inflammation or anti-inflammation depending on factors like the cellular context. Here, we further discuss current suggestions of the possible contribution of succinate to blood stem cell function and blood formation. Further study will be required in the future to better understand succinate biology in blood cells. This promising field may open new avenues to modulate inflammatory responses and to preserve blood cell homeostasis in the clinical setting.
Collapse
Affiliation(s)
- Franco Grimolizzi
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Norway
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Norway .,Department of Hematology, University Hospital of North Norway, Norway.,Young Associate Investigator, Norwegian Center for Molecular Medicine (NCMM), Tromsø, Norway
| |
Collapse
|
28
|
Serena C, Ceperuelo-Mallafré V, Keiran N, Queipo-Ortuño MI, Bernal R, Gomez-Huelgas R, Urpi-Sarda M, Sabater M, Pérez-Brocal V, Andrés-Lacueva C, Moya A, Tinahones FJ, Fernández-Real JM, Vendrell J, Fernández-Veledo S. Elevated circulating levels of succinate in human obesity are linked to specific gut microbiota. THE ISME JOURNAL 2018; 12:1642-1657. [PMID: 29434314 PMCID: PMC6018807 DOI: 10.1038/s41396-018-0068-2] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/21/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023]
Abstract
Gut microbiota-related metabolites are potential clinical biomarkers for cardiovascular disease (CVD). Circulating succinate, a metabolite produced by both microbiota and the host, is increased in hypertension, ischemic heart disease, and type 2 diabetes. We aimed to analyze systemic levels of succinate in obesity, a major risk factor for CVD, and its relationship with gut microbiome. We explored the association of circulating succinate with specific metagenomic signatures in cross-sectional and prospective cohorts of Caucasian Spanish subjects. Obesity was associated with elevated levels of circulating succinate concomitant with impaired glucose metabolism. This increase was associated with specific changes in gut microbiota related to succinate metabolism: a higher relative abundance of succinate-producing Prevotellaceae (P) and Veillonellaceae (V), and a lower relative abundance of succinate-consuming Odoribacteraceae (O) and Clostridaceae (C) in obese individuals, with the (P + V/O + C) ratio being a main determinant of plasma succinate. Weight loss intervention decreased (P + V/O + C) ratio coincident with the reduction in circulating succinate. In the spontaneous evolution after good dietary advice, alterations in circulating succinate levels were linked to specific metagenomic signatures associated with carbohydrate metabolism and energy production with independence of body weight change. Our data support the importance of microbe-microbe interactions for the metabolite signature of gut microbiome and uncover succinate as a potential microbiota-derived metabolite related to CVD risk.
Collapse
Affiliation(s)
- Carolina Serena
- Unitat de Recerca, 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
| | - Victoria Ceperuelo-Mallafré
- Unitat de Recerca, 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
| | - Noelia Keiran
- Unitat de Recerca, 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
| | - Maria Isabel Queipo-Ortuño
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario de Málaga Virgen de la Victoria, Universidad de Málaga, 29010, Málaga, Spain
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Bernal
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Medicina Interna, IBIMA, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Ricardo Gomez-Huelgas
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Medicina Interna, IBIMA, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Mireia Urpi-Sarda
- Biomarkers and Nutrimetabolomics Laboratory, Nutrition, Food Science and Gastronomy Department, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, 08028, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, 28028, Spain
| | - Mónica Sabater
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
| | - Vicente Pérez-Brocal
- Genomics and Health Area, Foundation for the Promotion of Sanitary and Biomedical Research (FISABIO), València, Spain
- CIBER de Epidemiology y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Andrés-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Nutrition, Food Science and Gastronomy Department, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, 08028, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, 28028, Spain
| | - Andres Moya
- Genomics and Health Area, Foundation for the Promotion of Sanitary and Biomedical Research (FISABIO), València, Spain
- CIBER de Epidemiology y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Integrative Systems Biology, Universitat de València, València, Spain
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario de Málaga Virgen de la Victoria, Universidad de Málaga, 29010, Málaga, Spain
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Manuel Fernández-Real
- CIBER de Obesidad y Nutrición (CIBERObN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
| | - Joan Vendrell
- Unitat de Recerca, 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
- Unitat de Recerca, 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.
| |
Collapse
|
29
|
Activation of intestinal tuft cell-expressed Sucnr1 triggers type 2 immunity in the mouse small intestine. Proc Natl Acad Sci U S A 2018; 115:5552-5557. [PMID: 29735652 DOI: 10.1073/pnas.1720758115] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The hallmark features of type 2 mucosal immunity include intestinal tuft and goblet cell expansion initiated by tuft cell activation. How infectious agents that induce type 2 mucosal immunity are detected by tuft cells is unknown. Published microarray analysis suggested that succinate receptor 1 (Sucnr1) is specifically expressed in tuft cells. Thus, we hypothesized that the succinate-Sucnr1 axis may be utilized by tuft cells to detect certain infectious agents. Here we confirmed that Sucnr1 is specifically expressed in intestinal tuft cells but not in other types of intestinal epithelial cells, and demonstrated that dietary succinate induces tuft and goblet cell hyperplasia via Sucnr1 and the tuft cell-expressed chemosensory signaling elements gustducin and Trpm5. Conventional mice with a genetic Sucnr1 deficiency (Sucnr1-/-) showed diminished immune responses to treatment with polyethylene glycol and streptomycin, which are known to enhance microbiota-derived succinate, but responded normally to inoculation with the parasitic worm Nippostrongylus brasiliensis that also produces succinate. Thus, Sucnr1 is required for microbiota-induced but not for a generalized worm-induced type 2 immunity.
Collapse
|
30
|
Abstract
Traditionally cellular respiration or metabolism has been viewed as catabolic and anabolic pathways generating energy and biosynthetic precursors required for growth and general cellular maintenance. However, growing literature provides evidence of a much broader role for metabolic reactions and processes in controlling immunological effector functions. Much of this research into immunometabolism has focused on macrophages, cells that are central in pro- as well as anti-inflammatory responses—responses that in turn are a direct result of metabolic reprogramming. As we learn more about the precise role of metabolic pathways and pathway intermediates in immune function, a novel opportunity to target immunometabolism therapeutically has emerged. Here, we review the current understanding of the regulation of macrophage function through metabolic remodeling.
Collapse
Affiliation(s)
- Ciana Diskin
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Science Institute, Dublin, Ireland
| | - Eva M Pålsson-McDermott
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Science Institute, Dublin, Ireland
| |
Collapse
|
31
|
Ryan DG, O'Neill LAJ. Krebs cycle rewired for macrophage and dendritic cell effector functions. FEBS Lett 2017; 591:2992-3006. [PMID: 28685841 DOI: 10.1002/1873-3468.12744] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 01/03/2025]
Abstract
The Krebs cycle is an amphibolic pathway operating in the mitochondrial matrix of all eukaryotic organisms. In response to proinflammatory stimuli, macrophages and dendritic cells undergo profound metabolic remodelling to support the biosynthetic and bioenergetic requirements of the cell. Recently, it has been discovered that this metabolic shift also involves the rewiring of the Krebs cycle to regulate cellular metabolic flux and the accumulation of Krebs cycle intermediates, notably, citrate, succinate and fumarate. Interestingly, a new role for Krebs cycle intermediates as signalling molecules and immunomodulators that dictate the inflammatory response has begun to emerge. This review will discuss the latest developments in Krebs cycle rewiring and immune cell effector functions, with a particular focus on the regulation of cytokine production.
Collapse
Affiliation(s)
- Dylan Gerard Ryan
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| |
Collapse
|
32
|
Rubić-Schneider T, Carballido-Perrig N, Regairaz C, Raad L, Jost S, Rauld C, Christen B, Wieczorek G, Kreutzer R, Dawson J, Lametschwandner G, Littlewood-Evans A, Carballido JM. GPR91 deficiency exacerbates allergic contact dermatitis while reducing arthritic disease in mice. Allergy 2017; 72:444-452. [PMID: 27527650 PMCID: PMC5324651 DOI: 10.1111/all.13005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2016] [Indexed: 12/12/2022]
Abstract
Background Succinate, in addition to its role as an intermediary of the citric acid cycle, acts as an alarmin, initiating and propagating danger signals resulting from tissue injury or inflammatory stimuli. The contribution of this immune sensing pathway to the development of allergic and inflammatory responses is unknown. Methods Ear thickness of wild‐type (wt) and Sucnr1‐deficient (Sucnr1−/−) mice, sensitized and challenged with oxazolone, was used as a criterion to assess the relevance of SUCNR1/GPR91 expression mediating allergic contact dermatitis (ACD). Results obtained in this system were contrasted with data generated using passive cutaneous anaphylaxis, ovalbumin‐induced asthma and arthritis models. Results We found augmented ACD reactions in Sucnr1−/− mice. This observation correlated with increased mast cell activation in vitro and in vivo. However, exacerbated mast cell activation in Sucnr1−/− mice did not contribute to the enhancement of asthma or arthritis and seemed to be due to alterations during mast cell development as augmented mast cell responses could be recapitulated in wt mast cells differentiated in the absence of succinate. Conclusions A deficiency in succinate sensing during mast cell development confers these cells with a hyperactive phenotype. Such a phenomenon does not translate into exacerbation of asthma or mast cell‐dependent arthritis. On the contrary, the fact that Sucnr1−/− mice developed reduced arthritic disease, using two different in vivo models, indicates that GPR91 antagonists may have therapeutic potential for the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- T. Rubić-Schneider
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
- Former NIBR; Vienna Austria
| | - N. Carballido-Perrig
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
- Former NIBR; Vienna Austria
| | - C. Regairaz
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | - L. Raad
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | | | - C. Rauld
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | - B. Christen
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | - G. Wieczorek
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | - R. Kreutzer
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | - J. Dawson
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
| | | | | | - J. M. Carballido
- Novartis Institutes for Biomedical Research (NIBR); Basel Switzerland
- Former NIBR; Vienna Austria
| |
Collapse
|