1
|
Zhao X, Li Y, Yang L, Chen X, Zhang J, Chen T, Wang H, Li F, Cheng C, Wu J, Cong J, Yin W, Li J, Wang X. GPR35 prevents drug-induced liver injury via the Gαs-cAMP-PKA axis in macrophages. Cell Mol Life Sci 2025; 82:219. [PMID: 40437264 PMCID: PMC12119454 DOI: 10.1007/s00018-025-05751-4] [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: 12/10/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 06/01/2025]
Abstract
Acetaminophen (APAP) overdose induces acute liver injury and represents the most frequent cause of drug-induced liver injury worldwide. Macrophage-mediated inflammation plays detrimental roles during the early stage of liver injury. However, the potential targets regulating inflammation to improve drug-induced liver injury remains undefined. In this study, we reported that G protein-coupled receptor 35 (GPR35) improves drug-induced liver injury by blocking macrophage-mediated inflammation via the Gαs-cyclic AMP-protein kinase A (Gαs-cAMP-PKA) pathway. The ablation of GPR35 exacerbates APAP-induced liver injury, characterized by higher levels of alanine aminotransferase and aspartate aminotransferase in sera, larger damaged areas, and increased levels of pro-inflammatory cytokines. More hepatic macrophages appeared in the inflamed liver of mice with GPR35 deficiency. In contrast, the agonists of GPR35 alleviated APAP-induced liver injury. The depletion of macrophages abolished GPR35-mediated protection. Mechanistically, GPR35 ablation facilitated the activation of pro-inflammatory AKT, MAPK, and NF-κB signaling pathways at the downstream of Toll-like receptors in macrophages. GPR35 agonists activated Gαs-cAMP-PKA signaling to inhibit the activation of these pro-inflammatory signaling pathways and then suppress the inflammatory response in macrophages. Thus, our findings demonstrate that GPR35 prevents drug-induced liver injury by blocking macrophage-mediated inflammation via the Gαs-cAMP-PKA pathway, indicating that GPR35 is a potential target for the development of novel medicines that control drug-induced liver injury.
Collapse
Affiliation(s)
- Xueqin Zhao
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yuanhao Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Liu Yang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xi Chen
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jialong Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tong Chen
- School of Life Sciences, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Haoqi Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fei Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chen Cheng
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Wu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jingjing Cong
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Wenwei Yin
- Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| | - Jing Li
- School of Life Sciences, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Xuefu Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
2
|
Wu SN, Wang YJ, Gao ZH, Liutkevičienė R, Rovite V. Recent Advances in Ionic Mechanisms in Pituitary Cells: Implications for Electrophysiological and Electropharmacological Research. J Clin Med 2025; 14:3117. [PMID: 40364147 PMCID: PMC12072979 DOI: 10.3390/jcm14093117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pituitary cells are specialized cells located within the pituitary gland, a small, pea-sized gland situated at the base of the brain. Through the use of cellular electrophysiological techniques, the electrical properties of these cells have been revealed. This review paper aims to introduce the ion currents that are known to be functionally expressed in pituitary cells. These currents include a voltage-gated Na+ current (INa), erg-mediated K+ current (IK(erg)), M-type K+ current (IK(M)), hyperpolarization-activated cation current (Ih), and large-conductance Ca2+-activated K+ (BKCa) channel. The biophysical characteristics of the respective ion current were described. Additionally, we also provide explanations for the effect of various drugs or compounds on each of these currents. GH3-cell exposure to GV-58 can increase the magnitude of INa with a concurrent rise in the inactivation time constant of the current. The presence of esaxerenone, an antagonist of the aldosterone receptor, directly suppresses the magnitude of peak and late INa. Risperidone, an atypical antipsychotic agent, is effective at suppressing the IK(erg) amplitude directly, and di(2-ethylhexyl)-phthalate suppressed IK(erg). Solifenacin and kynurenic acid can interact with the KM channel to stimulate IK(M), while carisbamate and cannabidiol inhibit the Ih amplitude activated by sustained hyperpolarization. Moreover, the presence of either rufinamide or QO-40 can enhance the activity of single BKCa channels. To summarize, alterations in ion currents within native pituitary cells or pituitary tumor cells can influence their functional activity, particularly in processes like stimulus-secretion coupling. The effects of small-molecule modulators, as demonstrated here, bear significance in clinical, therapeutic, and toxicological contexts.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Research and Education, An-Nan Hospital, China Medical University, No. 66, Section 2, Changhe Road, An Nan District, Tainan 70965, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701401, Taiwan
| | - Ya-Jean Wang
- Department of Senior Services Industry Management, Minghsin University of Science and Technology, Hsinchu 300401, Taiwan
| | - Zi-Han Gao
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701401, Taiwan
| | - Rasa Liutkevičienė
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu 2, 50106 Kaunas, Lithuania
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre (BMC), LV-1067 Riga, Latvia
| |
Collapse
|
3
|
Zheng W, Wang X, Chen H, He K, Yan X, Zhang Y, Yang Y, Zhang P, Zhu W, Zhu S, Li H. KYNA Ameliorates Hepatic Ischemia-Reperfusion Injury by Activating the Hippo Signalling Pathway via FTO-Dependent m6A Demethylation of LATS1. Cell Prolif 2025:e70048. [PMID: 40277037 DOI: 10.1111/cpr.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) substantially influences the prognosis of liver transplant recipients. Although kynurenic acid (KYNA) has been associated with protective effects against ischemia-reperfusion injury in various organs, the precise mechanisms underlying its protective role in HIRI are not well elucidated. In this study, a 70% mouse HIRI model and an in vitro hypoxia/reoxygenation model were employed to examine the protective effects of KYNA on HIRI. In this study, we illustrate that KYNA influences the methylation status of the Hippo signalling pathway by enhancing the expression of the fat mass and obesity-associated gene (FTO). Within this pathway, large tumour suppressor kinase 1 (LATS1) is identified as a direct target of FTO. Moreover, the stability of LATS1 mRNA exhibits an inverse correlation with FTO levels and is modulated through its interaction with YTH N6-Methyladenosine RNA Binding Protein F2 (YTHDF2). The reduction in LATS1 expression facilitated Yes-associated protein (YAP) nuclear translocation, decreased hepatocyte apoptosis, and mitigated HIRI. Clinically, elevated levels of serum KYNA correlate with a diminished severity of liver injury post-transplantation. our work revealed that KYNA possesses significant clinical translational potential for the prevention of HIRI, and further exploration of its underlying mechanisms was conducted.
Collapse
Affiliation(s)
- Wenjie Zheng
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Zhejiang, Hangzhou, China
| | - Xiaowen Wang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Thyroid and Breast Surgery, The Third Affiliated Lingnan Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Haoqi Chen
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kaiming He
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xijing Yan
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Thyroid and Breast Surgery, The Third Affiliated Lingnan Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Zhang
- Department of Pathology, Guangdong Provincial Hospital of Chiese Medicine, Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng Zhang
- Department of Thyroid and Breast Surgery, The Third Affiliated Lingnan Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenfeng Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuguang Zhu
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hua Li
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
4
|
Li Y, Zhang L, Wu D, Zhang Z, Zhou Y, Li J. Kynurenic Acid, a Small Foodborne Molecule with the Potential to Affect Human Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8729-8739. [PMID: 40176362 DOI: 10.1021/acs.jafc.4c12355] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Kynurenic acid (KYNA) is one of the metabolites of tryptophan (Trp), which can be supplied by endogenous synthesis or diet and involves various aspects of nutrition and metabolism. KYNA has been proven to be an antagonist of α7-nicotinic acetylcholine and NMDA receptors, which can regulate the neurotransmission of cholinergic and glutaminergic acid, so KYNA is mainly associated with neurodegenerative diseases. Other metabolites in the Trp and kynurenine metabolic pathways, such as 3-hydroxy-l-kynurenine (3-HK), 5-hydroxytryptophan (5-HT), and quinolinic acid (QA), are also involved in immune regulation and have pro-stimulatory or anti-excitatory toxicity. KYNA is present in both the human body and the daily diet, so it has the potential as a new dietary supplement. It may be of great significance in treating psychiatric disorders.
Collapse
Affiliation(s)
- Yuqi Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Daren Wu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Yongbo Zhou
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| |
Collapse
|
5
|
Elias JE, Debela M, Sewell GW, Stopforth RJ, Partl H, Heissbauer S, Holland LM, Karlsen TH, Kaser A, Kaneider NC. GPR35 prevents osmotic stress induced cell damage. Commun Biol 2025; 8:478. [PMID: 40121360 PMCID: PMC11929815 DOI: 10.1038/s42003-025-07848-9] [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: 07/16/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
GPR35 is an orphan G-protein coupled receptor that has been implicated in the development of cancer. GPR35 regulates the Na+/K+-ATPase's pump and signalling function. Here we show GPR35's critical role in ion flux that in turn controls cellular osmotic pressure and Na+-dependent transport in HepG2 and SW480 cells. GPR35 deficiency results in increased levels of intracellular Na+, osmotic stress and changes in osmolytes leading to increased cells size and decreased glutamine import in vitro and in vivo. The GPR35-T108M risk variant, which increases risk for primary sclerosing cholangitis and inflammatory bowel disease, leads to lower intracellular Na+ levels, and enhanced glutamine uptake. High salt diet (HSD) in wildtype mice resembles the intestinal epithelial phenotype of their Gpr35-/- littermates with decreased Goblet cell size and numbers. This indicates that GPR35's regulation of the Na+/K+-ATPase controls ion homeostasis, osmosis and Na+-dependent transporters.
Collapse
Affiliation(s)
- Joshua E Elias
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Mekdes Debela
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Gavin W Sewell
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Richard J Stopforth
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Hannah Partl
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Sophie Heissbauer
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Lorraine M Holland
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Tom H Karlsen
- Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nicole C Kaneider
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK.
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
6
|
Sánchez-Melgar A, Vultaggio-Poma V, Falzoni S, Fructuoso C, Albasanz JL, Di Virgilio F, Martín M. Mitochondrial Localization and Function of Adenosine Receptors. Int J Biol Sci 2025; 21:1874-1893. [PMID: 40083698 PMCID: PMC11900819 DOI: 10.7150/ijbs.101930] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/11/2025] [Indexed: 03/16/2025] Open
Abstract
G-protein coupled receptors (GPCRs) are typically expressed on the cell surface where they mediate extracellular signals from hormones, neurotransmitters and growth factors, among others. However, growing evidence support the intracellular localization of GPCRs, including mitochondria. In the present work, we assessed the presence and functionality of adenosine receptors in mitochondria by combining techniques such as western blotting, radioligand binding, electron microscopy, enzymatic activities determination, oxygen consumption measurement and 3D morphological analysis of mitochondrial networks. Our results demonstrate the mitochondrial localization of adenosine A1 and A2 receptors in pure mitochondria fractions isolated from mouse brain and liver, human brain, HeLa and SH-SY5Y cells. Adenylyl cyclase activity assays revealed that these receptors are functional in the mitochondria. Moreover, exposure of isolated mitochondria to selective A1, A2A and A2B receptors agonists revealed these receptors as potential modulators of mitochondrial energy metabolism, since ATP production and coupling efficiency increased in the presence of BAY 60-6583 (A2B agonist) whereas proton leak and acute response were higher with CGS 21680 (A2A agonist). Also, proton leak, ATP production, acute response and acute respiration were increased in the presence of CPA (A1 agonist). Interestingly, different mitochondrial morphological changes were detected in HeLa cells exposed to these receptors' agonists.
Collapse
Affiliation(s)
- Alejandro Sánchez-Melgar
- Department of Inorganic, Organic Chemistry and Biochemistry. Faculty of Medicine of Ciudad Real / Faculty of Chemical Sciences and Technologies. Institute of Biomedicine (IB-UCLM). IDISCAM. University of Castilla-La Mancha. Ciudad Real, Spain
| | | | - Simoneta Falzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Clara Fructuoso
- Department of Inorganic, Organic Chemistry and Biochemistry. Faculty of Medicine of Ciudad Real / Faculty of Chemical Sciences and Technologies. Institute of Biomedicine (IB-UCLM). IDISCAM. University of Castilla-La Mancha. Ciudad Real, Spain
| | - José Luis Albasanz
- Department of Inorganic, Organic Chemistry and Biochemistry. Faculty of Medicine of Ciudad Real / Faculty of Chemical Sciences and Technologies. Institute of Biomedicine (IB-UCLM). IDISCAM. University of Castilla-La Mancha. Ciudad Real, Spain
| | | | - Mairena Martín
- Department of Inorganic, Organic Chemistry and Biochemistry. Faculty of Medicine of Ciudad Real / Faculty of Chemical Sciences and Technologies. Institute of Biomedicine (IB-UCLM). IDISCAM. University of Castilla-La Mancha. Ciudad Real, Spain
| |
Collapse
|
7
|
Kirkik D, Hacimustafaoglu F, Gündogdu B, Dogantekin B, Kariksiz M, Kalkanli Tas S. Genetic Susceptibility and Disease Activity in Ankylosing Spondylitis: The Role of G Protein-Coupled Receptor 35rs4676410 Polymorphism in a Turkish Population. Genet Test Mol Biomarkers 2025; 29:32-38. [PMID: 39918909 DOI: 10.1089/gtmb.2024.0482] [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] [Indexed: 02/09/2025] Open
Abstract
Background: Ankylosing spondylitis (AS) is a chronic inflammatory disorder with a significant genetic predisposition. Genome-wide association studies (GWAS) have identified immune-related loci, including the G Protein-Coupled Receptor 35 (GPR35) gene, as potential contributors to AS pathogenesis. This study aimed to evaluate the association between the rs4676410 polymorphism in the GPR35 gene and both AS susceptibility and disease activity in a Turkish population. Methods: This case-control study included 200 participants (100 AS patients and 100 healthy controls). DNA was isolated from blood samples, and the rs4676410 polymorphism was analyzed using real-time polymerase chain reaction (PCR). Disease activity in AS patients was assessed using the Bath AS Functional Index (BASFI), Bath AS Disease Activity Index (BASDAI), and disease activity scores including C-reactive protein (ASDAS-CRP) scores. Statistical analyses were conducted using IBM SPSS 26. Results: The rs4676410 polymorphism was significantly associated with AS susceptibility. The AA genotype and A allele were more prevalent in AS patients, indicating an increased risk of developing AS. Among disease activity measures, ASDAS-CRP scores were significantly higher in patients with the AA genotype (p = 0.043), while no significant differences were observed for BASFI and BASDAI scores across genotypes. Conclusion: The findings suggest that the rs4676410 polymorphism in the GPR35 gene is associated with AS susceptibility and may influence disease activity through elevated inflammatory responses. These results highlight the potential of the AA genotype and A allele as genetic markers for AS and underscore the importance of integrating genetic insights into personalized treatment approaches.
Collapse
Affiliation(s)
- Duygu Kirkik
- Hamidiye Medicine Faculty, Department of Medical Biology, University of Health Sciences, Istanbul, Turkey
- Hamidiye Medicine Faculty, Department of Immunology, University of Health Sciences, Istanbul, Turkey
| | - Fatih Hacimustafaoglu
- Medical Laboratory Techniques Programme, Hamidiye Vocational School of Health Services, University of Health Sciences, Istanbul, Turkey
| | - Barış Gündogdu
- Department of Internal Medicine, University of Health Sciences Sultan Abdulhamid Han Training and Research Hospital, Istanbul, Turkey
| | - Betül Dogantekin
- Department of Internal Medicine, University of Health Sciences Sultan Abdulhamid Han Training and Research Hospital, Istanbul, Turkey
| | - Mesut Kariksiz
- Department of Orthopedic and Traumatology, Cam and Sakura City Hospital, Istanbul, Turkey
| | - Sevgi Kalkanli Tas
- Hamidiye Medicine Faculty, Department of Immunology, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
8
|
Sadeesh EM, Lahamge MS, Malik A, Ampadi AN. Differential Expression of Nuclear-Encoded Mitochondrial Protein Genes of ATP Synthase Across Different Tissues of Female Buffalo. Mol Biotechnol 2025; 67:705-722. [PMID: 38305843 DOI: 10.1007/s12033-024-01085-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
The physiological well-being of buffaloes, encompassing phenotypic traits, reproductive health, and productivity, depends on their energy status. Mitochondria, the architects of energy production, orchestrate a nuanced interplay between nuclear and mitochondrial domains. Oxidative phosphorylation complexes and associated proteins wield significant influence over metabolic functions, energy synthesis, and organelle dynamics, often linked to tissue-specific pathologies. The unexplored role of ATP synthase in buffalo tissues prompted a hypothesis: in-depth exploration of nuclear-derived mitochondrial genes, notably ATP synthase, reveals distinctive tissue-specific diversity. RNA extraction and sequencing of buffalo tissues (kidney, heart, brain, and ovary) enabled precise quantification of nuclear-derived mitochondrial protein gene expression. The analysis unveiled 24 ATP synthase transcript variants, each with unique tissue-specific patterns. Kidney, brain, and heart exhibited elevated gene expression compared to ovaries, with 10, 8, and 19 up-regulated genes, respectively. The kidney showed 3 and 12 down-regulated genes compared to the brain and heart. The heart-brain comparison highlighted ten highly expressed genes in ATP synthase functions. Gene ontology and pathway analyses revealed enriched functions linked to ATP synthesis and oxidative phosphorylation, offering a comprehensive understanding of energy production in buffalo tissues. This analysis enhances understanding of tissue-specific gene expression, emphasizing the influence of energy demands. Revealing intricate links between mitochondrial gene expression and tissue specialization in buffaloes, it provides nuanced insights into tissue-specific expression of nuclear-encoded mitochondrial protein genes, notably ATP synthase, advancing the comprehension of buffalo tissue biology.
Collapse
Affiliation(s)
- E M Sadeesh
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India.
| | - Madhuri S Lahamge
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Anuj Malik
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - A N Ampadi
- Laboratory of Mitochondrial Biology of Farm Animals, Animal Biochemistry Division, ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
9
|
Quon T, Lin LC, Ganguly A, Hudson BD, Tobin AB, Milligan G. Biased constitutive signaling of the G protein-coupled receptor GPR35 suppresses gut barrier permeability. J Biol Chem 2025; 301:108035. [PMID: 39615676 PMCID: PMC11732441 DOI: 10.1016/j.jbc.2024.108035] [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: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Agonist-independent, or constitutive, activity is an integral feature of G protein-coupled receptors, but its relevance in pathophysiological settings is generally poorly explored. GPR35 is a therapeutic target in inflammatory diseases of the lower gut. In colonic organoids from a human GPR35a-expressing transgenic mouse line, the GPR35 inverse agonist CID-2745687 increased barrier permeability substantially, indicating that constitutive receptor activity contributes to maintaining epithelial barrier integrity. High constitutive activity of GPR35 was also observed in both HT-29 and HEPG2 cells that express GPR35 endogenously. Mechanistic investigations in recombinant in vitro systems revealed that the constitutive activity of GPR35a was biased and not equivalent across signaling pathways. Hence, no constitutive interactions of the receptor with arrestin-adaptor proteins or activation of Gαo-containing G protein heterotrimers were detected while, even at low GPR35a expression levels, substantial constitutive activation of heterotrimers containing either Gα12 or Gα13 was observed. Similar biased constitutive activity was observed for the human GPR35b isoform. The extent of constitutive and agonist-mediated activity was dependent on receptor expression level. At high receptor levels, constitutive activation of Gα12 or Gα13 masked any agonist-induced effects while low expression levels with low constitutive activity allowed measurement of agonist-induced responses. These results highlight roles, selectivity, and the extent of constitutive activity of GPR35 in cells and tissues that express this receptor endogenously and highlight the contribution of its constitutive activity to maintaining the colonic epithelial barrier, potentially limiting the development of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Tezz Quon
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Li-Chiung Lin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Amlan Ganguly
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Brian D Hudson
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom.
| |
Collapse
|
10
|
Qian Y, Yan Z, Ye T, Shahin V, Jiang J, Fan C. Decoding the regulatory role of ATP synthase inhibitory factor 1 (ATPIF1) in Wallerian degeneration and peripheral nerve regeneration. EXPLORATION (BEIJING, CHINA) 2024; 4:20230098. [PMID: 39713198 PMCID: PMC11655313 DOI: 10.1002/exp.20230098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/02/2024] [Indexed: 12/24/2024]
Abstract
ATP synthase inhibitory factor 1 (ATPIF1), a key modulator of ATP synthase complex activity, has been implicated in various physiological and pathological processes. While its role is established in conditions such as hypoxia, ischemia-reperfusion injury, apoptosis, and cancer, its involvement remains elusive in peripheral nerve regeneration. Leveraging ATPIF1 knockout transgenic mice, this study reveals that the absence of ATPIF1 impedes neural structural reconstruction, leading to delayed sensory and functional recovery. RNA-sequencing unveils a significant attenuation in immune responses following peripheral nerve injury, which attributes to the CCR2/CCL2 signaling axis and results in decreased macrophage infiltration and activation. Importantly, macrophages, not Schwann cells, are identified as key contributors to the delayed Wallerian degeneration in ATPIF1 knockout mice, and affect the overall outcome of peripheral nerve regeneration. These results shed light on the translational potential of ATPIF1 for improving peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yun Qian
- Department of OrthopedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue RegenerationShanghaiPeople's Republic of China
| | - Zhiwen Yan
- Department of OrthopedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue RegenerationShanghaiPeople's Republic of China
| | - Tianbao Ye
- Department of CardiologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Victor Shahin
- Institute of Physiology IIUniversity of MünsterMünsterGermany
| | - Jia Jiang
- Department of OrthopedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue RegenerationShanghaiPeople's Republic of China
| | - Cunyi Fan
- Department of OrthopedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue RegenerationShanghaiPeople's Republic of China
| |
Collapse
|
11
|
Otkur W, Zhang Y, Li Y, Bao W, Feng T, Wu B, Ma Y, Shi J, Wang L, Pei S, Wang W, Wang J, Zhao Y, Liu Y, Li X, Xia T, Wang F, Chen D, Liang X, Piao HL. Spatial multi-omics characterizes GPR35-relevant lipid metabolism signatures across liver zonation in MASLD. LIFE METABOLISM 2024; 3:loae021. [PMID: 39873004 PMCID: PMC11748505 DOI: 10.1093/lifemeta/loae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 01/30/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a metabolic disease that can progress to metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and cancer. The zonal distribution of biomolecules in the liver is implicated in mediating the disease progression. Recently, G-protein-coupled receptor 35 (GPR35) has been highlighted to play a role in MASLD, but the precise mechanism is not fully understood, particularly, in a liver-zonal manner. Here, we aimed to identify spatially distributed specific genes and metabolites in different liver zonation that are regulated by GPR35 in MASLD, by combining lipid metabolomics, spatial transcriptomics (ST), and spatial metabolomics (SM). We found that GPR35 influenced lipid accumulation, inflammatory and metabolism-related factors in specific regions, notably affecting the anti-inflammation factor ELF4 (E74 like E-twenty six (ETS) transcription factor 4), lipid homeostasis key factor CIDEA (cell death-inducing DNA fragmentation factor alpha (DFFA)-like effector A), and the injury response-related genes SAA1/2/3 (serum amyloid A1/2/3), thereby impacting MASLD progression. Furthermore, SM elucidated specific metabolite distributions across different liver regions, such as C10H11N4O7P (3',5'-cyclic inosine monophosphate (3',5'-IMP)) for the central vein, and this metabolite significantly decreased in the liver zones of GPR35-deficient mice during MASLD progression. Taken together, GPR35 regulates hepatocyte damage repair, controls inflammation, and prevents MASLD progression by influencing phospholipid homeostasis and gene expression in a zonal manner.
Collapse
Affiliation(s)
- Wuxiyar Otkur
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shengyang, Liaoning 110016, China
| | - Yiran Zhang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yirong Li
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjun Bao
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingze Feng
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Wu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Yaolu Ma
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Shi
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Li Wang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- Laboratory of High-Resolution Mass Spectrometry Technologies, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Shaojun Pei
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen Wang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Jixia Wang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Yaopeng Zhao
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Yanfang Liu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Xiuling Li
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Tian Xia
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
| | - Fangjun Wang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinmiao Liang
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hai-long Piao
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
12
|
Guo L. F-ATP synthase inhibitory factor 1 and mitochondria-organelle interactions: New insight and implications. Pharmacol Res 2024; 208:107393. [PMID: 39233058 DOI: 10.1016/j.phrs.2024.107393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/08/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitochondria are metabolic hub, and act as primary sites for reactive oxygen species (ROS) and metabolites generation. Mitochondrial Ca2+ uptake contributes to Ca2+ storage. Mitochondria-organelle interactions are important for cellular metabolic adaptation, biosynthesis, redox balance, cell fate. Organelle communications are mediated by Ca2+/ROS signals, vesicle transport and membrane contact sites. The permeability transition pore (PTP) is an unselective channel that provides a release pathway for Ca2+/ROS, mtDNA and metabolites. F-ATP synthase inhibitory factor 1 (IF1) participates in regulation of PTP opening and is required for the translocation of transcriptional factors c-Myc/PGC1α to mitochondria to stimulate metabolic switch. IF1, a mitochondrial specific protein, has been suggested to regulate other organelles including nucleus, endoplasmic reticulum and lysosomes. IF1 may be able to mediate mitochondria-organelle interactions and cellular physiology through regulation of PTP activity.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
13
|
Gáspár R, Nógrádi-Halmi D, Demján V, Diószegi P, Igaz N, Vincze A, Pipicz M, Kiricsi M, Vécsei L, Csont T. Kynurenic acid protects against ischemia/reperfusion injury by modulating apoptosis in cardiomyocytes. Apoptosis 2024; 29:1483-1498. [PMID: 39153038 PMCID: PMC11416393 DOI: 10.1007/s10495-024-02004-w] [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] [Accepted: 07/09/2024] [Indexed: 08/19/2024]
Abstract
Acute myocardial infarction, often associated with ischemia/reperfusion injury (I/R), is a leading cause of death worldwide. Although the endogenous tryptophan metabolite kynurenic acid (KYNA) has been shown to exert protection against I/R injury, its mechanism of action at the cellular and molecular level is not well understood yet. Therefore, we examined the potential involvement of antiapoptotic mechanisms, as well as N-methyl-D-aspartate (NMDA) receptor modulation in the protective effect of KYNA in cardiac cells exposed to simulated I/R (SI/R). KYNA was shown to attenuate cell death induced by SI/R dose-dependently in H9c2 cells or primary rat cardiomyocytes. Analysis of morphological and molecular markers of apoptosis (i.e., membrane blebbing, apoptotic nuclear morphology, DNA double-strand breaks, activation of caspases) revealed considerably increased apoptotic activity in cardiac cells undergoing SI/R. The investigated apoptotic markers were substantially improved by treatment with the cytoprotective dose of KYNA. Although cardiac cells were shown to express NMDA receptors, another NMDA antagonist structurally different from KYNA was unable to protect against SI/R-induced cell death. Our findings provide evidence that the protective effect of KYNA against SI/R-induced cardiac cell injury involves antiapoptotic mechanisms, that seem to evoke independently of NMDA receptor signaling.
Collapse
Affiliation(s)
- Renáta Gáspár
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Dóra Nógrádi-Halmi
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Virág Demján
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Petra Diószegi
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Vincze
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE-Neuroscience Research Group, Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm Tér 9, 6720, Szeged, Hungary.
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary.
| |
Collapse
|
14
|
Keifi Bajestani A, Alavi MS, Etemad L, Roohbakhsh A. Role of orphan G-protein coupled receptors in tissue ischemia: A comprehensive review. Eur J Pharmacol 2024; 978:176762. [PMID: 38906238 DOI: 10.1016/j.ejphar.2024.176762] [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: 02/23/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/23/2024]
Abstract
Ischemic events lead to many diseases and deaths worldwide. Ischemia/reperfusion (I/R) occurs due to reduced blood circulation in tissues followed by blood reflow. Reoxygenation of ischemic tissues is characterized by oxidative stress, inflammation, energy distress, and endoplasmic reticulum stress. There are still no adequate clinical protocols or pharmacological approaches to address the consequences of I/R damage. G protein-coupled receptors (GPCRs) are important therapeutic targets. They compose a large family of seven transmembrane-spanning proteins that are involved in many biological functions. Orphan GPCRs are a large subgroup of these receptors expressed in different organs. In the present review, we summarized the literature regarding the role of orphan GPCRs in I/R in different organs. We focused on the effect of these receptors on modulating cellular and molecular processes underlying ischemia including apoptosis, inflammation, and autophagy. The study showed that GPR3, GPR4, GPR17, GPR30, GPR31, GPR35, GPR37, GPR39, GPR55, GPR65, GPR68, GPR75, GPR81, and GPR91 are involved in ischemic events, mainly in the brain and heart. These receptors offer new possibilities for treating I/R injuries in the body.
Collapse
Affiliation(s)
- Alireza Keifi Bajestani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Wyant GA, Jiang Q, Singh M, Qayyum S, Levrero C, Maron BA, Kaelin WG. Induction of DEPP1 by HIF Mediates Multiple Hallmarks of Ischemic Cardiomyopathy. Circulation 2024; 150:770-786. [PMID: 38881449 PMCID: PMC11361356 DOI: 10.1161/circulationaha.123.066628] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 05/22/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND HIF (hypoxia inducible factor) regulates many aspects of cardiac function. We and others previously showed that chronic HIF activation in the heart in mouse models phenocopies multiple features of ischemic cardiomyopathy in humans, including mitochondrial loss, lipid accumulation, and systolic cardiac dysfunction. In some settings, HIF also causes the loss of peroxisomes. How, mechanistically, HIF promotes cardiac dysfunction is an open question. METHODS We used mice lacking cardiac pVHL (von Hippel-Lindau protein) to investigate how chronic HIF activation causes multiple features of ischemic cardiomyopathy, such as autophagy induction and lipid accumulation. We performed immunoblot assays, RNA sequencing, mitochondrial and peroxisomal autophagy flux measurements, and live cell imaging on isolated cardiomyocytes. We used CRISPR-Cas9 gene editing in mice to validate a novel mediator of cardiac dysfunction in the setting of chronic HIF activation. RESULTS We identify a previously unknown pathway by which cardiac HIF activation promotes the loss of mitochondria and peroxisomes. We found that DEPP1 (decidual protein induced by progesterone 1) is induced under hypoxia in a HIF-dependent manner and localizes inside mitochondria. DEPP1 is both necessary and sufficient for hypoxia-induced autophagy and triglyceride accumulation in cardiomyocytes ex vivo. DEPP1 loss increases cardiomyocyte survival in the setting of chronic HIF activation ex vivo, and whole-body Depp1 loss decreases cardiac dysfunction in hearts with chronic HIF activation caused by VHL loss in vivo. CONCLUSIONS Our findings identify DEPP1 as a key component in the cardiac remodeling that occurs with chronic ischemia.
Collapse
Affiliation(s)
- Gregory A. Wyant
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Qinqin Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
| | - Madhu Singh
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Shariq Qayyum
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA (G.A.W., M.S., S.Q.)
| | - Clara Levrero
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
| | - Bradley A. Maron
- Department of Cardiovascular Medicine (B.A.M.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.A.W., Q.J., C.L., W.G.K.)
- Department of Medicine (W.G.K.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Chevy Chase, MD (W.G.K.)
| |
Collapse
|
16
|
Stone TW, Williams RO. Tryptophan metabolism as a 'reflex' feature of neuroimmune communication: Sensor and effector functions for the indoleamine-2, 3-dioxygenase kynurenine pathway. J Neurochem 2024; 168:3333-3357. [PMID: 38102897 DOI: 10.1111/jnc.16015] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/16/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Although the central nervous system (CNS) and immune system were regarded as independent entities, it is now clear that immune system cells can influence the CNS, and neuroglial activity influences the immune system. Despite the many clinical implications for this 'neuroimmune interface', its detailed operation at the molecular level remains unclear. This narrative review focuses on the metabolism of tryptophan along the kynurenine pathway, since its products have critical actions in both the nervous and immune systems, placing it in a unique position to influence neuroimmune communication. In particular, since the kynurenine pathway is activated by pro-inflammatory mediators, it is proposed that physical and psychological stressors are the stimuli of an organismal protective reflex, with kynurenine metabolites as the effector arm co-ordinating protective neural and immune system responses. After a brief review of the neuroimmune interface, the general perception of tryptophan metabolism along the kynurenine pathway is expanded to emphasize this environmentally driven perspective. The initial enzymes in the kynurenine pathway include indoleamine-2,3-dioxygenase (IDO1), which is induced by tissue damage, inflammatory mediators or microbial products, and tryptophan-2,3-dioxygenase (TDO), which is induced by stress-induced glucocorticoids. In the immune system, kynurenic acid modulates leucocyte differentiation, inflammatory balance and immune tolerance by activating aryl hydrocarbon receptors and modulates pain via the GPR35 protein. In the CNS, quinolinic acid activates N-methyl-D-aspartate (NMDA)-sensitive glutamate receptors, whereas kynurenic acid is an antagonist: the balance between glutamate, quinolinic acid and kynurenic acid is a significant regulator of CNS function and plasticity. The concept of kynurenine and its metabolites as mediators of a reflex coordinated protection against stress helps to understand the variety and breadth of their activity. It should also help to understand the pathological origin of some psychiatric and neurodegenerative diseases involving the immune system and CNS, facilitating the development of new pharmacological strategies for treatment.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [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: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
18
|
Stone TW, Darlington LG, Badawy AAB, Williams RO. The Complex World of Kynurenic Acid: Reflections on Biological Issues and Therapeutic Strategy. Int J Mol Sci 2024; 25:9040. [PMID: 39201726 PMCID: PMC11354734 DOI: 10.3390/ijms25169040] [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: 07/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
It has been unequivocally established that kynurenic acid has a number of actions in a variety of cells and tissues, raising, in principle, the possibility of targeting its generation, metabolism or sites of action to manipulate those effects to a beneficial therapeutic end. However, many basic aspects of the biology of kynurenic acid remain unclear, potentially leading to some confusion and misinterpretations of data. They include questions of the source, generation, targets, enzyme expression, endogenous concentrations and sites of action. This essay is intended to raise and discuss many of these aspects as a source of reference for more balanced discussion. Those issues are followed by examples of situations in which modulating and correcting kynurenic acid production or activity could bring significant therapeutic benefit, including neurological and psychiatric conditions, inflammatory diseases and cell protection. More information is required to obtain a clear overall view of the pharmacological environment relevant to kynurenic acid, especially with respect to the active concentrations of kynurenine metabolites in vivo and changed levels in disease. The data and ideas presented here should permit a greater confidence in appreciating the sites of action and interaction of kynurenic acid under different local conditions and pathologies, enhancing our understanding of kynurenic acid itself and the many clinical conditions in which manipulating its pharmacology could be of clinical value.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| | - L. Gail Darlington
- Worthing Hospital, University Hospitals Sussex NHS Foundation Trust, Worthing BN11 2DH, UK
| | - Abdulla A.-B. Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK;
| |
Collapse
|
19
|
Zhang T, An W, You S, Chen S, Zhang S. G protein-coupled receptors and traditional Chinese medicine: new thinks for the development of traditional Chinese medicine. Chin Med 2024; 19:92. [PMID: 38956679 PMCID: PMC11218379 DOI: 10.1186/s13020-024-00964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) widely exist in vivo and participate in many physiological processes, thus emerging as important targets for drug development. Approximately 30% of the Food and Drug Administration (FDA)-approved drugs target GPCRs. To date, the 'one disease, one target, one molecule' strategy no longer meets the demands of drug development. Meanwhile, small-molecule drugs account for 60% of FDA-approved drugs. Traditional Chinese medicine (TCM) has garnered widespread attention for its unique theoretical system and treatment methods. TCM involves multiple components, targets and pathways. Centered on GPCRs and TCM, this paper discusses the similarities and differences between TCM and GPCRs from the perspectives of syndrome of TCM, the consistency of TCM's multi-component and multi-target approaches and the potential of GPCRs and TCM in the development of novel drugs. A novel strategy, 'simultaneous screening of drugs and targets', was proposed and applied to the study of GPCRs. We combine GPCRs with TCM to facilitate the modernisation of TCM, provide valuable insights into the rational application of TCM and facilitate the research and development of novel drugs. This study offers theoretical support for the modernisation of TCM and introduces novel ideas for development of safe and effective drugs.
Collapse
Affiliation(s)
- Ting Zhang
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China
| | - Wenqiao An
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China
| | - Shengjie You
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shilin Chen
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sanyin Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China.
| |
Collapse
|
20
|
Fujikawa M, Ueda M, Maruyama K. Role of Kynurenine and Its Derivatives in the Neuroimmune System. Int J Mol Sci 2024; 25:7144. [PMID: 39000249 PMCID: PMC11241229 DOI: 10.3390/ijms25137144] [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: 05/28/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
In recent years, there has been a growing realization of intricate interactions between the nervous and immune systems, characterized by shared humoral factors and receptors. This interplay forms the basis of the neuroimmune system, the understanding of which will provide insights into the pathogenesis of neurological diseases, in which the involvement of the immune system has been overlooked. Kynurenine and its derivatives derived from tryptophan have long been implicated in the pathogenesis of various neurological diseases. Recent studies have revealed their close association not only with neurological disorders but also with sepsis-related deaths. This review provides an overview of the biochemistry of kynurenine and its derivatives, followed by a discussion of their role via the modulation of the neuroimmune system in various diseases.
Collapse
Affiliation(s)
- Makoto Fujikawa
- Department of Pharmacology, School of Medicine, Aichi Medical University, Nagakute 480-1195, Aichi, Japan
| | - Masashi Ueda
- Department of Pharmacology, School of Medicine, Aichi Medical University, Nagakute 480-1195, Aichi, Japan
| | - Kenta Maruyama
- Department of Pharmacology, School of Medicine, Aichi Medical University, Nagakute 480-1195, Aichi, Japan
| |
Collapse
|
21
|
Sathyasaikumar KV, Blanco-Ayala T, Zheng Y, Schwieler L, Erhardt S, Tufvesson-Alm M, Poeggeler B, Schwarcz R. The Tryptophan Metabolite Indole-3-Propionic Acid Raises Kynurenic Acid Levels in the Rat Brain In Vivo. Int J Tryptophan Res 2024; 17:11786469241262876. [PMID: 38911967 PMCID: PMC11191616 DOI: 10.1177/11786469241262876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Alterations in the composition of the gut microbiota may be causally associated with several brain diseases. Indole-3-propionic acid (IPrA) is a tryptophan-derived metabolite, which is produced by intestinal commensal microbes, rapidly enters the circulation, and crosses the blood-brain barrier. IPrA has neuroprotective properties, which have been attributed to its antioxidant and bioenergetic effects. Here, we evaluate an alternative and/or complementary mechanism, linking IPrA to kynurenic acid (KYNA), another neuroprotective tryptophan metabolite. Adult Sprague-Dawley rats received an oral dose of IPrA (200 mg/kg), and both IPrA and KYNA were measured in plasma and frontal cortex 90 minutes, 6 or 24 hours later. IPrA and KYNA levels increased after 90 minutes and 6 hours (brain IPrA: ~56- and ~7-fold; brain KYNA: ~4- and ~3-fold, respectively). In vivo microdialysis, performed in the medial prefrontal cortex and in the striatum, revealed increased KYNA levels (~2.5-fold) following the administration of IPrA (200 mg/kg, p.o), but IPrA failed to affect extracellular KYNA when applied locally. Finally, treatment with 100 or 350 mg IPrA, provided daily to the animals in the chow for a week, resulted in several-fold increases of IPrA and KYNA levels in both plasma and brain. These results suggest that exogenously supplied IPrA may provide a novel strategy to affect the function of KYNA in the mammalian brain.
Collapse
Affiliation(s)
- Korrapati V Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, USA
| | - Tonali Blanco-Ayala
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez,” Mexico City, Mexico
| | - Yiran Zheng
- Departments of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Lilly Schwieler
- Departments of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Sophie Erhardt
- Departments of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | | | - Burkhard Poeggeler
- Department of Physiology, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Georg-August-Universität Göttingen, Germany
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
22
|
Yang Y, Liu X, Liu X, Xie C, Shi J. The role of the kynurenine pathway in cardiovascular disease. Front Cardiovasc Med 2024; 11:1406856. [PMID: 38883986 PMCID: PMC11176437 DOI: 10.3389/fcvm.2024.1406856] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
The kynurenine pathway (KP) serves as the primary route for tryptophan metabolism in most mammalian organisms, with its downstream metabolites actively involved in various physiological and pathological processes. Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) serve as the initial and pivotal enzymes of the KP, with IDO playing important and intricate roles in cardiovascular diseases. Multiple metabolites of KP have been observed to exhibit elevated concentrations in plasma across various cardiovascular diseases, such as atherosclerosis, hypertension, and acute myocardial infarction. Multiple studies have indicated that kynurenine (KYN) may serve as a potential biomarker for several adverse cardiovascular events. Furthermore, Kynurenine and its downstream metabolites have complex roles in inflammation, exhibiting both inhibitory and stimulatory effects on inflammatory responses under different conditions. In atherosclerosis, upregulation of IDO stimulates KYN production, mediating aromatic hydrocarbon receptor (AhR)-induced exacerbation of vascular inflammation and promotion of foam cell formation. Conversely, in arterial calcification, this mediation alleviates osteogenic differentiation of vascular smooth muscle cells. Additionally, in cardiac remodeling, KYN-mediated AhR activation exacerbates pathological left ventricular hypertrophy and fibrosis. Interventions targeting components of the KP, such as IDO inhibitors, 3-hydroxyanthranilic acid, and anthranilic acid, demonstrate cardiovascular protective effects. This review outlines the mechanistic roles of KP in coronary atherosclerosis, arterial calcification, and myocardial diseases, highlighting the potential diagnostic, prognostic, and therapeutic value of KP in cardiovascular diseases, thus providing novel insights for the development and application of related drugs in future research.
Collapse
Affiliation(s)
- Yuehang Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chiyang Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Lhuissier C, Desquiret-Dumas V, Girona A, Alban J, Faure J, Cassereau J, Codron P, Lenaers G, Baris OR, Gueguen N, Chevrollier A. Mitochondrial F0F1-ATP synthase governs the induction of mitochondrial fission. iScience 2024; 27:109808. [PMID: 38741710 PMCID: PMC11089353 DOI: 10.1016/j.isci.2024.109808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/22/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Mitochondrial dynamics is a process that balances fusion and fission events, the latter providing a mechanism for segregating dysfunctional mitochondria. Fission is controlled by the mitochondrial membrane potential (ΔΨm), optic atrophy 1 (OPA1) cleavage, and DRP1 recruitment. It is thought that this process is closely linked to the activity of the mitochondrial respiratory chain (MRC). However, we report here that MRC inhibition does not decrease ΔΨm nor increase fission, as evidenced by hyperconnected mitochondria. Conversely, blocking F0F1-ATP synthase activity induces fragmentation. We show that the F0F1-ATP synthase is sensing the inhibition of MRC activity by immediately promoting its reverse mode of action to hydrolyze matrix ATP and restoring ΔΨm, thus preventing fission. While this reverse mode is expected to be inhibited by the ATPase inhibitor ATPIF1, we show that this sensing is independent of this factor. We have unraveled an unexpected role of F0F1-ATP synthase in controlling the induction of fission by sensing and maintaining ΔΨm.
Collapse
Affiliation(s)
- Charlène Lhuissier
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Valérie Desquiret-Dumas
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
- Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France
| | - Anaïs Girona
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Jennifer Alban
- Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France
| | - Justine Faure
- Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France
| | - Julien Cassereau
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
- Department of Neurology, Angers University Hospital, Angers, France
| | - Philippe Codron
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
- Department of Neurology, Angers University Hospital, Angers, France
| | - Guy Lenaers
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
- Department of Neurology, Angers University Hospital, Angers, France
| | - Olivier R. Baris
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Naïg Gueguen
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
- Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France
| | - Arnaud Chevrollier
- University Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| |
Collapse
|
24
|
Zhang YW, Pang X, Yang Y. Hydrogels containing KYNA promote angiogenesis and inhibit inflammation to improve the survival rate of multi-territory perforator flaps. Biomed Pharmacother 2024; 174:116454. [PMID: 38640710 DOI: 10.1016/j.biopha.2024.116454] [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: 11/07/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND A new spray adhesive (KYNA-PF127) was established through the combination of thermosensitive hydrogel (Pluronic F127) and KYNA, aimed to investigate the effect of KYNA-PF127 on multi-territory perforator flaps and its possible molecular mechanism. MATERIALS AND METHODS 36 SD male rats with 250-300 g were randomly divided into 3 groups (n = 12): control group, blank glue group and KYNA-PF127 group. KYNA-PF127 hydrogel was prepared and characterized for its morphology and properties using scanning electron microscopy. CCK-8 assay, scratch wound assay, transwell assay, tube formation assay and Ki67 staining were used to study the effect of KYNA-PF127 on the proliferation, migration, and tube formation of HUVECs. VEGF and FGF2 were measured by qPCR to evaluate the angiogenesis capacity of HUVECs in vitro. In vivo, the effect of each group on the survival area of the cross-zone perforator flap was evaluated, and angiogenesis was evaluated by HE and immunofluorescence (CD31 and MMP-9). The effect of inflammation on skin collagen fibers was assessed by Masson. Immunohistochemistry (SOD1, IL-1β, TNF-α) was used to evaluate the effects of oxidative stress and inflammatory factors on multi-territory flaps. RESULTS KYNA-PF127 has good sustained release and biocompatibility at 25% concentration. KYNA-PF127 promoted the proliferation, migration, and angiogenesis of HUVECs in vitro. In vivo, the survival area of multi-territory perforator flaps and angiogenic capability have increased after KYNA-PF127 intervention. KYNA-PF127 could effectively reduce the oxidative stress and inflammation of multi-territory perforator flaps. CONCLUSION KYNA-PF127 promotes angiogenesis through its antioxidant stress and anti-inflammatory effects, and shows potential clinical value in promoting the survival viability and drug delivery of multi-territory perforator flaps.
Collapse
Affiliation(s)
- Ya-Wei Zhang
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiaoyang Pang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yan Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.
| |
Collapse
|
25
|
Donlon J, Kumari P, Varghese SP, Bai M, Florentin OD, Frost ED, Banks J, Vadlapatla N, Kam O, Shad MU, Rahman S, Abulseoud OA, Stone TW, Koola MM. Integrative Pharmacology in the Treatment of Substance Use Disorders. J Dual Diagn 2024; 20:132-177. [PMID: 38117676 DOI: 10.1080/15504263.2023.2293854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The detrimental physical, mental, and socioeconomic effects of substance use disorders (SUDs) have been apparent to the medical community for decades. However, it has become increasingly urgent in recent years to develop novel pharmacotherapies to treat SUDs. Currently, practitioners typically rely on monotherapy. Monotherapy has been shown to be superior to no treatment at all for most substance classes. However, many randomized controlled trials (RCTs) have revealed that monotherapy leads to poorer outcomes when compared with combination treatment in all specialties of medicine. The results of RCTs suggest that monotherapy frequently fails since multiple dysregulated pathways, enzymes, neurotransmitters, and receptors are involved in the pathophysiology of SUDs. As such, research is urgently needed to determine how various neurobiological mechanisms can be targeted by novel combination treatments to create increasingly specific yet exceedingly comprehensive approaches to SUD treatment. This article aims to review the neurobiology that integrates many pathophysiologic mechanisms and discuss integrative pharmacology developments that may ultimately improve clinical outcomes for patients with SUDs. Many neurobiological mechanisms are known to be involved in SUDs including dopaminergic, nicotinic, N-methyl-D-aspartate (NMDA), and kynurenic acid (KYNA) mechanisms. Emerging evidence indicates that KYNA, a tryptophan metabolite, modulates all these major pathophysiologic mechanisms. Therefore, achieving KYNA homeostasis by harmonizing integrative pathophysiology and pharmacology could prove to be a better therapeutic approach for SUDs. We propose KYNA-NMDA-α7nAChRcentric pathophysiology, the "conductor of the orchestra," as a novel approach to treat many SUDs concurrently. KYNA-NMDA-α7nAChR pathophysiology may be the "command center" of neuropsychiatry. To date, extant RCTs have shown equivocal findings across comparison conditions, possibly because investigators targeted single pathophysiologic mechanisms, hit wrong targets in underlying pathophysiologic mechanisms, and tested inadequate monotherapy treatment. We provide examples of potential combination treatments that simultaneously target multiple pathophysiologic mechanisms in addition to KYNA. Kynurenine pathway metabolism demonstrates the greatest potential as a target for neuropsychiatric diseases. The investigational medications with the most evidence include memantine, galantamine, and N-acetylcysteine. Future RCTs are warranted with novel combination treatments for SUDs. Multicenter RCTs with integrative pharmacology offer a promising, potentially fruitful avenue to develop novel therapeutics for the treatment of SUDs.
Collapse
Affiliation(s)
- Jack Donlon
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Pooja Kumari
- Community Living Trent Highlands, Peterborough, Canada
| | - Sajoy P Varghese
- Addiction Recovery Treatment Services, Veterans Affairs Northern California Health Care System, University of California, Davis, Sacramento, California, USA
| | - Michael Bai
- Columbia University, New York, New York, USA
| | - Ori David Florentin
- Department of Psychiatry, Westchester Medical Center, Valhalla, New York, USA
| | - Emma D Frost
- Department of Neurology, Cooper University Health Care, Camden, New Jersey, USA
| | - John Banks
- Talkiatry Mental Health Clinic, New York, New York, USA
| | - Niyathi Vadlapatla
- Thomas Jefferson High School for Science and Technology, Alexandria, Virginia, USA
| | - Olivia Kam
- Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| | - Mujeeb U Shad
- Department of Psychiatry, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Alix School of Medicine at Mayo Clinic, Phoenix, Arizona, USA
| | - Trevor W Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Cooper University Health Care, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| |
Collapse
|
26
|
Xu Y, Deng C, Chen H, Song Y, Xu H, Song G, Wang X, Luo T, Chen W, Ma J, Zeng A, Huang S, Chen Z, Fu J, Gong M, Tai Y, Huang A, Feng H, Hu J, Zhu X, Tang Q, Lu J, Wang J. Osteosarcoma Cells Secrete CXCL14 That Activates Integrin α11β1 on Fibroblasts to Form a Lung Metastatic Niche. Cancer Res 2024; 84:994-1012. [PMID: 38295227 DOI: 10.1158/0008-5472.can-23-1307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/02/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024]
Abstract
Cooperation between primary malignant cells and stromal cells can mediate the establishment of lung metastatic niches. Here, we characterized the landscape of cell populations in the tumor microenvironment in treatment-naïve osteosarcoma using single-cell RNA sequencing and identified a stem cell-like cluster with tumor cell-initiating properties and prometastatic traits. CXCL14 was specifically enriched in the stem cell-like cluster and was also significantly upregulated in lung metastases compared with primary tumors. CXCL14 induced stromal reprogramming and evoked a malignant phenotype in fibroblasts to form a supportive lung metastatic niche. Binding of CXCL14 to heterodimeric integrin α11β1 on fibroblasts activated actomyosin contractility and matrix remodeling properties. CXCL14-stimulated fibroblasts produced TGFβ and increased osteosarcoma invasion and migration. mAbs targeting the CXCL14-integrin α11β1 axis inhibited fibroblast TGFβ production, enhanced CD8+ T cell-mediated antitumor immunity, and suppressed osteosarcoma lung metastasis. Taken together, these findings identify cross-talk between osteosarcoma cells and fibroblasts that promotes metastasis and demonstrate that targeting the CXCL14-integrin α11β1 axis is a potential strategy to inhibit osteosarcoma lung metastasis. SIGNIFICANCE Cooperation between stem-like osteosarcoma cells and fibroblasts mediated by a CXCL14-integrin α11β1 axis creates a tumor-supportive lung metastatic niche and represents a therapeutic target to suppress osteosarcoma metastasis.
Collapse
Affiliation(s)
- Yanyang Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Chuangzhong Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Hongmin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - YiJiang Song
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Huaiyuan Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Guohui Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xinliang Wang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Tianqi Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Weiqing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jiahui Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Anyu Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shujing Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Zhihao Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jianchang Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Ming Gong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi Tai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Anfei Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Huixiong Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jinxin Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xiaojun Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Qinglian Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jinchang Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| |
Collapse
|
27
|
Lauterboeck L, Kang SW, White D, Bao R, Mobasheran P, Yang Q. IF1 Promotes Cellular Proliferation and Inhibits Oxidative Phosphorylation in Mouse Embryonic Fibroblasts under Normoxia and Hypoxia. Cells 2024; 13:551. [PMID: 38534395 PMCID: PMC10969582 DOI: 10.3390/cells13060551] [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: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
ATP synthase inhibitory factor subunit 1 (IF1) is an inhibitory subunit of mitochondrial ATP synthase, playing a crucial role in regulating mitochondrial respiration and energetics. It is well-established that IF1 interacts with the F1 sector of ATP synthase to inhibit the reversal rotation and, thus, ATP hydrolysis. Recent evidence supports that IF1 also inhibits forward rotation or the ATP synthesis activity. Adding to the complexity, IF1 may also facilitate mitophagy and cristae formation. The implications of these complex actions of IF1 for cellular function remain obscure. In the present study, we found that IF1 expression was markedly upregulated in hypoxic MEFs relative to normoxic MEFs. We investigate how IF1 affects cellular growth and function in cultured mouse embryonic fibroblasts derived from mouse lines with systemic IF1 overexpression and knockout under normoxia and hypoxia. Cell survival and proliferation analyses revealed that IF1 overexpression exerted limited effects on cellular viability but substantially increased proliferation under normoxia, whereas it facilitated both cellular viability and proliferation under hypoxia. The absence of IF1 may have a pro-survival effect but not a proliferative one in both normoxia and hypoxia. Cellular bioenergetic analyses revealed that IF1 suppressed cellular respiration when subjected to normoxia and was even more pronounced when subjected to hypoxia with increased mitochondrial ATP production. In contrast, IF1 knockout MEFs showed markedly increased cellular respiration under both normoxia and hypoxia with little change in mitochondrial ATP. Glycolytic stress assay revealed that IF1 overexpression modestly increased glycolysis in normoxia and hypoxia. Interestingly, the absence of IF1 in MEFs led to substantial increases in glycolysis. Therefore, we conclude that IF1 mainly inhibits cellular respiration and enhances cellular glycolysis to preserve mitochondrial ATP. On the other hand, IF1 deletion can significantly facilitate cellular respiration and glycolysis without leading to mitochondrial ATP deficit.
Collapse
Affiliation(s)
- Lothar Lauterboeck
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Cell Biology, Life Science Solutions, Thermo Fisher Scientific, Frederick, MD 21704, USA
| | - Sung Wook Kang
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
| | - Donnell White
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Rong Bao
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Parnia Mobasheran
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Qinglin Yang
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
28
|
Oka M, Akaki S, Ohno O, Terasaki M, Hamaoka-Tamura Y, Saito M, Kato S, Inoue A, Aoki J, Matsuno K, Furuta K, Tanaka S. Suppression of Mast Cell Activation by GPR35: GPR35 Is a Primary Target of Disodium Cromoglycate. J Pharmacol Exp Ther 2024; 389:76-86. [PMID: 38290974 DOI: 10.1124/jpet.123.002024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
Mast cell stabilizers, including disodium cromoglycate (DSCG), were found to have potential as the agonists of an orphan G protein-coupled receptor, GPR35, although it remains to be determined whether GPR35 is expressed in mast cells and involved in suppression of mast cell degranulation. Our purpose in this study is to verify the expression of GPR35 in mast cells and to clarify how GPR35 modulates the degranulation. We explored the roles of GPR35 using an expression system, a mast cell line constitutively expressing rat GPR35, peritoneal mast cells, and bone marrow-derived cultured mast cells. Immediate allergic responses were assessed using the IgE-mediated passive cutaneous anaphylaxis (PCA) model. Various known GPR35 agonists, including DSCG and newly designed compounds, suppressed IgE-mediated degranulation. GPR35 was expressed in mature mast cells but not in immature bone marrow-derived cultured mast cells and the rat mast cell line. Degranulation induced by antigens was significantly downmodulated in the mast cell line stably expressing GPR35. A GPR35 agonist, zaprinast, induced a transient activation of RhoA and a transient decrease in the amount of filamentous actin. GPR35 agonists suppressed the PCA responses in the wild-type mice but not in the GPR35-/- mice. These findings suggest that GPR35 should prevent mast cells from undergoing degranulation induced by IgE-mediated antigen stimulation and be the primary target of mast cell stabilizers. SIGNIFICANCE STATEMENT: The agonists of an orphan G protein-coupled receptor, GPR35, including disodium cromoglycate, were found to suppress degranulation of rat and mouse mature mast cells, and their antiallergic effects were abrogated in the GPR35-/- mice, indicating that the primary target of mast cell stabilizers should be GPR35.
Collapse
Affiliation(s)
- Masumi Oka
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Sohta Akaki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Osamu Ohno
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Maho Terasaki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Yuho Hamaoka-Tamura
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Michiko Saito
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Shinichi Kato
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Asuka Inoue
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Junken Aoki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Kenji Matsuno
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Kazuyuki Furuta
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Satoshi Tanaka
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| |
Collapse
|
29
|
Cuezva JM, Domínguez-Zorita S. The ATPase Inhibitory Factor 1 (IF1) Contributes to the Warburg Effect and Is Regulated by Its Phosphorylation in S39 by a Protein Kinase A-like Activity. Cancers (Basel) 2024; 16:1014. [PMID: 38473373 DOI: 10.3390/cancers16051014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
The relevant role played by the ATPase Inhibitory Factor 1 (IF1) as a physiological in vivo inhibitor of mitochondrial ATP synthase in cancer and non-cancer cells, and in the mitochondria of different mouse tissues, as assessed in different genetic loss- and gain-of-function models of IF1 has been extensively documented. In this review we summarize our findings and those of others that favor the implication of IF1 in metabolic reprogramming to an enhanced glycolytic phenotype, which is mediated by its binding and inhibition of the ATP synthase. Moreover, we emphasize that IF1 is phosphorylated in vivo in its S39 by the c-AMP-dependent PKA activity of mitochondria to render an inactive inhibitor that is unable to interact with the enzyme, thus triggering the activation of ATP synthase. Overall, we discuss and challenge the results that argue against the role of IF1 as in vivo inhibitor of mitochondrial ATP synthase and stress that IF1 cannot be regarded solely as a pro-oncogenic protein because in some prevalent carcinomas, it prevents metastatic disease.
Collapse
Affiliation(s)
- José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
30
|
Yang X, Zhang W, Wang L, Zhao Y, Wei W. Metabolite-sensing GPCRs in rheumatoid arthritis. Trends Pharmacol Sci 2024; 45:118-133. [PMID: 38182481 DOI: 10.1016/j.tips.2023.12.001] [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: 11/13/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024]
Abstract
Persistent inflammation in damaged joints results in metabolic dysregulation of the synovial microenvironment, causing pathogenic alteration of cell activity in rheumatoid arthritis (RA). Recently, the role of metabolite and metabolite-sensing G protein-coupled receptors (GPCRs) in the RA-related inflammatory immune response (IIR) has become a focus of research attention. These GPCRs participate in the progression of RA by modulating immune cell activation, migration, and inflammatory responses. Here, we discuss recent evidence implicating metabolic dysregulation in RA pathogenesis, focusing on the connection between RA-related IIR and GPCR signals originating from the synovial joint and gut. Furthermore, we discuss future directions for targeting metabolite-sensing GPCRs for therapeutic benefit, emphasizing the importance of identifying endogenous ligands and investigating the various transduction mechanisms involved.
Collapse
Affiliation(s)
- Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
31
|
Dixit D, Hallisey VM, Zhu EY, Okuniewska M, Cadwell K, Chipuk JE, Axelrad JE, Schwab SR. S1PR1 inhibition induces proapoptotic signaling in T cells and limits humoral responses within lymph nodes. J Clin Invest 2024; 134:e174984. [PMID: 38194271 PMCID: PMC10869180 DOI: 10.1172/jci174984] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
Effective immunity requires a large, diverse naive T cell repertoire circulating among lymphoid organs in search of antigen. Sphingosine 1-phosphate (S1P) and its receptor S1PR1 contribute by both directing T cell migration and supporting T cell survival. Here, we addressed how S1P enables T cell survival and the implications for patients treated with S1PR1 antagonists. We found that S1PR1 limited apoptosis by maintaining the appropriate balance of BCL2 family members via restraint of JNK activity. Interestingly, the same residues of S1PR1 that enable receptor internalization were required to prevent this proapoptotic cascade. Findings in mice were recapitulated in ulcerative colitis patients treated with the S1PR1 antagonist ozanimod, and the loss of naive T cells limited B cell responses. Our findings highlighted an effect of S1PR1 antagonists on the ability to mount immune responses within lymph nodes, beyond their effect on lymph node egress, and suggested both limitations and additional uses of this important class of drugs.
Collapse
Affiliation(s)
- Dhaval Dixit
- Departments of Cell Biology and Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victoria M. Hallisey
- Departments of Cell Biology and Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ethan Y.S. Zhu
- Departments of Cell Biology and Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Martyna Okuniewska
- Departments of Cell Biology and Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ken Cadwell
- Department of Medicine and Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jerry E. Chipuk
- Department of Oncological Sciences, Department of Dermatology, and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordan E. Axelrad
- Division of Gastroenterology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Susan R. Schwab
- Departments of Cell Biology and Pathology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
32
|
Rao B, Yu X, Bai J, Hu J. E2EATP: Fast and High-Accuracy Protein-ATP Binding Residue Prediction via Protein Language Model Embedding. J Chem Inf Model 2024; 64:289-300. [PMID: 38127815 DOI: 10.1021/acs.jcim.3c01298] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Identifying the ATP-binding sites of proteins is fundamentally important to uncover the mechanisms of protein functions and explore drug discovery. Many computational methods are proposed to predict ATP-binding sites. However, due to the limitation of the quality of feature representation, the prediction performance still has a big room for improvement. In this study, we propose an end-to-end deep learning model, E2EATP, to dig out more discriminative information from a protein sequence for improving the ATP-binding site prediction performance. Concretely, we employ a pretrained deep learning-based protein language model (ESM2) to automatically extract high-latent discriminative representations of protein sequences relevant for protein functions. Based on ESM2, we design a residual convolutional neural network to train a protein-ATP binding site prediction model. Furthermore, a weighted focal loss function is used to reduce the negative impact of imbalanced data on the model training stage. Experimental results on the two independent testing data sets demonstrate that E2EATP could achieve higher Matthew's correlation coefficient and AUC values than most existing state-of-the-art prediction methods. The speed (about 0.05 s per protein) of E2EATP is much faster than the other existing prediction methods. Detailed data analyses show that the major advantage of E2EATP lies at the utilization of the pretrained protein language model that extracts more discriminative information from the protein sequence only. The standalone package of E2EATP is freely available for academic at https://github.com/jun-csbio/e2eatp/.
Collapse
Affiliation(s)
- Bing Rao
- School of Information and Electrical Engineering, Hangzhou City University, Hangzhou 310015, China
| | - Xuan Yu
- Glasgow College, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jie Bai
- School of Information and Electrical Engineering, Hangzhou City University, Hangzhou 310015, China
| | - Jun Hu
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| |
Collapse
|
33
|
Pearson K, Beier K, Mardis T, Munoz B, Zaidi A. The Neurochemistry of Depression: The Good, The Bad and The Ugly. MISSOURI MEDICINE 2024; 121:68-75. [PMID: 38404431 PMCID: PMC10887465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A large constellation of experimental evidence suggests that neuroinflammation is involved in the onset of depression and neurodegenerative disorders. Many studies have shown impairments in tryptophan metabolism, the major pathway for the synthesis of serotonin, the mood regulating neurotransmitter. This article reviews the various metabolites generated in the competing pathways of tryptophan metabolism including the kynurenine pathway. Increased synthesis of the neurotoxic compound quinolinic acid occurs at the expense of the synthesis of the neuroprotective metabolite kynurenic acid. This shift in equilibrium plays a critical role in the induction of oxidative stress, neuroinflammation, and neurotoxicity. Sufficient protein intake with adequate amounts of tryptophan along with dietary antioxidants and flavonoids may offer protection against major depressive and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kami Pearson
- Research Associate, Kansas City University, Kansas City, Missouri
| | - Kimberley Beier
- Medical student, Kansas City University, Kansas City, Missouri
| | - Thornton Mardis
- Medical student, Kansas City University, Kansas City, Missouri
| | - Bryan Munoz
- Medical student, Kansas City University, Kansas City, Missouri
| | - Asma Zaidi
- Professor of Biochemistry, Kansas City University, Kansas City, Missouri
| |
Collapse
|
34
|
Lemieux GA, Yoo S, Lin L, Vohra M, Ashrafi K. The steroid hormone ADIOL promotes learning by reducing neural kynurenic acid levels. Genes Dev 2023; 37:998-1016. [PMID: 38092521 PMCID: PMC10760639 DOI: 10.1101/gad.350745.123] [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: 04/24/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023]
Abstract
Reductions in brain kynurenic acid levels, a neuroinhibitory metabolite, improve cognitive function in diverse organisms. Thus, modulation of kynurenic acid levels is thought to have therapeutic potential in a range of brain disorders. Here we report that the steroid 5-androstene 3β, 17β-diol (ADIOL) reduces kynurenic acid levels and promotes associative learning in Caenorhabditis elegans We identify the molecular mechanisms through which ADIOL links peripheral metabolic pathways to neural mechanisms of learning capacity. Moreover, we show that in aged animals, which normally experience rapid cognitive decline, ADIOL improves learning capacity. The molecular mechanisms that underlie the biosynthesis of ADIOL as well as those through which it promotes kynurenic acid reduction are conserved in mammals. Thus, rather than a minor intermediate in the production of sex steroids, ADIOL is an endogenous hormone that potently regulates learning capacity by causing reductions in neural kynurenic acid levels.
Collapse
Affiliation(s)
- George A Lemieux
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Shinja Yoo
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lin Lin
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Mihir Vohra
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
35
|
Wu Y, Zhang P, Fan H, Zhang C, Yu P, Liang X, Chen Y. GPR35 acts a dual role and therapeutic target in inflammation. Front Immunol 2023; 14:1254446. [PMID: 38035084 PMCID: PMC10687457 DOI: 10.3389/fimmu.2023.1254446] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
GPR35 is a G protein-coupled receptor with notable involvement in modulating inflammatory responses. Although the precise role of GPR35 in inflammation is not yet fully understood, studies have suggested that it may have both pro- and anti-inflammatory effects depending on the specific cellular environment. Some studies have shown that GPR35 activation can stimulate the production of pro-inflammatory cytokines and facilitate the movement of immune cells towards inflammatory tissues or infected areas. Conversely, other investigations have suggested that GPR35 may possess anti-inflammatory properties in the gastrointestinal tract, liver and certain other tissues by curbing the generation of inflammatory mediators and endorsing the differentiation of regulatory T cells. The intricate role of GPR35 in inflammation underscores the requirement for more in-depth research to thoroughly comprehend its functional mechanisms and its potential significance as a therapeutic target for inflammatory diseases. The purpose of this review is to concurrently investigate the pro-inflammatory and anti-inflammatory roles of GPR35, thus illuminating both facets of this complex issue.
Collapse
Affiliation(s)
- Yetian Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Pei Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Hongjie Fan
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Caiying Zhang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Pengfei Yu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yang Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| |
Collapse
|
36
|
Kamel R, Baetz D, Gueguen N, Lebeau L, Barbelivien A, Guihot AL, Allawa L, Gallet J, Beaumont J, Ovize M, Henrion D, Reynier P, Mirebeau-Prunier D, Prunier F, Tamareille S. Kynurenic Acid: A Novel Player in Cardioprotection against Myocardial Ischemia/Reperfusion Injuries. Pharmaceuticals (Basel) 2023; 16:1381. [PMID: 37895852 PMCID: PMC10610491 DOI: 10.3390/ph16101381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Myocardial infarction is one of the leading causes of mortality worldwide; hence, there is an urgent need to discover novel cardioprotective strategies. Kynurenic acid (KYNA), a metabolite of the kynurenine pathway, has been previously reported to have cardioprotective effects. However, the mechanisms by which KYNA may be protective are still unclear. The current study addressed this issue by investigating KYNA's cardioprotective effect in the context of myocardial ischemia/reperfusion. METHODS H9C2 cells and rats were exposed to hypoxia/reoxygenation or myocardial infarction, respectively, in the presence or absence of KYNA. In vitro, cell death was quantified using flow cytometry analysis of propidium iodide staining. In vivo, TTC-Evans Blue staining was performed to evaluate infarct size. Mitochondrial respiratory chain complex activities were measured using spectrophotometry. Protein expression was evaluated by Western blot, and mRNA levels by RT-qPCR. RESULTS KYNA treatment significantly reduced H9C2-relative cell death as well as infarct size. KYNA did not exhibit any effect on the mitochondrial respiratory chain complex activity. SOD2 mRNA levels were increased by KYNA. A decrease in p62 protein levels together with a trend of increase in PARK2 may mark a stimulation of mitophagy. Additionally, ERK1/2, Akt, and FOXO3α phosphorylation levels were significantly reduced after the KYNA treatment. Altogether, KYNA significantly reduced myocardial ischemia/reperfusion injuries in both in vitro and in vivo models. CONCLUSION Here we show that KYNA-mediated cardioprotection was associated with enhanced mitophagy and antioxidant defense. A deeper understanding of KYNA's cardioprotective mechanisms is necessary to identify promising novel therapeutic targets and their translation into the clinical arena.
Collapse
Affiliation(s)
- Rima Kamel
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Delphine Baetz
- Laboratoire CarMeN, INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, F-69500 Bron, France; (D.B.); (M.O.)
| | - Naïg Gueguen
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
- Service de Biochimie et Biologie Moléculaire, CHU Angers, F-49000 Angers, France
| | - Lucie Lebeau
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Agnès Barbelivien
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Anne-Laure Guihot
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Louwana Allawa
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Jean Gallet
- Service de Cardiologie, CHU Angers, F-49000 Angers, France;
| | - Justine Beaumont
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Michel Ovize
- Laboratoire CarMeN, INSERM U1060, INRA U1397, Université Claude Bernard Lyon 1, F-69500 Bron, France; (D.B.); (M.O.)
- Service d’Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, F-69000 Lyon, France
| | - Daniel Henrion
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| | - Pascal Reynier
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
- Service de Biochimie et Biologie Moléculaire, CHU Angers, F-49000 Angers, France
| | - Delphine Mirebeau-Prunier
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
- Service de Biochimie et Biologie Moléculaire, CHU Angers, F-49000 Angers, France
| | - Fabrice Prunier
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
- Service de Cardiologie, CHU Angers, F-49000 Angers, France;
| | - Sophie Tamareille
- MITOVASC, SFR ICAT, CNRS 6015, INSERM U1083, Université d’Angers, F-49000 Angers, France; (R.K.); (N.G.); (L.L.); (A.-L.G.); (L.A.); (D.H.); (P.R.); (D.M.-P.); (F.P.)
| |
Collapse
|
37
|
Pellicano C, Vaiarello V, Colalillo A, Gigante A, Iannazzo F, Rosato E. Role of kinurenic acid in the systemic sclerosis renal involvement. Clin Exp Med 2023; 23:1713-1719. [PMID: 36436115 DOI: 10.1007/s10238-022-00962-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/22/2022] [Indexed: 11/28/2022]
Abstract
Systemic sclerosis (SSc) subclinical renal vasculopathy is characterized by progressive increase of intrarenal stiffness and reduction of parenchymal thickness due to post ischemic fibrosis secondary to the renal Raynaud phenomenon. Aims of this study were to evaluate kinurenic acid (KYNA) serum level in SSc patients and healthy controls (HC) and to assess the role of KYNA in SSc subclinical nephropathy. Serum level of KYNA was evaluated in 52 SSc patients and 20 HC, matched for sex and age. Renal function was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation to estimate glomerular filtration rate (eGFR) and renal doppler ultrasound was performed to evaluate kidneys' morphology and indices of intrarenal stiffness. The parameters registered were renal longitudinal length, atrophy index (AI), renal sinus, parenchymal thickness, renal resistive index (RRI), pulsatile index (PI) and systolic/diastolic ratio (S/D). SSc patients had lower median value of KYNA than HC [54.43 ng/ml (IQR 44.44-63.64) vs 61.94 ng/ml (IQR 55.23-88.75), p < 0.001]. SSc patients with AI ≥ 0.70 had lower KYNA than SSc patients with AI < 0.70 [47.85 ng/ml (IQR 41.16-59.91) vs 55.5 ng/ml (IQR 49.99-67.33), p < 0.05] and a slightly significant negative linear correlation was found between KYNA and AI (r = - 0.249, p < 0.05). SSc patient with RRI ≥ 0.70 had higher KYNA than SSc patients with RRI < 0.70 [58.25 ng/ml (IQR 50.49-69.68) vs 50.07 ng/ml (IQR 42.70-56.31), p < 0.05] and a significant positive correlation was found between KYNA and RRI (r = 0.318, p < 0.05). KYNA may be used as a marker to evaluate the renal involvement in SSc patients.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy
| | - Valentina Vaiarello
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy
| | - Amalia Colalillo
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy
| | - Francesco Iannazzo
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University, Viale Dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
38
|
Prag HA, Murphy MP, Krieg T. Preventing mitochondrial reverse electron transport as a strategy for cardioprotection. Basic Res Cardiol 2023; 118:34. [PMID: 37639068 PMCID: PMC10462584 DOI: 10.1007/s00395-023-01002-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
In the context of myocardial infarction, the burst of superoxide generated by reverse electron transport (RET) at complex I in mitochondria is a crucial trigger for damage during ischaemia/reperfusion (I/R) injury. Here we outline the necessary conditions for superoxide production by RET at complex I and how it can occur during reperfusion. In addition, we explore various pathways that are implicated in generating the conditions for RET to occur and suggest potential therapeutic strategies to target RET, aiming to achieve cardioprotection.
Collapse
Affiliation(s)
- Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
39
|
Bigelman E, Pasmanik-Chor M, Dassa B, Itkin M, Malitsky S, Dorot O, Pichinuk E, Kleinberg Y, Keren G, Entin-Meer M. Kynurenic acid, a key L-tryptophan-derived metabolite, protects the heart from an ischemic damage. PLoS One 2023; 18:e0275550. [PMID: 37616231 PMCID: PMC10449225 DOI: 10.1371/journal.pone.0275550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 06/20/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Renal injury induces major changes in plasma and cardiac metabolites. Using a small- animal in vivo model, we sought to identify a key metabolite whose levels are significantly modified following an acute kidney injury (AKI) and to analyze whether this agent could offer cardiac protection once an ischemic event has occurred. METHODS AND RESULTS Metabolomics profiling of cardiac lysates and plasma samples derived from rats that underwent AKI 1 or 7 days earlier by 5/6 nephrectomy versus sham-operated controls was performed. We detected 26 differential metabolites in both heart and plasma samples at the two selected time points, relative to sham. Out of which, kynurenic acid (kynurenate, KYNA) seemed most relevant. Interestingly, KYNA given at 10 mM concentration significantly rescued the viability of H9C2 cardiac myoblast cells grown under anoxic conditions and largely increased their mitochondrial content and activity as determined by flow cytometry and cell staining with MitoTracker dyes. Moreover, KYNA diluted in the drinking water of animals induced with an acute myocardial infarction, highly enhanced their cardiac recovery according to echocardiography and histopathology. CONCLUSION KYNA may represent a key metabolite absorbed by the heart following AKI as part of a compensatory mechanism aiming at preserving the cardiac function. KYNA preserves the in vitro myocyte viability following exposure to anoxia in a mechanism that is mediated, at least in part, by protection of the cardiac mitochondria. A short-term administration of KYNA may be highly beneficial in the treatment of the acute phase of kidney disease in order to attenuate progression to reno-cardiac syndrom and to reduce the ischemic myocardial damage following an ischemic event.
Collapse
Affiliation(s)
- Einat Bigelman
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Cardiology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Bareket Dassa
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Maxim Itkin
- Metabolic Profiling Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Malitsky
- Metabolic Profiling Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Orly Dorot
- Bio-Imaging Core, Blavatnik Center for Drug Discovery, Tel-Aviv University, Tel-Aviv, Israel
| | - Edward Pichinuk
- Bio-Imaging Core, Blavatnik Center for Drug Discovery, Tel-Aviv University, Tel-Aviv, Israel
| | - Yuval Kleinberg
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Bio-Imaging Core, Blavatnik Center for Drug Discovery, Tel-Aviv University, Tel-Aviv, Israel
| | - Gad Keren
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Cardiology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michal Entin-Meer
- Laboratory of Cardiovascular Research, Tel Aviv Sourasky Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Cardiology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
40
|
Dixit D, Hallisey VM, Zhu EYS, Okuniewska M, Cadwell K, Chipuk JE, Axelrad JE, Schwab SR. Sphingosine 1-phosphate receptor 1 inhibition induces a pro-apoptotic signaling cascade in T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554104. [PMID: 37662380 PMCID: PMC10473648 DOI: 10.1101/2023.08.21.554104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Effective immunity requires a large, diverse naïve T cell repertoire circulating among lymphoid organs in search of antigen. Sphingosine 1-phosphate (S1P) and its receptor S1PR1 contribute by both directing T cell migration and supporting T cell survival. Here, we address how S1P enables T cell survival, and the implications for patients treated with S1PR1 antagonists. Contrary to expectations, we found that S1PR1 limits apoptosis by maintaining the appropriate balance of BCL2 family members via restraint of JNK activity. Interestingly, the same residues of S1PR1 that enable receptor internalization are required to prevent this pro-apoptotic cascade. Findings in mice were recapitulated in ulcerative colitis patients treated with the S1PR1 antagonist ozanimod, and the loss of naïve T cells limited B cell responses. Our findings highlight an unexpected effect of S1PR1 antagonists on the ability to mount immune responses within lymph nodes, beyond their effect on lymph node egress, and suggest both limitations and novel uses of this important class of drugs.
Collapse
|
41
|
Romero-Carramiñana I, Esparza-Moltó PB, Domínguez-Zorita S, Nuevo-Tapioles C, Cuezva JM. IF1 promotes oligomeric assemblies of sluggish ATP synthase and outlines the heterogeneity of the mitochondrial membrane potential. Commun Biol 2023; 6:836. [PMID: 37573449 PMCID: PMC10423274 DOI: 10.1038/s42003-023-05214-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/04/2023] [Indexed: 08/14/2023] Open
Abstract
The coexistence of two pools of ATP synthase in mitochondria has been largely neglected despite in vitro indications for the existence of reversible active/inactive state transitions in the F1-domain of the enzyme. Herein, using cells and mitochondria from mouse tissues, we demonstrate the existence in vivo of two pools of ATP synthase: one active, the other IF1-bound inactive. IF1 is required for oligomerization and inactivation of ATP synthase and for proper cristae formation. Immunoelectron microscopy shows the co-distribution of IF1 and ATP synthase, placing the inactive "sluggish" ATP synthase preferentially at cristae tips. The intramitochondrial distribution of IF1 correlates with cristae microdomains of high membrane potential, partially explaining its heterogeneous distribution. These findings support that IF1 is the in vivo regulator of the active/inactive state transitions of the ATP synthase and suggest that local regulation of IF1-ATP synthase interactions is essential to activate the sluggish ATP synthase.
Collapse
Affiliation(s)
- Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
42
|
De Giovanni M, Chen H, Li X, Cyster JG. GPR35 and mediators from platelets and mast cells in neutrophil migration and inflammation. Immunol Rev 2023; 317:187-202. [PMID: 36928841 PMCID: PMC10504419 DOI: 10.1111/imr.13194] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Neutrophil recruitment from circulation to sites of inflammation is guided by multiple chemoattractant cues emanating from tissue cells, immune cells, and platelets. Here, we focus on the function of one G-protein coupled receptor, GPR35, in neutrophil recruitment. GPR35 has been challenging to study due the description of multiple ligands and G-protein couplings. Recently, we found that GPR35-expressing hematopoietic cells respond to the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA). We discuss distinct response profiles of GPR35 to 5-HIAA compared to other ligands. To place the functions of 5-HIAA in context, we summarize the actions of serotonin in vascular biology and leukocyte recruitment. Important sources of serotonin and 5-HIAA are platelets and mast cells. We discuss the dynamics of cell migration into inflamed tissues and how multiple platelet and mast cell-derived mediators, including 5-HIAA, cooperate to promote neutrophil recruitment. Additional actions of GPR35 in tissue physiology are reviewed. Finally, we discuss how clinically approved drugs that modulate serotonin uptake and metabolism may influence 5-HIAA-GPR35 function, and we speculate about broader influences of the GPR35 ligand-receptor system in immunity and disease.
Collapse
Affiliation(s)
- Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hongwen Chen
- Departments of Molecular Genetics and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaochun Li
- Departments of Molecular Genetics and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jason G. Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
43
|
Domínguez-Zorita S, Cuezva JM. The Mitochondrial ATP Synthase/IF1 Axis in Cancer Progression: Targets for Therapeutic Intervention. Cancers (Basel) 2023; 15:3775. [PMID: 37568591 PMCID: PMC10417293 DOI: 10.3390/cancers15153775] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer poses a significant global health problem with profound personal and economic implications on National Health Care Systems. The reprograming of metabolism is a major trait of the cancer phenotype with a clear potential for developing effective therapeutic strategies to combat the disease. Herein, we summarize the relevant role that the mitochondrial ATP synthase and its physiological inhibitor, ATPase Inhibitory Factor 1 (IF1), play in metabolic reprogramming to an enhanced glycolytic phenotype. We stress that the interplay in the ATP synthase/IF1 axis has additional functional roles in signaling mitohormetic programs, pro-oncogenic or anti-metastatic phenotypes depending on the cell type. Moreover, the same axis also participates in cell death resistance of cancer cells by restrained mitochondrial permeability transition pore opening. We emphasize the relevance of the different post-transcriptional mechanisms that regulate the specific expression and activity of ATP synthase/IF1, to stimulate further investigations in the field because of their potential as future targets to treat cancer. In addition, we review recent findings stressing that mitochondria metabolism is the primary altered target in lung adenocarcinomas and that the ATP synthase/IF1 axis of OXPHOS is included in the most significant signature of metastatic disease. Finally, we stress that targeting mitochondrial OXPHOS in pre-clinical mouse models affords a most effective therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| |
Collapse
|
44
|
Domínguez-Zorita S, Romero-Carramiñana I, Santacatterina F, Esparza-Moltó PB, Simó C, Del-Arco A, Núñez de Arenas C, Saiz J, Barbas C, Cuezva JM. IF1 ablation prevents ATP synthase oligomerization, enhances mitochondrial ATP turnover and promotes an adenosine-mediated pro-inflammatory phenotype. Cell Death Dis 2023; 14:413. [PMID: 37433784 DOI: 10.1038/s41419-023-05957-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/07/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
ATPase Inhibitory Factor 1 (IF1) regulates the activity of mitochondrial ATP synthase. The expression of IF1 in differentiated human and mouse cells is highly variable. In intestinal cells, the overexpression of IF1 protects against colon inflammation. Herein, we have developed a conditional IF1-knockout mouse model in intestinal epithelium to investigate the role of IF1 in mitochondrial function and tissue homeostasis. The results show that IF1-ablated mice have increased ATP synthase/hydrolase activities, leading to profound mitochondrial dysfunction and a pro-inflammatory phenotype that impairs the permeability of the intestinal barrier compromising mouse survival upon inflammation. Deletion of IF1 prevents the formation of oligomeric assemblies of ATP synthase and alters cristae structure and the electron transport chain. Moreover, lack of IF1 promotes an intramitochondrial Ca2+ overload in vivo, minimizing the threshold to Ca2+-induced permeability transition (mPT). Removal of IF1 in cell lines also prevents the formation of oligomeric assemblies of ATP synthase, minimizing the threshold to Ca2+-induced mPT. Metabolomic analyses of mice serum and colon tissue highlight that IF1 ablation promotes the activation of de novo purine and salvage pathways. Mechanistically, lack of IF1 in cell lines increases ATP synthase/hydrolase activities and installs futile ATP hydrolysis in mitochondria, resulting in the activation of purine metabolism and in the accumulation of adenosine, both in culture medium and in mice serum. Adenosine, through ADORA2B receptors, promotes an autoimmune phenotype in mice, stressing the role of the IF1/ATP synthase axis in tissue immune responses. Overall, the results highlight that IF1 is required for ATP synthase oligomerization and that it acts as a brake to prevent ATP hydrolysis under in vivo phosphorylating conditions in intestinal cells.
Collapse
Affiliation(s)
- Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carolina Simó
- Molecular Nutrition and Metabolism, Institute of Food Science Research (CIAL, CSIC-UAM), 28049, Madrid, Spain
| | - Araceli Del-Arco
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla la Mancha, Toledo, 45071, Spain
- Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina, Toledo, 45071, Spain
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jorge Saiz
- Centre of Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - Coral Barbas
- Centre of Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
45
|
De Giovanni M, Dang EV, Chen KY, An J, Madhani HD, Cyster JG. Platelets and mast cells promote pathogenic eosinophil recruitment during invasive fungal infection via the 5-HIAA-GPR35 ligand-receptor system. Immunity 2023; 56:1548-1560.e5. [PMID: 37279752 PMCID: PMC10360074 DOI: 10.1016/j.immuni.2023.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Cryptococcus neoformans is the leading cause of fungal meningitis and is characterized by pathogenic eosinophil accumulation in the context of type-2 inflammation. The chemoattractant receptor GPR35 is expressed by granulocytes and promotes their migration to the inflammatory mediator 5-hydroxyindoleacetic acid (5-HIAA), a serotonin metabolite. Given the inflammatory nature of cryptococcal infection, we examined the role of GPR35 in the circuitry underlying cell recruitment to the lung. GPR35 deficiency dampened eosinophil recruitment and fungal growth, whereas overexpression promoted eosinophil homing to airways and fungal replication. Activated platelets and mast cells were the sources of GPR35 ligand activity and pharmacological inhibition of serotonin conversion to 5-HIAA, or genetic deficiency in 5-HIAA production by platelets and mast cells resulted in more efficient clearance of Cryptococcus. Thus, the 5-HIAA-GPR35 axis is an eosinophil chemoattractant receptor system that modulates the clearance of a lethal fungal pathogen, with implications for the use of serotonin metabolism inhibitors in the treatment of fungal infections.
Collapse
Affiliation(s)
- Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Eric V Dang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin Y Chen
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
46
|
Balestrieri N, Palzkill V, Pass C, Tan J, Salyers ZR, Moparthy C, Murillo A, Kim K, Thome T, Yang Q, O’Malley KA, Berceli SA, Yue F, Scali ST, Ferreira LF, Ryan TE. Activation of the Aryl Hydrocarbon Receptor in Muscle Exacerbates Ischemic Pathology in Chronic Kidney Disease. Circ Res 2023; 133:158-176. [PMID: 37325935 PMCID: PMC10330629 DOI: 10.1161/circresaha.123.322875] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) accelerates the development of atherosclerosis, decreases muscle function, and increases the risk of amputation or death in patients with peripheral artery disease (PAD). However, the mechanisms underlying this pathobiology are ill-defined. Recent work has indicated that tryptophan-derived uremic solutes, which are ligands for AHR (aryl hydrocarbon receptor), are associated with limb amputation in PAD. Herein, we examined the role of AHR activation in the myopathy of PAD and CKD. METHODS AHR-related gene expression was evaluated in skeletal muscle obtained from mice and human PAD patients with and without CKD. AHRmKO (skeletal muscle-specific AHR knockout) mice with and without CKD were subjected to femoral artery ligation, and a battery of assessments were performed to evaluate vascular, muscle, and mitochondrial health. Single-nuclei RNA sequencing was performed to explore intercellular communication. Expression of the constitutively active AHR was used to isolate the role of AHR in mice without CKD. RESULTS PAD patients and mice with CKD displayed significantly higher mRNA expression of classical AHR-dependent genes (Cyp1a1, Cyp1b1, and Aldh3a1) when compared with either muscle from the PAD condition with normal renal function (P<0.05 for all 3 genes) or nonischemic controls. AHRmKO significantly improved limb perfusion recovery and arteriogenesis, preserved vasculogenic paracrine signaling from myofibers, increased muscle mass and strength, as well as enhanced mitochondrial function in an experimental model of PAD/CKD. Moreover, viral-mediated skeletal muscle-specific expression of a constitutively active AHR in mice with normal kidney function exacerbated the ischemic myopathy evidenced by smaller muscle masses, reduced contractile function, histopathology, altered vasculogenic signaling, and lower mitochondrial respiratory function. CONCLUSIONS These findings establish AHR activation in muscle as a pivotal regulator of the ischemic limb pathology in CKD. Further, the totality of the results provides support for testing of clinical interventions that diminish AHR signaling in these conditions.
Collapse
Affiliation(s)
- Nicholas Balestrieri
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Caroline Pass
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Zachary R. Salyers
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Chatick Moparthy
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Ania Murillo
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Kerri A. O’Malley
- Department of Surgery, The University of Florida, Gainesville, FL, USA
| | - Scott A. Berceli
- Department of Surgery, The University of Florida, Gainesville, FL, USA
| | - Feng Yue
- Department of Animal Sciences, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| | | | - Leonardo F. Ferreira
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
47
|
Yiu SPT, Zerbe C, Vanderwall D, Huttlin EL, Weekes MP, Gewurz BE. An Epstein-Barr virus protein interaction map reveals NLRP3 inflammasome evasion via MAVS UFMylation. Mol Cell 2023; 83:2367-2386.e15. [PMID: 37311461 PMCID: PMC10372749 DOI: 10.1016/j.molcel.2023.05.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/05/2023] [Accepted: 05/14/2023] [Indexed: 06/15/2023]
Abstract
Epstein-Barr virus (EBV) causes infectious mononucleosis, triggers multiple sclerosis, and is associated with 200,000 cancers/year. EBV colonizes the human B cell compartment and periodically reactivates, inducing expression of 80 viral proteins. However, much remains unknown about how EBV remodels host cells and dismantles key antiviral responses. We therefore created a map of EBV-host and EBV-EBV interactions in B cells undergoing EBV replication, uncovering conserved herpesvirus versus EBV-specific host cell targets. The EBV-encoded G-protein-coupled receptor BILF1 associated with MAVS and the UFM1 E3 ligase UFL1. Although UFMylation of 14-3-3 proteins drives RIG-I/MAVS signaling, BILF1-directed MAVS UFMylation instead triggered MAVS packaging into mitochondrial-derived vesicles and lysosomal proteolysis. In the absence of BILF1, EBV replication activated the NLRP3 inflammasome, which impaired viral replication and triggered pyroptosis. Our results provide a viral protein interaction network resource, reveal a UFM1-dependent pathway for selective degradation of mitochondrial cargo, and highlight BILF1 as a novel therapeutic target.
Collapse
Affiliation(s)
- Stephanie Pei Tung Yiu
- Division of Infectious Diseases, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Graduate Program in Virology, Boston, MA 02115, USA; Center for Integrated Solutions to Infectious Diseases, Broad Institute and Harvard Medical School, Cambridge, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Cassie Zerbe
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - David Vanderwall
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| | - Benjamin E Gewurz
- Division of Infectious Diseases, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Graduate Program in Virology, Boston, MA 02115, USA; Center for Integrated Solutions to Infectious Diseases, Broad Institute and Harvard Medical School, Cambridge, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Guo L, Gu Z. F-ATP synthase inhibitory factor 1 regulates metabolic reprogramming involving its interaction with c-Myc and PGC1α. Front Oncol 2023; 13:1207603. [PMID: 37469400 PMCID: PMC10352482 DOI: 10.3389/fonc.2023.1207603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/13/2023] [Indexed: 07/21/2023] Open
Abstract
F-ATP synthase inhibitory factor 1 (IF1) is an intrinsic inhibitor of F-ATP synthase. It is known that IF1 mediates metabolic phenotypes and cell fate, yet the molecular mechanisms through which IF1 fulfills its physiological functions are not fully understood. Ablation of IF1 favors metabolic switch to oxidative metabolism from glycolysis. c-Myc and PGC1α are critical for metabolic reprogramming. This work identified that IF1 interacted with Thr-58 phosphorylated c-Myc, which might thus mediate the activity of c-Myc and promote glycolysis. The interaction of IF1 with PGC1α inhibited oxidative respiration. c-Myc and PGC1α were localized to mitochondria under mitochondrial stress in an IF1-dependent manner. Furthermore, IF1 was found to be required for the protective effect of hypoxia on c-Myc- and PGC1α-induced cell death. This study suggested that the interactions of IF1 with transcription factors c-Myc and PGC1α might be involved in IF1-regulatory metabolic reprogramming and cell fate.
Collapse
Affiliation(s)
- Lishu Guo
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenglong Gu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| |
Collapse
|
49
|
Noble AJ, Nowak JK, Adams AT, Uhlig HH, Satsangi J. Defining Interactions Between the Genome, Epigenome, and the Environment in Inflammatory Bowel Disease: Progress and Prospects. Gastroenterology 2023; 165:44-60.e2. [PMID: 37062395 DOI: 10.1053/j.gastro.2023.03.238] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 04/18/2023]
Abstract
Recent advances in our understanding of the pathogenesis of inflammatory bowel disease (IBD) have highlighted the complex interplay between the genome, the epigenome, and the environment. Despite the exciting advances in genomics that have enabled the identification of over 200 susceptibility loci, these only account for a small proportion of the disease variance and the estimated heritability in IBD. It is likely that gene-environment (GxE) interactions contribute to "missing heritability" and these may act through epigenetic mechanisms. Several environmental factors, such as the microbiome, nutrition, and tobacco smoking, induce alterations in the epigenome of children and adults, which may impact disease susceptibility. Other mechanisms for GxE interactions are also directly pertinent in early life. We discuss a model in which environmental factors imprint disease risk in a window of susceptibility during infancy that may contribute to later disease onset, whereas other elements of the exposome act later in life and contribute directly to the pathogenesis and course of the disease. Understanding the mechanisms underlying GxE interactions may provide the basis for new therapeutic targets or preventative strategies for IBD.
Collapse
Affiliation(s)
- Alexandra J Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom.
| | - Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Alex T Adams
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Department of Pediatrics, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom; Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Seo SK, Kwon B. Immune regulation through tryptophan metabolism. Exp Mol Med 2023; 55:1371-1379. [PMID: 37394584 PMCID: PMC10394086 DOI: 10.1038/s12276-023-01028-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 07/04/2023] Open
Abstract
Amino acids are fundamental units of molecular components that are essential for sustaining life; however, their metabolism is closely interconnected to the control systems of cell function. Tryptophan (Trp) is an essential amino acid catabolized by complex metabolic pathways. Several of the resulting Trp metabolites are bioactive and play central roles in physiology and pathophysiology. Additionally, various physiological functions of Trp metabolites are mutually regulated by the gut microbiota and intestine to coordinately maintain intestinal homeostasis and symbiosis under steady state conditions and during the immune response to pathogens and xenotoxins. Cancer and inflammatory diseases are associated with dysbiosis- and host-related aberrant Trp metabolism and inactivation of the aryl hydrocarbon receptor (AHR), which is a receptor of several Trp metabolites. In this review, we focus on the mechanisms through which Trp metabolism converges to AHR activation for the modulation of immune function and restoration of tissue homeostasis and how these processes can be targeted using therapeutic approaches for cancer and inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Su-Kil Seo
- Department of Microbiology and Immunology, College of Medicine Inje University, Busan, 47392, Republic of Korea.
- Parenchyma Biotech, Busan, 47392, Republic of Korea.
| | - Byungsuk Kwon
- Parenchyma Biotech, Busan, 47392, Republic of Korea.
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea.
| |
Collapse
|