1
|
Wang W, Wang XM, Zhang HL, Zhao R, Wang Y, Zhang HL, Song ZJ. Molecular and metabolic landscape of adenosine triphosphate-induced cell death in cardiovascular disease. World J Cardiol 2024; 16:689-706. [PMID: 39734818 PMCID: PMC11669974 DOI: 10.4330/wjc.v16.i12.689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
The maintenance of intracellular and extracellular adenosine triphosphate (ATP) levels plays a pivotal role in cardiac function. In recent years, burgeoning attention has been directed towards ATP-induced cell death (AICD), revealing it as a distinct cellular demise pathway triggered by heightened extracellular ATP concentrations, distinguishing it from other forms of cell death such as apoptosis and necrosis. AICD is increasingly acknowledged as a critical mechanism mediating the pathogenesis and progression of various cardiovascular maladies, encompassing myocardial ischemia-reperfusion injury, sepsis-induced cardiomyopathy, hypertrophic cardiomyopathy, arrhythmia, and diabetic cardiomyopathy. Consequently, a comprehensive understanding of the molecular and metabolic underpinnings of AICD in cardiac tissue holds promise for the prevention and amelioration of cardiovascular diseases. This review first elucidates the vital physiological roles of ATP in the cardiovascular system, subsequently delving into the intricate molecular mechanisms and metabolic signatures governing AICD. Furthermore, it addresses the potential therapeutic targets implicated in mitigating AICD for treating cardiovascular diseases, while also delineating the current constraints and future avenues for these innovative therapeutic targets, thereby furnishing novel insights and strategies for the prevention and management of cardiovascular disorders.
Collapse
Affiliation(s)
- Wei Wang
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Xue-Mei Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 73000, Gansu Province, China
| | - Hao-Long Zhang
- University Sains Malaysia, Advanced Medical and Dental Institute, Penang 13200, Malaysia
| | - Rui Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yong Wang
- Department of Pathology Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Hao-Ling Zhang
- Department of Biomedical Science, Advanced Medical and Dental Institute, University Sains Malaysia, Penang 13200, Malaysia
| | - Zhi-Jing Song
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
2
|
Zhang J, Wang J, Gu Z, Liu X. Study on the Mechanism and Potential of Corbicula fluminea (Asian Clam) in Removing Copper and Cadmium from Aquaculture Ponds. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 27:2. [PMID: 39560786 DOI: 10.1007/s10126-024-10389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/01/2024] [Indexed: 11/20/2024]
Abstract
The issue of heavy metal pollution in aquaculture ponds is becoming increasingly severe, posing a significant threat to the healthy development of the aquaculture industry. Heavy metals such as cadmium and copper accumulate in ponds, not only exerting toxic effects on aquatic organisms and affecting their growth and reproduction but also endangering human health through the food chain. Bioremediation, as a green and environmentally friendly technology, utilizes specific organisms to absorb, transform, and immobilize heavy metals. We examined metal accumulation, traditional metal-related biomarkers, alongside transcriptomic and tissue histological analyses, in the hepatopancreas of Corbicula fluminea following a 14-day exposure to copper (20 µg/L), cadmium (20 µg/L), or combined copper-cadmium treatments (20 µg/L Cu and 20 µg/L Cd). Metal exposure led to notable metal accumulation in the clam's hepatopancreas. Analysis of traditional biomarkers revealed signs of cellular injury and oxidative stress in clams post-metal exposure. Transcriptomic analysis across the three treatment groups revealed disruptions in immune response, response to metal ion, and energy metabolism, characterized by differential expression levels of key genes such as ABCA3, MYD88, TOLLIP, TBK1, C2, C4, c-Myc, SYK, and SAMHD1. These findings deepen our understanding of the adverse effects of metal exposure on freshwater organisms and evaluate the potential of Corbicula fluminea for removing heavy metals from aquaculture ponds.
Collapse
Affiliation(s)
- Jiahua Zhang
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200092, China.
- Key Laboratory of Aquaculture Facilities Engineering, Ministry of Agriculture and Rural Affairs, Shanghai, China.
| | - Jie Wang
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200092, China
- Key Laboratory of Aquaculture Facilities Engineering, Ministry of Agriculture and Rural Affairs, Shanghai, China
| | - Zhaojun Gu
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200092, China
- Key Laboratory of Aquaculture Facilities Engineering, Ministry of Agriculture and Rural Affairs, Shanghai, China
| | - Xingguo Liu
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200092, China
- Key Laboratory of Aquaculture Facilities Engineering, Ministry of Agriculture and Rural Affairs, Shanghai, China
| |
Collapse
|
3
|
Gerasimovskaya E, Patil RS, Davies A, Maloney ME, Simon L, Mohamed B, Cherian-Shaw M, Verin AD. Extracellular purines in lung endothelial permeability and pulmonary diseases. Front Physiol 2024; 15:1450673. [PMID: 39234309 PMCID: PMC11372795 DOI: 10.3389/fphys.2024.1450673] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The purinergic signaling system is an evolutionarily conserved and critical regulatory circuit that maintains homeostatic balance across various organ systems and cell types by providing compensatory responses to diverse pathologies. Despite cardiovascular diseases taking a leading position in human morbidity and mortality worldwide, pulmonary diseases represent significant health concerns as well. The endothelium of both pulmonary and systemic circulation (bronchial vessels) plays a pivotal role in maintaining lung tissue homeostasis by providing an active barrier and modulating adhesion and infiltration of inflammatory cells. However, investigations into purinergic regulation of lung endothelium have remained limited, despite widespread recognition of the role of extracellular nucleotides and adenosine in hypoxic, inflammatory, and immune responses within the pulmonary microenvironment. In this review, we provide an overview of the basic aspects of purinergic signaling in vascular endothelium and highlight recent studies focusing on pulmonary microvascular endothelial cells and endothelial cells from the pulmonary artery vasa vasorum. Through this compilation of research findings, we aim to shed light on the emerging insights into the purinergic modulation of pulmonary endothelial function and its implications for lung health and disease.
Collapse
Affiliation(s)
| | - Rahul S Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adrian Davies
- Department of Internal Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - McKenzie E Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liselle Simon
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Basmah Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
4
|
Cash E, Goodwin AT, Tatler AL. Adenosine receptor signalling as a driver of pulmonary fibrosis. Pharmacol Ther 2023; 249:108504. [PMID: 37482099 DOI: 10.1016/j.pharmthera.2023.108504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Pulmonary fibrosis is a debilitating and life-limiting lung condition in which the damage- response mechanisms of mixed-population cells within the lungs go awry. The tissue microenvironment is drastically remodelled by aberrantly activated fibroblasts which deposit ECM components into the surrounding lung tissue, detrimentally affecting lung function and capacity for gas exchange. Growing evidence suggests a role for adenosine signalling in the pathology of tissue fibrosis in a variety of organs, including the lung, but the molecular pathways through which this occurs remain largely unknown. This review explores the role of adenosine in fibrosis and evaluates the contribution of the different adenosine receptors to fibrogenesis. Therapeutic targeting of the adenosine receptors is also considered, along with clinical observations pointing towards a role for adenosine in fibrosis. In addition, the interaction between adenosine signalling and other profibrotic signalling pathways, such as TGFβ1 signalling, is discussed.
Collapse
Affiliation(s)
- Emily Cash
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - Amanda T Goodwin
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - Amanda L Tatler
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, Nottingham, UK.
| |
Collapse
|
5
|
Zhao Y, Huang S, Xie R, Liu J. Extracellular ATP accelerates cell death and decreases tight junction protein ZO-1 in hypoxic cochlear strial marginal cells in neonatal rats. Cell Signal 2023:110732. [PMID: 37245680 DOI: 10.1016/j.cellsig.2023.110732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023]
Abstract
In the cochlear, extracellular ATP (eATP) plays an important role in both physiological and pathological processes, but its role in the hypoxic cochlear remains unclear. The present study aims to investigate the relationship between eATP and hypoxic marginal cells (MCs) in the stria vascularis in cochlear. Combining various methodologies, we found that eATP accelerates cell death and decreases tight junction protein zonula occludens-1 (ZO-1) in hypoxic MCs. Flow cytometry and western blot analyses revealed an increase in apoptosis levels and suppression of autophagy, indicating that eATP causes additional cell death by increasing the apoptosis of hypoxic MCs. Given that autophagy inhibits apoptosis to protect MCs under hypoxia, apoptosis is probably enchanced by suppressing autophagy. Interleukin-33(IL-33)/suppression of tumorigenicity-2(ST-2)/matrix metalloprotein 9(MMP9) pathway activation was also observed during the process. Further experiments involving the use of additional IL-33 protein and an MMP9 inhibitor indicated that this pathway is responsible for the damage to the ZO-1 protein in hypoxic MCs. Our study revealed the adverse effect of eATP on the survival and ZO-1 protein expression of hypoxic MCs, as well as the underlying mechanism.
Collapse
Affiliation(s)
- Yanyun Zhao
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sihan Huang
- Department of Otorhinolaryngology, Zhangzhou Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Renwei Xie
- Department of Otorhinolaryngology, Renhe Hospital, Baoshan District, Shanghai, China
| | - Jun Liu
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
6
|
Fu Z, Zhao PY, Yang XP, Li H, Hu SD, Xu YX, Du XH. Cannabidiol regulates apoptosis and autophagy in inflammation and cancer: A review. Front Pharmacol 2023; 14:1094020. [PMID: 36755953 PMCID: PMC9899821 DOI: 10.3389/fphar.2023.1094020] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Cannabidiol (CBD) is a terpenoid naturally found in plants. The purified compound is used in the treatment of mental disorders because of its antidepressive, anxiolytic, and antiepileptic effects. CBD can affect the regulation of several pathophysiologic processes, including autophagy, cytokine secretion, apoptosis, and innate and adaptive immune responses. However, several authors have reported contradictory findings concerning the magnitude and direction of CBD-mediated effects. For example, CBD treatment can increase, decrease, or have no significant effect on autophagy and apoptosis. These variable results can be attributed to the differences in the biological models, cell types, and CBD concentration used in these studies. This review focuses on the mechanism of regulation of autophagy and apoptosis in inflammatory response and cancer by CBD. Further, we broadly elaborated on the prospects of using CBD as an anti-inflammatory agent and in cancer therapy in the future.
Collapse
Affiliation(s)
- Ze Fu
- Medical School of Chinese PLA, Beijing, China
| | | | | | - Hao Li
- Medical School of Chinese PLA, Beijing, China
| | - Shi-Dong Hu
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ying-Xin Xu
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Hui Du
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiao-Hui Du,
| |
Collapse
|
7
|
Kinsella S, Evandy CA, Cooper K, Cardinale A, Iovino L, deRoos P, Hopwo KS, Smith CW, Granadier D, Sullivan LB, Velardi E, Dudakov JA. Damage-induced pyroptosis drives endog thymic regeneration via induction of Foxn1 by purinergic receptor activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524800. [PMID: 36711570 PMCID: PMC9882324 DOI: 10.1101/2023.01.19.524800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Endogenous thymic regeneration is a crucial process that allows for the renewal of immune competence following stress, infection or cytoreductive conditioning. Fully understanding the molecular mechanisms driving regeneration will uncover therapeutic targets to enhance regeneration. We previously demonstrated that high levels of homeostatic apoptosis suppress regeneration and that a reduction in the presence of damage-induced apoptotic thymocytes facilitates regeneration. Here we identified that cell-specific metabolic remodeling after ionizing radiation steers thymocytes towards mitochondrial-driven pyroptotic cell death. We further identified that a key damage-associated molecular pattern (DAMP), ATP, stimulates the cell surface purinergic receptor P2Y2 on cortical thymic epithelial cells (cTECs) acutely after damage, enhancing expression of Foxn1, the critical thymic transcription factor. Targeting the P2Y2 receptor with the agonist UTPγS promotes rapid regeneration of the thymus in vivo following acute damage. Together these data demonstrate that intrinsic metabolic regulation of pyruvate processing is a critical process driving thymus repair and identifies the P2Y2 receptor as a novel molecular therapeutic target to enhance thymus regeneration.
Collapse
Affiliation(s)
- Sinéad Kinsella
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Cindy A Evandy
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Kirsten Cooper
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Antonella Cardinale
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, 00146, Italy
| | - Lorenzo Iovino
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Paul deRoos
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Kayla S Hopwo
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - Colton W Smith
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
| | - David Granadier
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
- Medical Scientist Training Program, University of Washington, Seattle WA, 98195, US
| | - Lucas B Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, 00146, Italy
| | - Jarrod A Dudakov
- Program in Immunology, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle WA, 98109, US
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle WA, 98109, US
- Department of Immunology, University of Washington, Seattle WA, 98195, US
| |
Collapse
|
8
|
Feliu C, Konecki C, Cazaubon Y, Binet L, Vautier D, Fouley A, Gozalo C, Djerada Z. Development and Validation of a Non-Targeted Screening Method for Most Psychoactive, Analgesic, Anaesthetic, Anti-Diabetic, Anti-Coagulant and Anti-Hypertensive Drugs in Human Whole Blood and Plasma Using High-Resolution Mass Spectrometry. Pharmaceuticals (Basel) 2023; 16:ph16010076. [PMID: 36678573 PMCID: PMC9865759 DOI: 10.3390/ph16010076] [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: 11/21/2022] [Revised: 12/16/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
(1) Background: In toxicological laboratories, various screening methods can be used to identify compounds involved in intoxication. High-resolution mass spectrometry has been increasingly used in this context for the last years, because of its sensitivity and reliability. Here, we present the development and validation of a screening method that uses liquid chromatography coupled with a high-resolution mass spectrometer. (2) Methods: This method required only 100 µL of whole blood or plasma sample. Pretreatment consisted of a rapid and simple deproteinisation with methanol/acetonitrile and zinc sulphate. This new assay was validated according to international guidelines. (3) Results: To perform the method validation, 53 compounds were selected. The selection criteria were as follows: various chemical structures and therapeutic families (>15), large m/z distribution, positive or negative ionisation mode, and various elution times. The assays showed high selectivity and specificity, with optimal process efficiency. The identification limits, determined using predefined criteria, were established at sub-therapeutic or therapeutic concentrations. Applicability was evaluated using spiked plasma controls and external quality controls. (4) Conclusions: The new method was then successfully applied to routine clinical and forensic samples.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
- Correspondence: (C.F.); (Z.D.)
| | - Celine Konecki
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
| | - Yoann Cazaubon
- Institute Desbrest of Epidemiology and Public Health, INSERM, Montpellier University, Department of Pharmacology, Montpellier University Hospital, Avenue du Doyen Gaston Giraud, 34090 Montpellier, France
| | - Laurent Binet
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
| | - Damien Vautier
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
| | - Aurélie Fouley
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
| | - Claire Gozalo
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
| | - Zoubir Djerada
- Department of Pharmacology, EA 3801, SFR CAP-Santé, Reims University Hospital, 51 Rue Cognacq-Jay, CEDEX, 51095 Reims, France
- Correspondence: (C.F.); (Z.D.)
| |
Collapse
|
9
|
Zhao D, Hu G, Chen R, Xiao G, Teng S. Molecular cloning, characterization, and tissue distribution of c-Myc from blood clam Tegillarca granosa and its role in cadmium-induced stress response. Gene 2022; 834:146611. [PMID: 35618219 DOI: 10.1016/j.gene.2022.146611] [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: 12/23/2021] [Revised: 04/28/2022] [Accepted: 05/19/2022] [Indexed: 11/20/2022]
Abstract
Cadmium (Cd) pollution threatens the cultivation of the blood clam Tegillarca granosa (T. granosa) in coastal regions of the East China Sea. The molecular mechanisms regulating Cd stress response and detoxification in blood clams are largely unclear. In the present study, the full-length T. granosa c-Myc (Tgc-Myc) cDNA was cloned for the first time. The 3063-bp cDNA consisted of a 129-bp 5' untranslated region (UTR), a 1746-bp 3' UTR, and a 1188-bp open reading frame encoding a predicted protein of 395 amino acid residues. The predicted protein had a calculated molecular weight of 44.9 kDa and an estimated isoelectric point of 6.82. The predicted protein contained an N-terminal transactivation domain and a C-terminal basic helix-loop-helix leucine zipper domain, which are conserved functional domains of c-Myc proteins. Tgc-Myc showed broad tissue distribution in blood clams, with the highest expression detected in the gill and hepatopancreas. Exposure to Cd, a major heavy metal pollutant in coastal regions of the East China Sea, induced Tgc-Myc expression in gill tissues. Tgc-Myc knockdown led to reduced expression of a variety of stress response/detoxification genes in blood clams cultivated in Cd-contaminated seawater. Tgc-Myc knockdown also led to decreased expression of IGF1R, a proto-oncogene that promotes cell proliferation. These findings indicated that Tgc-Myc regulates Cd-induced stress response and detoxification in blood clams. The upregulation of Tgc-Myc may serve as an approach to generate strains with an enhanced detoxification response and consequently a low heavy metal buildup.
Collapse
Affiliation(s)
- Defeng Zhao
- Zhejiang Mariculture Research Institute, Zhejiang Key Lab of Exploitation and Preservation of Coastal Bio-Resource, Wenzhou Key Laboratory of Marine Biological Genetics and Breeding, Wenzhou 325005, China; Zhejiang Ocean University, Zhoushan 316022, China
| | - Gaoyu Hu
- Zhejiang Mariculture Research Institute, Zhejiang Key Lab of Exploitation and Preservation of Coastal Bio-Resource, Wenzhou Key Laboratory of Marine Biological Genetics and Breeding, Wenzhou 325005, China
| | - Ran Chen
- Zhejiang Mariculture Research Institute, Zhejiang Key Lab of Exploitation and Preservation of Coastal Bio-Resource, Wenzhou Key Laboratory of Marine Biological Genetics and Breeding, Wenzhou 325005, China
| | - Guoqiang Xiao
- Zhejiang Mariculture Research Institute, Zhejiang Key Lab of Exploitation and Preservation of Coastal Bio-Resource, Wenzhou Key Laboratory of Marine Biological Genetics and Breeding, Wenzhou 325005, China; Zhejiang Ocean University, Zhoushan 316022, China
| | - Shuangshuang Teng
- Zhejiang Mariculture Research Institute, Zhejiang Key Lab of Exploitation and Preservation of Coastal Bio-Resource, Wenzhou Key Laboratory of Marine Biological Genetics and Breeding, Wenzhou 325005, China.
| |
Collapse
|
10
|
Simulated hypoxia modulates P2X7 receptor function in mice peritoneal macrophages. Int Immunopharmacol 2022; 110:109062. [PMID: 35863257 DOI: 10.1016/j.intimp.2022.109062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
The inflammatory focus is similar to the tumor microenvironment, which contains a complex milieu with immune cells and macrophages. The accumulation of cells promotes local pH and O2 tension decline (hypoxia). Local O2 tension decline activates hypoxia-inducible factor α and β (HIF-1α and HIF-1β adenosine triphosphate (ATP) release. ATP activates the P2X7 receptor and modulates ischemic/hypoxic conditions. Similarly, α1α may regulate P2X7 receptor expression in the hypoxic microenvironment. Therefore, we investigated P2X7 receptor function under simulated hypoxic conditions by pretreating peritoneal macrophages with mitochondrial electron transport chain complex inhibitors (simulated hypoxia). Treatment with mitochondrial electron transport chain complex inhibitors until three hours of exposure did not cause LDH release. Additionally, mitochondrial electron transport chain complex inhibitors increased ATP-induced P2X7 receptor function without being able to directly activate this receptor. Other P2 receptor subtypes do not appear to participate in this mechanism. Simulated hypoxia augmented HIF-1α levels and suppressed HIF-1α and P2X7 receptor antagonists. Similarly, simulated hypoxia increased ATP-induced dye uptake and inhibited HIF-1α antagonists. Another factor activated in simulated hypoxic conditions was the intracellular P2X7 receptor regulator PIP2. Treatment with HIF-1α agonists increased PIP2 levels and reversed the effects of HIF-1α and P2X7 receptor antagonists. Additionally, the improved ATP-induced dye uptake caused by the simulated hypoxia stimulus was inhibited by P2X7 receptor and PIP2 antagonists. Therefore, simulated hypoxia may augment P2X7 receptor activity for a pathway dependent on HIF-1α and PIP2 activation.
Collapse
|
11
|
Blocking connexin 43 and its promotion of ATP release from renal tubular epithelial cells ameliorates renal fibrosis. Cell Death Dis 2022; 13:511. [PMID: 35641484 PMCID: PMC9156700 DOI: 10.1038/s41419-022-04910-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023]
Abstract
Whether metabolites derived from injured renal tubular epithelial cells (TECs) participate in renal fibrosis is poorly explored. After TEC injury, various metabolites are released and among the most potent is adenosine triphosphate (ATP), which is released via ATP-permeable channels. In these hemichannels, connexin 43 (Cx43) is the most common member. However, its role in renal interstitial fibrosis (RIF) has not been fully examined. We analyzed renal samples from patients with obstructive nephropathy and mice with unilateral ureteral obstruction (UUO). Cx43-KSP mice were generated to deplete Cx43 in TECs. Through transcriptomics, metabolomics, and single-cell sequencing multi-omics analysis, the relationship among tubular Cx43, ATP, and macrophages in renal fibrosis was explored. The expression of Cx43 in TECs was upregulated in both patients and mice with obstructive nephropathy. Knockdown of Cx43 in TECs or using Cx43-specific inhibitors reduced UUO-induced inflammation and fibrosis in mice. Single-cell RNA sequencing showed that ATP specific receptors, including P2rx4 and P2rx7, were distributed mainly on macrophages. We found that P2rx4- or P2rx7-positive macrophages underwent pyroptosis after UUO, and in vitro ATP directly induced pyroptosis by macrophages. The administration of P2 receptor or P2X7 receptor blockers to UUO mice inhibited macrophage pyroptosis and demonstrated a similar degree of renoprotection as Cx43 genetic depletion. Further, we found that GAP 26 (a Cx43 hemichannel inhibitor) and A-839977 (an inhibitor of the pyroptosis receptor) alleviated UUO-induced fibrosis, while BzATP (the agonist of pyroptosis receptor) exacerbated fibrosis. Single-cell sequencing demonstrated that the pyroptotic macrophages upregulated the release of CXCL10, which activated intrarenal fibroblasts. Cx43 mediates the release of ATP from TECs during renal injury, inducing peritubular macrophage pyroptosis, which subsequently leads to the release of CXCL10 and activation of intrarenal fibroblasts and acceleration of renal fibrosis.
Collapse
|
12
|
Wang P, Wang S, Chen H, Deng X, Zhang L, Xu H, Yang H. TCMIP v2.0 Powers the Identification of Chemical Constituents Available in Xinglou Chengqi Decoction and the Exploration of Pharmacological Mechanisms Acting on Stroke Complicated With Tanre Fushi Syndrome. Front Pharmacol 2021; 12:598200. [PMID: 34335236 PMCID: PMC8320350 DOI: 10.3389/fphar.2021.598200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Xinglou Chengqi (XLCQ) decoction, composed of three botanical drugs and one inorganic drug, is used in clinics during the treatment of acute stroke complicated with Tanre Fushi (TRFS) syndrome in China. However, its active ingredients and the molecular mechanism have not been clarified. So, we aimed to preliminarily characterize its chemical constituents and investigate its pharmacological mechanisms using an integrative pharmacology strategy, including component analysis, network prediction, and experimental verification. We employed UPLC-QTOF-MS/MS to describe the chemical profile of XLCQ, Integrative Pharmacology-based Network Computational Research Platform of Traditional Chinese Medicine (TCMIP v2.0, http://www.tcmip.cn/), to assist in identifying the chemical components and predict the putative molecular mechanism against acute stroke complicated with TRFS, and LPS-stimulated BV-2 cells to verify the anti-neuroinflammatory effects of luteolin, apigenin, and chrysoeriol. Altogether, 197 chemical compounds were identified or tentatively characterized in the water extraction of XLCQ, 22 of them were selected as the key active constituents that may improve the pathological state by regulating 27 corresponding targets that are mainly involved in inflammation/immune-related pathways, and furthermore, luteolin, apigenin, and chrysoeriol exhibited good anti-neuroinflammatory effects from both protein and mRNA levels. In summary, it is the first time to employ an integrative pharmacology strategy to delineate 22 constituents that may improve the pathological state of stroke with TRFS by regulating 27 corresponding targets, which may offer a highly efficient way to mine the scientific connotation of traditional Chinese medicine prescriptions. This study might be a supplement for the deficiency of the basic research of XLCQ.
Collapse
Affiliation(s)
- Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hong Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofang Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luoqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Shaanxi Institute of International Trade and Commerce, Xianyang, China
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Yang H, Su J, Meng W, Chen X, Xu Y, Sun B. MiR-518a-5p Targets GZMB to Extenuate Vascular Endothelial Cell Injury Induced by Hypoxia-Reoxygenation and Thereby Improves Myocardial Ischemia. Int Heart J 2021; 62:658-665. [PMID: 33994508 DOI: 10.1536/ihj.20-619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To probe the function of miR-518a-5p/Granzyme B (GZMB) in hypoxia/reoxygenation (H/R) -induced vascular endothelial cell injury.The key genes of myocardial infarction were screened by bioinformatic methods. The upstream micro RNAs (miRNAs) of GZMB were predicted by TargetScan. The binding of miR-518a-5p to GZMB was verified with luciferase reporter assay. The H/R model was constructed with human vascular endothelial cell (HUVEC) in vitro. Cell Counting Kit-8 (CCK8) assay was performed to detect cell proliferation. Western blot was utilized to evaluate the levels of indicated proteins.GZMB was up-regulated in patients with myocardial infarction and identified as the key gene by the bioinformatics analysis. Then the prediction from TargetScan indicated that miR-518a-5p, which is down-regulated in myocardial infarction patients, might be the potential upstream miRNA for GZMB. The following experiments verified that miR-518a-5p could bind to the 3'UTR of GZMB and negatively modulates GZMB expression. More importantly, the miR-518a-5p mimic enhanced cell proliferation and repressed apoptosis of H/R-injured HUVEC cells by inhibiting GZMB expression.We proved that miR-518a-5p could partly attenuate H/R-induced HUVEC cell injury by targeting GZMB, and perhaps the miR-518a-5p/GZMB axis could be potential therapeutic targets for myocardial infarction.
Collapse
Affiliation(s)
- Hui Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Jingjing Su
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Weixin Meng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Xiaoya Chen
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University
| | - Ying Xu
- Editorial Department, Journal of Harbin Medical University
| | - Bo Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| |
Collapse
|
14
|
Pereira-Figueiredo D, Nascimento AA, Cunha-Rodrigues MC, Brito R, Calaza KC. Caffeine and Its Neuroprotective Role in Ischemic Events: A Mechanism Dependent on Adenosine Receptors. Cell Mol Neurobiol 2021; 42:1693-1725. [PMID: 33730305 DOI: 10.1007/s10571-021-01077-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Ischemia is characterized by a transient, insufficient, or permanent interruption of blood flow to a tissue, which leads to an inadequate glucose and oxygen supply. The nervous tissue is highly active, and it closely depends on glucose and oxygen to satisfy its metabolic demand. Therefore, ischemic conditions promote cell death and lead to a secondary wave of cell damage that progressively spreads to the neighborhood areas, called penumbra. Brain ischemia is one of the main causes of deaths and summed with retinal ischemia comprises one of the principal reasons of disability. Although several studies have been performed to investigate the mechanisms of damage to find protective/preventive interventions, an effective treatment does not exist yet. Adenosine is a well-described neuromodulator in the central nervous system (CNS), and acts through four subtypes of G-protein-coupled receptors. Adenosine receptors, especially A1 and A2A receptors, are the main targets of caffeine in daily consumption doses. Accordingly, caffeine has been greatly studied in the context of CNS pathologies. In fact, adenosine system, as well as caffeine, is involved in neuroprotection effects in different pathological situations. Therefore, the present review focuses on the role of adenosine/caffeine in CNS, brain and retina, ischemic events.
Collapse
Affiliation(s)
- D Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil
| | - A A Nascimento
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - M C Cunha-Rodrigues
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - R Brito
- Laboratory of Neuronal Physiology and Pathology, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil. .,Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
15
|
Meyer AV, Klein D, de Leve S, Szymonowicz K, Stuschke M, Robson SC, Jendrossek V, Wirsdörfer F. Host CD39 Deficiency Affects Radiation-Induced Tumor Growth Delay and Aggravates Radiation-Induced Normal Tissue Toxicity. Front Oncol 2020; 10:554883. [PMID: 33194619 PMCID: PMC7649817 DOI: 10.3389/fonc.2020.554883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
The ectonucleoside triphosphate diphosphohydrolase (CD39)/5′ ectonuclotidase (CD73)-dependent purinergic pathway emerges as promising cancer target. Yet, except for own previous work revealing a pathogenic role of CD73 and adenosine in radiation-induced lung fibrosis, the role of purinergic signaling for radiotherapy outcome remained elusive. Here we used C57BL/6 wild-type (WT), CD39 knockout (CD39−/−), and CD73 knockout (CD73−/−) mice and hind-leg tumors of syngeneic murine Lewis lung carcinoma cells (LLC1) to elucidate how host purinergic signaling shapes the growth of LLC1 tumors to a single high-dose irradiation with 10 Gy in vivo. In complementary in vitro experiments, we examined the radiation response of LLC1 cells in combination with exogenously added ATP or adenosine, the proinflammatory and anti-inflammatory arms of purinergic signaling. Finally, we analyzed the impact of genetic loss of CD39 on pathophysiologic lung changes associated with lung fibrosis induced by a single-dose whole-thorax irradiation (WTI) with 15 Gy. Loss of CD73 in the tumor host did neither significantly affect tumor growth nor the radiation response of the CD39/CD73-negative LLC1 tumors. In contrast, LLC1 tumors exhibited a tendency to grow faster in CD39−/− mice compared to WT mice. Even more important, tumors grown in the CD39-deficient background displayed a significantly reduced tumor growth delay upon irradiation when compared to irradiated tumors grown on WT mice. CD39 deficiency caused only subtle differences in the immune compartment of irradiated LLC1 tumors compared to WT mice. Instead, we could associate the tumor growth and radioresistance-promoting effects of host CD39 deficiency to alterations in the tumor endothelial compartment. Importantly, genetic deficiency of CD39 also augmented the expression level of fibrosis-associated osteopontin in irradiated normal lungs and exacerbated radiation-induced lung fibrosis at 25 weeks after irradiation. We conclude that genetic loss of host CD39 alters the tumor microenvironment, particularly the tumor microvasculature, and thereby promotes growth and radioresistance of murine LLC1 tumors. In the normal tissue loss of host, CD39 exacerbates radiation-induced adverse late effects. The suggested beneficial roles of host CD39 on the therapeutic ratio of radiotherapy suggest that therapeutic strategies targeting CD39 in combination with radiotherapy have to be considered with caution.
Collapse
Affiliation(s)
- Alina V Meyer
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Simone de Leve
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Klaudia Szymonowicz
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- Department of Radiotherapy, University Hospital Essen, Essen, Germany
| | - Simon C Robson
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Verena Jendrossek
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
16
|
Strassheim D, Verin A, Batori R, Nijmeh H, Burns N, Kovacs-Kasa A, Umapathy NS, Kotamarthi J, Gokhale YS, Karoor V, Stenmark KR, Gerasimovskaya E. P2Y Purinergic Receptors, Endothelial Dysfunction, and Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21186855. [PMID: 32962005 PMCID: PMC7555413 DOI: 10.3390/ijms21186855] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Purinergic G-protein-coupled receptors are ancient and the most abundant group of G-protein-coupled receptors (GPCRs). The wide distribution of purinergic receptors in the cardiovascular system, together with the expression of multiple receptor subtypes in endothelial cells (ECs) and other vascular cells demonstrates the physiological importance of the purinergic signaling system in the regulation of the cardiovascular system. This review discusses the contribution of purinergic P2Y receptors to endothelial dysfunction (ED) in numerous cardiovascular diseases (CVDs). Endothelial dysfunction can be defined as a shift from a “calm” or non-activated state, characterized by low permeability, anti-thrombotic, and anti-inflammatory properties, to a “activated” state, characterized by vasoconstriction and increased permeability, pro-thrombotic, and pro-inflammatory properties. This state of ED is observed in many diseases, including atherosclerosis, diabetes, hypertension, metabolic syndrome, sepsis, and pulmonary hypertension. Herein, we review the recent advances in P2Y receptor physiology and emphasize some of their unique signaling features in pulmonary endothelial cells.
Collapse
Affiliation(s)
- Derek Strassheim
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | - Robert Batori
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | - Hala Nijmeh
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
| | - Nana Burns
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Anita Kovacs-Kasa
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | | | - Janavi Kotamarthi
- The Department of BioMedical Engineering, University of Wisconsin, Madison, WI 53706, USA; (J.K.); (Y.S.G.)
| | - Yash S. Gokhale
- The Department of BioMedical Engineering, University of Wisconsin, Madison, WI 53706, USA; (J.K.); (Y.S.G.)
| | - Vijaya Karoor
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Kurt R. Stenmark
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
| | - Evgenia Gerasimovskaya
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
- Correspondence: ; Tel.: +1-303-724-5614
| |
Collapse
|
17
|
Feliu C, Peyret H, Vautier D, Djerada Z. Simultaneous quantification of 8 nucleotides and adenosine in cells and their medium using UHPLC-HRMS. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122156. [PMID: 32446186 DOI: 10.1016/j.jchromb.2020.122156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Purinergic signalling is involved in physiological processes, particularly during ischemia-reperfusion injuries for which it has a protective effect. The purpose of this work was to develop a method for simultaneous quantification of eight nucleotides and adenosine in biological matrices by liquid chromatography coupled with high-resolution mass spectrometry. A method was developed that was sufficiently robust to quantify the targeted analytes in 20 min with good sensitivity. Analysis of extracellular media from cultured endothelial cells detected the release of nucleotides and adenosine during 2 h of hypoxia. The quantification of cylic adenosine monophosphate (cAMP) allowed to establish a dose-response curve after receptor stimulation. Therefore, our method allows us to study the involvement of nucleotides in various processes in both the intracellular and extracellular compartment.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Damien Vautier
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France.
| |
Collapse
|
18
|
Feliu C, Peyret H, Brassart-Pasco S, Oszust F, Poitevin G, Nguyen P, Millart H, Djerada Z. Ticagrelor Prevents Endothelial Cell Apoptosis through the Adenosine Signalling Pathway in the Early Stages of Hypoxia. Biomolecules 2020; 10:biom10050740. [PMID: 32397519 PMCID: PMC7277469 DOI: 10.3390/biom10050740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several studies have reported the beneficial effects of anti-platelet drugs in cardioprotection against ischaemia-reperfusion injuries. To date, no studies have focused on the indirect cytoprotective effects of ticagrelor via adenosine receptor on the endothelium. METHOD By evaluating cell viability and cleaved caspase 3 expression, we validated a model of endothelial cell apoptosis induced by hypoxia. In hypoxic endothelial cells treated with ticagrelor, we quantified the extracellular concentration of adenosine, and then we studied the involvement of adenosine pathways in the cytoprotective effect of ticagrelor. RESULTS Our results showed that 10 µM ticagrelor induced an anti-apoptotic effect in our model associated with an increase of extracellular adenosine concentration. Similar experiments were conducted with cangrelor but did not demonstrate an anti-apoptotic effect. We also found that A2B and A3 adenosine receptors were involved in the anti-apoptotic effect of ticagrelor in endothelial cells exposed to 2 h of hypoxia stress. CONCLUSION we described an endothelial cytoprotective mechanism of ticagrelor against hypoxia stress, independent of blood elements. We highlighted a mechanism triggered mainly by the increased extracellular bioavailability of adenosine, which activates A2B and A3 receptors on the endothelium.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Hélène Peyret
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Sylvie Brassart-Pasco
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims University Hospital, SFR CAP-santé, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France;
| | - Floriane Oszust
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Gaël Poitevin
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (G.P.); (P.N.)
| | - Philippe Nguyen
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (G.P.); (P.N.)
| | - Hervé Millart
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Zoubir Djerada
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
- Correspondence: ; Tel.: +33-3-26-83-27-82; Fax: +33-3-26-78-84-56
| |
Collapse
|
19
|
Xin M, Feng J, Hao Y, You J, Wang X, Yin X, Shang P, Ma D. Cyclic adenosine monophosphate in acute ischemic stroke: some to update, more to explore. J Neurol Sci 2020; 413:116775. [PMID: 32197118 DOI: 10.1016/j.jns.2020.116775] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/14/2022]
Abstract
The development of effective treatment for ischemic stroke, which is a common cause of morbidity and mortality worldwide, remains an unmet goal because the current first-line treatment management interventional therapy has a strict time window and serious complications. In recent years, a growing body of evidence has shown that the elevation of intracellular and extracellular cyclic adenosine monophosphate (cAMP) alleviates brain damage after ischemic stroke by attenuating neuroinflammation in the central nervous system and peripheral immune system. In the central nervous system, upregulated intracellular cAMP signaling can alleviate immune-mediated damage by restoring neuronal morphology and function, inhibiting microglia migration and activation, stabilizing the membrane potential of astrocytes and improving the cellular functions of endothelial cells and oligodendrocytes. Enhancement of the extracellular cAMP signaling pathway can improve neurological function by activating the cAMP-adenosine pathway to reduce immune-mediated damage. In the peripheral immune system, cAMP can act on various immune cells to suppress peripheral immune function, which can alleviate the inflammatory response in the central nervous system and improve the prognosis of acute cerebral ischemic injury. Therefore, cAMP may play key roles in reducing post-stroke neuroinflammatory damage. The protective roles of the cAMP indicate that the cAMP enhancing drugs such as cAMP supplements, phosphodiesterase inhibitors, adenylate cyclase agonists, which are currently used in the treatment of heart and lung diseases. They are potentially able to be applied as a new therapeutic strategy in ischemic stroke. This review focuses on the immune-regulating roles and the clinical implication of cAMP in acute ischemic stroke.
Collapse
Affiliation(s)
- Meiying Xin
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China.
| | - Yulei Hao
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Jiulin You
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Xiang Yin
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Pei Shang
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China
| | - Di Ma
- Department of Neurology, Jilin University First Hospital, Changchun, Jilin, China.
| |
Collapse
|
20
|
The role of purinergic P2Y 12 and P2Y 13 receptors in ADPβS-induced inhibition of the cardioaccelerator sympathetic drive in pithed rats. Purinergic Signal 2020; 16:73-84. [PMID: 32067141 DOI: 10.1007/s11302-020-09689-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/23/2020] [Indexed: 11/27/2022] Open
Abstract
ATP is a cotransmitter released with other neurotransmitters from sympathetic nerves, where it stimulates purinergic receptors. Purinergic adenosine P1 receptors (coupled to Gi/o proteins) produce sympatho-inhibition in several autonomic effectors by prejunctional inhibition of neurotransmitter release. Similarly, signalling through P2Y12 and P2Y13 receptors coupled to Gi/o proteins is initiated by the ATP breakdown product ADP. Hence, this study has pharmacologically investigated a possible role of ADP-induced inhibition of the cardioaccelerator sympathetic drive in pithed rats, using a stable ADP analogue (ADPβS) and selective antagonists for the purinergic P2Y1, P2Y12 and P2Y13 receptors. Accordingly, male Wistar rats were pithed and: (i) pretreated i.v. with gallamine (25 mg/kg) and desipramine (50 μg/kg) for preganglionic spinal (C7-T1) stimulation of the cardioaccelerator sympathetic drive (n = 78); or (ii) prepared for receiving i.v. injections of exogenous noradrenaline (n = 12). The i.v. continuous infusions of ADPβS (10 and 30 μg/kg/min) dose-dependently inhibited the tachycardic responses to electrical sympathetic stimulation, but not those to exogenous noradrenaline. The cardiac sympatho-inhibition produced by 30 μg/kg/min ADPβS was (after i.v. administration of compounds) (i) unchanged by 1-ml/kg bidistilled water or 300-μg/kg MRS 2500 (P2Y1 receptor antagonist), (ii) abolished by 300-μg/kg PSB 0739 (P2Y12 receptor antagonist) and (iii) partially blocked by 3000-μg/kg MRS 2211 (P2Y13 receptor antagonist). Our results suggest that ADPβS induces a cardiac sympatho-inhibition that mainly involves the P2Y12 receptor subtype and, probably to a lesser extent, the P2Y13 receptor subtype. These receptors may represent therapeutic targets for treating cardiovascular pathologies, including stroke and myocardial infarctions.
Collapse
|