1
|
Buja LM. Pathobiology of myocardial and cardiomyocyte injury in ischemic heart disease: Perspective from seventy years of cell injury research. Exp Mol Pathol 2024; 140:104944. [PMID: 39577392 DOI: 10.1016/j.yexmp.2024.104944] [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: 06/10/2024] [Revised: 10/11/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
This review presents a perspective on the pathobiology of acute myocardial infarction, a major manifestation of ischemic heart disease, and related mechanisms of ischemic and toxic cardiomyocyte injury, based on advances and insights that have accrued over the last seventy years, including my sixty years of involvement in the field as a physician-scientist-pathologist. This analysis is based on integration of my research within the broader context of research in the field. A particular focus has been on direct measurements in cardiomyocytes of electrolyte content by electron probe X-ray microanalysis (EPXMA) and Ca2+ fluxes by fura-2 microspectrofluorometry. These studies established that increased intracellular Ca2+ develops at a transitional stage in the progression of cardiomyocyte injury in association with ATP depletion, other electrolyte alterations, altered cell volume regulation, and altered membrane phospholipid composition. Subsequent increase in total calcium with mitochondrial calcium accumulation can occur. These alterations are characteristic of oncosis, which is an initial pre-lethal state of cell injury with cell swelling due to cell membrane dysfunction in ATP depleted cells; oncosis rapidly progresses to necrosis/necroptosis with physical disruption of the cell membrane, unless the adverse stimulus is rapidly reversed. The observed sequential changes fit a three-stage model of membrane injury leading to irreversible cell injury. The data establish oncosis as the primary mode of cardiomyocyte injury in evolving myocardial infarcts. Oncosis also has been documented to be the typical form of non-ischemic cell injury due to toxins. Cardiomyocytes with less energy impairment have the capability of undergoing apoptosis and autophagic death as well as oncosis, as is seen in pathological remodeling in chronic heart failure. Work is ongoing to apply the insights from experimental studies to better understand and ameliorate myocardial ischemia and reperfusion injury in patients. The perspective and insights in this review are derived from basic principles of pathology, an integrative discipline focused on mechanisms of disease affecting the cell, the organizing unit of living organisms.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, United States of America.
| |
Collapse
|
2
|
Beccacece L, Abondio P, Bini C, Pelotti S, Luiselli D. The Link between Prostanoids and Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24044193. [PMID: 36835616 PMCID: PMC9962914 DOI: 10.3390/ijms24044193] [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: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global deaths, and many risk factors contribute to their pathogenesis. In this context, prostanoids, which derive from arachidonic acid, have attracted attention for their involvement in cardiovascular homeostasis and inflammatory processes. Prostanoids are the target of several drugs, but it has been shown that some of them increase the risk of thrombosis. Overall, many studies have shown that prostanoids are tightly associated with cardiovascular diseases and that several polymorphisms in genes involved in their synthesis and function increase the risk of developing these pathologies. In this review, we focus on molecular mechanisms linking prostanoids to cardiovascular diseases and we provide an overview of genetic polymorphisms that increase the risk for cardiovascular disease.
Collapse
Affiliation(s)
- Livia Beccacece
- Computational Genomics Lab, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Paolo Abondio
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
- Correspondence: (L.B.); (P.A.)
| | - Carla Bini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Donata Luiselli
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
| |
Collapse
|
3
|
Zhang J, Hu Y, Wang H, Hou J, Xiao W, Wen X, Wang T, Long P, Jiang H, Wang Z, Liu H, Chen X. Advances in research on the protective mechanisms of traditional Chinese medicine (TCM) in myocardial ischaemia-reperfusion injury. PHARMACEUTICAL BIOLOGY 2022; 60:931-948. [PMID: 35587352 PMCID: PMC9132412 DOI: 10.1080/13880209.2022.2063342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Developing effective drugs to treat myocardial ischaemia-reperfusion (MI/R) injury is imperative. Traditional Chinese medicines (TCMs) have had considerable success in the treatment of cardiovascular diseases. Elucidating the mechanisms by which TCMs improve MI/R injury can supplement the literature on MI/R prevention and treatment. OBJECTIVE To summarise TCMs and their main protective mechanisms against MI/R injury reported over the past 40 years. METHODS Relevant literature published between 1980 and 2020 in Chinese and English was retrieved from the Web of Science, PubMed, SpringerLink, PubMed Central, Scopus, and Chinese National Knowledge Infrastructure (CNKI) databases. Search terms included 'medicinal plants', 'myocardial ischaemia reperfusion injury', 'Chinese medicine prescriptions', 'mechanisms', 'prevention', 'treatment' and 'protection'. For inclusion in the analysis, medicinal plants had to be searchable in the China Medical Information Platform and Plant Database. RESULTS We found 71 medicinal species (from 40 families) that have been used to prevent MI/R injury, of which Compositae species (8 species) and Leguminosae species (7 species) made up the majority. Most of the effects associated with these plants are described as antioxidant and anti-inflammatory. Furthermore, we summarised 18 kinds of Chinese compound prescriptions, including the compound Danshen tablet and Baoxin pill, which mainly reduce oxidative stress and regulate mitochondrial energy metabolism. DISCUSSION AND CONCLUSIONS We summarised TCMs that protect against MI/R injury and their pharmacological mechanisms. This in-depth explanation of the roles of TCMs in MI/R injury protection provides a theoretical basis for the research and development of TCM-based treatment drugs.
Collapse
Affiliation(s)
- Jiexin Zhang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yonghe Hu
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Han Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wenjing Xiao
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology, The First People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Tingting Wang
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Pan Long
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hezhong Jiang
- Faculty of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhanhao Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Huawei Liu
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Huang M, Zhang X, Yan W, Liu J, Wang H. Metabolomics reveals potential plateau adaptability by regulating
inflammatory response and oxidative stress-related metabolism and energy
metabolism pathways in yak. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 64:97-109. [PMID: 35174345 PMCID: PMC8819316 DOI: 10.5187/jast.2021.e129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/14/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
Species are facing strong selection pressures to adapt to inhospitable
high-altitude environments. Yaks are a valuable species and an iconic symbol of
the Qinghai-Tibet Plateau. Extensive studies of high-altitude adaptation have
been conducted, but few have focused on metabolism. In the present study, we
determined the differences in the serum metabolomics between yaks and the
closely related species of low-altitude yellow cattle and dairy cows. We
generated high-quality metabolite profiling data for 36 samples derived from the
three species, and a clear separation trend was obtained between yaks and the
other animals from principal component analysis. In addition, we identified a
total of 63 differentially expressed metabolites among the three species.
Functional analysis revealed that differentially expressed metabolites were
related to the innate immune activation, oxidative stress-related metabolism,
and energy metabolism in yaks, which indicates the important roles of
metabolites in high-altitude adaptation in yaks. The results provide new
insights into the mechanism of adaptation or acclimatization to high-altitude
environments in yaks and hypoxia-related diseases in humans.
Collapse
Affiliation(s)
- Meizhou Huang
- Department of Toxicology, School of Public
Health, Lanzhou University, Gansu 730000, China
- Academician (Expert) Workstation of
Sichuan Province, The Affiliated Hospital of Southwest Medical
University, Sichuan 646000, China
| | - Xin Zhang
- Department of Toxicology, School of Public
Health, Lanzhou University, Gansu 730000, China
| | - Wenjun Yan
- Agricultural and Rural Integrated Service
Center of Dachaigou Town, Tianzhu Tibetan Autonomous County,
Gansu 733202, China
| | - Jingjing Liu
- Department of Toxicology, School of Public
Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public
Health, Lanzhou University, Gansu 730000, China
- Corresponding author: Hui Wang, Department of
Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China.
Tel: +86-13919330832, E-mail:
| |
Collapse
|
5
|
Pils V, Terlecki-Zaniewicz L, Schosserer M, Grillari J, Lämmermann I. The role of lipid-based signalling in wound healing and senescence. Mech Ageing Dev 2021; 198:111527. [PMID: 34174292 DOI: 10.1016/j.mad.2021.111527] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Lipid-based signalling modulates several cellular processes and intercellular communication during wound healing and tissue regeneration. Bioactive lipids include but are not limited to the diverse group of eicosanoids, phospholipids, and extracellular vesicles and mediate the attraction of immune cells, initiation of inflammatory responses, and their resolution. In aged individuals, wound healing and tissue regeneration are greatly impaired, resulting in a delayed healing process and non-healing wounds. Senescent cells accumulate with age in vivo, preferably at sites implicated in age-associated pathologies and their elimination was shown to alleviate many age-associated diseases and disorders. In contrast to these findings, the transient presence of senescent cells in the process of wound healing exerts beneficial effects and limits fibrosis. Hence, clearance of senescent cells during wound healing was repeatedly shown to delay wound closure in vivo. Recent findings established a dysregulated synthesis of eicosanoids, phospholipids and extracellular vesicles as part of the senescent phenotype. This intriguing connection between cellular senescence, lipid-based signalling, and the process of wound healing and tissue regeneration prompts us to compile the current knowledge in this review and propose future directions for investigation.
Collapse
Affiliation(s)
- Vera Pils
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence - SKINMAGINE, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
6
|
Mechanistic insight on the role of leukotriene receptors in ischemic-reperfusion injury. Pharmacol Rep 2021; 73:1240-1254. [PMID: 33818747 DOI: 10.1007/s43440-021-00258-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Leukotrienes (LT) are a class of inflammatory mediators produced by the 5-lipoxygenase (5-LO) enzyme from arachidonic acid (AA). We discussed the various LT inhibitors and downstream pathway modulators, such as Mitogen-Activated Protein Kinases (MAPK), Phosphatidylinositol 3-Kinase/Protein Kinase B (PI3K/Akt), 5'-Adenosine Monophosphate-Activated Protein Kinase (AMPK), Protein Kinase C (PKC), Nitric Oxide (NO), Bradykinin, Early Growth Response-1 (Egr-1), Nuclear Factor-κB (NF-κB), and Tumor Necrosis Factor-Alpha (TNF-α), which in turn regulate various metabolic and physiological processes involving I/R injury. A systematic literature review of Bentham, Scopus, PubMed, Medline, and EMBASE (Elsevier) databases was carried out to understand the nature and mechanistic interventions of the leukotriene receptor modulations in ischemic injury. In the pathophysiology of I/R injuries, LT has been found to play an important role. I/R injury affects most of the vital organs and is characterized by inflammation, oxidative stress, cell death, and apoptosis leading to morbidity and mortality. sThis present review focuses on the various LT receptors, i.e., CysLT, LTC4, LTD4, and LTE4, involved in developing I/R injury in organs, such as the brain, spinal cord, heart, kidney, liver, and intestine.
Collapse
|
7
|
Kishi H, Yamaguchi K, Watanabe K, Nakamura K, Fujioka D, Kugiyama K. Deficiency of Phospholipase A 2 Receptor Exacerbates Autoimmune Myocarditis in Mice. Inflammation 2021; 43:1097-1109. [PMID: 32016628 DOI: 10.1007/s10753-020-01195-z] [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] [Indexed: 01/09/2023]
Abstract
Secretory phospholipase A2 (sPLA2) plays a critical role in the pathogenesis of various inflammatory diseases through production of pro-inflammatory eicosanoids. PLA2 receptor 1 (PLA2R) acts as a clearance receptor for sPLA2s. This study examined whether PLA2R plays a role in the pathogenesis of experimental autoimmune myocarditis using PLA2R-deficient (PLA2R KO) mice on a BALB/c background. Autoimmune myocarditis was induced by immunization with murine α-myosin heavy chain. In the immunostaining of PLA2R wild-type (WT) myocardium, PLA2R and sPLA2s were expressed in α-SMA+ cells and neutrophils, respectively. In immunoblot analyses, tissue from PLA2R KO myocardium after immunization had five to tenfold increases in the protein level of sPLA2-IB and sPLA2-IIA compared with PLA2R WT myocardium. However, the mRNA expression levels of these sPLA2s were similar in PLA2R KO and WT myocardium. Compared with PLA2R WT myocardium, PLA2R KO myocardium after immunization showed 40% increase in areas affected by infiltration of inflammatory cells, eight to tenfold increase in levels of PGE2 and TXB2, and a threefold increase in number of Th17 cells in heart infiltrates assessed by flow cytometric analysis. Finally, PGE2 promoted IL-23-induced expansion of Th17 cells in vitro. In conclusion, PLA2R-deficiency increased sPLA2-IB and sPLA2-IIA levels in the myocardium after immunization probably through impaired clearance, leading to increased levels of PGE2 in the myocardium. Elevated PGE2 induced Th17 cell expansion, exacerbating myocarditis in PLA2R KO mice. Thus, PLA2R plays an important role in pathogenesis of experimental autoimmune myocarditis.
Collapse
Affiliation(s)
- Hiroki Kishi
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kazuyuki Yamaguchi
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kazuhiro Watanabe
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kazuto Nakamura
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Daisuke Fujioka
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kiyotaka Kugiyama
- Department of Internal Medicine II, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan. .,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
8
|
Chang Y, Xing L, Zhou W, Zhang W. Up-regulating microRNA-138-5p enhances the protective role of dexmedetomidine on myocardial ischemia-reperfusion injury mice via down-regulating Ltb4r1. Cell Cycle 2021; 20:445-458. [PMID: 33509010 DOI: 10.1080/15384101.2021.1878330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Both microRNAs (miRs) and dexmedetomidine (Dex) have been verified to exert functional roles in myocardial ischemia-reperfusion injury (MI/RI). Given that, we concretely aim to discuss the effects of Dex and miR-138-5p on ventricular remodeling in mice affected by MI/RI via mediating leukotriene B4 receptor 1 (Ltb4r1). MI/RI mouse model was established by ligating left anterior descending coronary artery. The cardiac function, inflammatory factors and collagen fiber contents were detected after Dex/miR-138-5p/Ltb4r1 treatment. MiR-138-5p and Ltb4r1 expression in myocardial tissues were tested by RT-qPCR and western blot assay. The target relationship between miR-138-5p and Ltb4r1 was verified by online software prediction and luciferase activity assay. MiR-138-5p was down-regulated while Ltb4r1 was up-regulated in myocardial tissues of MI/RI mice. Dex improved cardiac function, alleviated myocardial damage, reduced inflammatory factor contents, collagen fibers, and Ltb4r1 expression while increased miR-138-5p expression in myocardial tissues of mice with MI/RI. Restored miR-138-5p and depleted Ltb4r1 improved cardiac function, abated inflammatory factor contents, myocardial damage, and content of collagen fibers in MI/RI mice. MiR-138-5p directly targeted Ltb4r1. The work evidence that Dex could ameliorate ventricular remodeling of MI/RI mice by up-regulating miR-138-3p and down-regulating Ltb4r1. Thus, Dex and miR-138-3p/Ltb4r1 may serve as potential targets for the ventricular remodeling of MI/RI.
Collapse
Affiliation(s)
- Yanzi Chang
- Department of Anesthesiology, Attending Doctor, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Lika Xing
- Department of Anesthesiology, Attending Doctor, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Wenjuan Zhou
- Department of Anesthesiology, Attending Doctor, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| | - Wei Zhang
- Department of Anesthesiology, Chief Physician, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, China
| |
Collapse
|
9
|
Dissecting Cellular Mechanisms of Long-Chain Acylcarnitines-Driven Cardiotoxicity: Disturbance of Calcium Homeostasis, Activation of Ca 2+-Dependent Phospholipases, and Mitochondrial Energetics Collapse. Int J Mol Sci 2020; 21:ijms21207461. [PMID: 33050414 PMCID: PMC7589681 DOI: 10.3390/ijms21207461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023] Open
Abstract
Long-chain acylcarnitines (LCAC) are implicated in ischemia-reperfusion (I/R)-induced myocardial injury and mitochondrial dysfunction. Yet, molecular mechanisms underlying involvement of LCAC in cardiac injury are not sufficiently studied. It is known that in cardiomyocytes, palmitoylcarnitine (PC) can induce cytosolic Ca2+ accumulation, implicating L-type calcium channels, Na+/Ca2+ exchanger, and Ca2+-release from sarcoplasmic reticulum (SR). Alternatively, PC can evoke dissipation of mitochondrial potential (ΔΨm) and mitochondrial permeability transition pore (mPTP). Here, to dissect the complex nature of PC action on Ca2+ homeostasis and oxidative phosphorylation (OXPHOS) in cardiomyocytes and mitochondria, the methods of fluorescent microscopy, perforated path-clamp, and mitochondrial assays were used. We found that LCAC in dose-dependent manner can evoke Ca2+-sparks and oscillations, long-living Ca2+ enriched microdomains, and, finally, Ca2+ overload leading to hypercontracture and cardiomyocyte death. Collectively, PC-driven cardiotoxicity involves: (I) redistribution of Ca2+ from SR to mitochondria with minimal contribution of external calcium influx; (II) irreversible inhibition of Krebs cycle and OXPHOS underlying limited mitochondrial Ca2+ buffering; (III) induction of mPTP reinforced by PC-calcium interplay; (IV) activation of Ca2+-dependent phospholipases cPLA2 and PLC. Based on the inhibitory analysis we may suggest that simultaneous inhibition of both phospholipases could be an effective strategy for protection against PC-mediated toxicity in cardiomyocytes.
Collapse
|
10
|
Involvement of the Endothelin Receptor Type A in the Cardiovascular Inflammatory Response Following Scorpion Envenomation. Toxins (Basel) 2020; 12:toxins12060389. [PMID: 32545475 PMCID: PMC7374423 DOI: 10.3390/toxins12060389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023] Open
Abstract
Elevated levels of endothelin-1 (ET-1) were recorded in sera of scorpion sting patients. However, no studies focused on the mechanism of ET-1 involvement in the pathogenesis of scorpion envenomation, particularly in the cardiovascular system which is seriously affected in severe cases of scorpion stings. Inflammation induced by Androctonus australis hector (Aah) scorpion venom in the heart together with the aorta was studied in mice pretreated with a specific endothelin A receptor (ETA-R) inhibitor. ETA-R inhibition resulted in the attenuation of the high amounts of cytokine (tumor necrosis factor alpha (TNF-α) and interleukin-17 (IL-17)) recorded in the sera of envenomed mice. The recovery of the oxidative stress marker balance and matrix metalloproteinase (MMP) expression were also observed, concomitantly with the reduction of tissular neutrophil infiltration. Additionally, the cardiac and the aortic tissue alterations, and the metabolic enzymes (creatine kinase (CK) and muscle–brain isoform creatine kinase (CK-MB)) overspread into sera were significantly attenuated. Obtained results suggest the implication of endothelin throughout its ETA receptors in the inflammatory response observed in the cardiovascular components during scorpion envenomation. Further knowledge is needed to better understand the implication of the endothelin axis and to improve the therapeutic management of severe scorpion sting cases.
Collapse
|
11
|
Xiong TY, Liu C, Liao YB, Zheng W, Li YJ, Li X, Ou Y, Wang ZJ, Wang X, Li CM, Zhao ZG, Feng Y, Liu XJ, Chen M. Differences in metabolic profiles between bicuspid and tricuspid aortic stenosis in the setting of transcatheter aortic valve replacement. BMC Cardiovasc Disord 2020; 20:229. [PMID: 32423380 PMCID: PMC7236099 DOI: 10.1186/s12872-020-01491-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/19/2020] [Indexed: 02/08/2023] Open
Abstract
Background To explore why bicuspid aortic stenosis has certain clinical differences from the tricuspid morphology, we evaluated the metabolomics profile involved in bicuspid aortic valve (BAV) aortic stenosis prior to and after transcatheter aortic valve replacement (TAVR) in comparison with tricuspid aortic valve (TAV). Methods In this TAVR cohort with prospectively collected data, blood samples were obtained before TAVR valve deployment and at the 7th day after TAVR, which were then sent for liquid and gas chromatography-mass spectrometry detection. Besides comparisons between BAV and TAV, BAV patients were also divided in subgroups according to baseline hemodynamics (i.e. maximal transaortic velocity, Vmax) and post-procedural reverse left ventricular (LV) remodeling (i.e. the change in LV mass index from baseline, ∆LVMI) for further analysis. Metabolic differences between groups were identified by integrating univariate test, multivariate analysis and weighted correlation network analysis algorithm. Results A total of 57 patients were enrolled including 33 BAV patients. The BAV group showed lower arginine and proline metabolism both before and post TAVR than TAV represented by decreased expression of L-Glutamine. In BAV subgroup analysis, patients with baseline Vmax > 5 m/s (n = 11) or the 4th quartile of change in ∆LVMI at one-year follow-up (i.e. poorly-recovered LV, n = 8) showed elevated arachidonic acid metabolism compared with Vmax < 4.5 m/s (n = 12) or the 1st quartile of ∆LVMI (i.e. well-recovered LV, n = 8) respectively. Conclusions Difference in arginine and proline metabolism was identified between BAV and TAV in TAVR recipients. Elevated arachidonic acid metabolism may reflect more severe baseline hemodynamics and worse LV reserve remodeling after TAVR in BAV.
Collapse
Affiliation(s)
- Tian-Yuan Xiong
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Chang Liu
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Yan-Biao Liao
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Wen Zheng
- Laboratory of Mitochondrial Biology, West China-Washington Mitochondria and Metabolism Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yi-Jian Li
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Xi Li
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Yuanweixiang Ou
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Zi-Jie Wang
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Xi Wang
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Chang-Ming Li
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Zhen-Gang Zhao
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Yuan Feng
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China
| | - Xiao-Jing Liu
- Laboratory of Mitochondrial Biology, West China-Washington Mitochondria and Metabolism Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China. .,Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, 610041, Chengdu, People's Republic of China.
| | - Mao Chen
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guo Xue Alley, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
12
|
Zhi W, Li K, Wang H, Lei M, Guo Y. Melatonin elicits protective effects on OGD/R‑insulted H9c2 cells by activating PGC‑1α/Nrf2 signaling. Int J Mol Med 2020; 45:1294-1304. [PMID: 32323734 PMCID: PMC7138270 DOI: 10.3892/ijmm.2020.4514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Melatonin (Mel) elicits beneficial effects on myocardial ischemia/reperfusion injury. However, the underlying mechanism of Mel against oxygen-glucose deprivation/ reperfusion (OGD/R)-induced H9c2 cardiomyocyte damage remains largely unknown. The aim of the present study was to investigate the biological roles and the potential mechanisms of Mel in OGD/R-exposed H9c2 cardiomyocytes. The results of the present study demonstrated that Mel significantly elevated the viability and reduced the activity of lactate dehydrogenase and creatine kinase myocardial band in a doseand time-dependent manner in OGD/R-insulted H9c2 cells. In addition, Mel suppressed OGD/R-induced oxidative stress in H9c2 cells, as demonstrated by the decreased reactive oxygen species and malondialdehyde levels, as well as the increased activities of superoxide dismutase, catalase and glutathione peroxidase. Mel exerted an antioxidant effect by activating the peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Mel reduced the expression of OGD/R-enhanced pro-inflammatory tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1β, IL-8 and monocyte chemotactic protein-1. Mel also abolished the OGD/R-induced increase in H9c2 apoptosis, as evidenced by mitochondrial membrane potential restoration and caspase-3 and caspase-9 inactivation, as well as the upregulation of Bcl-2 and down-regulation of cleaved caspase-3 and Bax. The Mel-induced antiapoptotic effects were dependent on PGC-1α/TNF-α signaling. Overall, the results of the present study demonstrated that Mel alleviated OGD/R-induced H9c2 cell injury via the inhibition of oxidative stress and inflammation by regulating the PGC-1α/Nrf2 and PGC-1α/TNF-α signaling pathways, suggesting a promising role for Mel in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Weiwei Zhi
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Kai Li
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Hongbing Wang
- Department of Cardiology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Ming Lei
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Yingqiang Guo
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| |
Collapse
|
13
|
Hoff U, Bubalo G, Fechner M, Blum M, Zhu Y, Pohlmann A, Hentschel J, Arakelyan K, Seeliger E, Flemming B, Gürgen D, Rothe M, Niendorf T, Manthati VL, Falck JR, Haase M, Schunck W, Dragun D. A synthetic epoxyeicosatrienoic acid analogue prevents the initiation of ischemic acute kidney injury. Acta Physiol (Oxf) 2019; 227:e13297. [PMID: 31077555 DOI: 10.1111/apha.13297] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
AIM Imbalances in cytochrome P450 (CYP)-dependent eicosanoid formation may play a central role in ischemic acute kidney injury (AKI). We reported previously that inhibition of 20-hydroxyeicosatetraenoic acid (20-HETE) action ameliorated ischemia/reperfusion (I/R)-induced AKI in rats. Now we tested the hypothesis that enhancement of epoxyeicosatrienoic acid (EET) actions may counteract the detrimental effects of 20-HETE and prevent the initiation of AKI. METHODS Male Lewis rats underwent right nephrectomy and ischemia was induced by 45 min clamping of the left renal pedicle followed by up to 48 h of reperfusion. Circulating CYP-eicosanoid profiles were compared in patients who underwent cardiac surgery with (n = 21) and without (n = 38) developing postoperative AKI. RESULTS Ischemia induced an about eightfold increase of renal 20-HETE levels, whereas free EETs were not accumulated. To compensate for this imbalance, a synthetic 14,15-EET analogue was administered by intrarenal infusion before ischemia. The EET analogue improved renal reoxygenation as monitored by in vivo parametric MRI during the initial 2 h reperfusion phase. The EET analogue improved PI3K- as well as mTORC2-dependent rephosphorylation of Akt, induced inactivation of GSK-3β, reduced the development of tubular apoptosis and attenuated inflammatory cell infiltration. The EET analogue also significantly alleviated the I/R-induced drop in creatinine clearance. Patients developing postoperative AKI featured increased preoperative 20-HETE and 8,9-EET levels. CONCLUSIONS Pharmacological interventions targeting the CYP-eicosanoid pathway could offer promising new options for AKI prevention. Individual differences in CYP-eicosanoid formation may contribute to the risk of developing AKI in clinical settings.
Collapse
Affiliation(s)
- Uwe Hoff
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
| | - Gordana Bubalo
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
| | - Mandy Fechner
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
| | | | - Ye Zhu
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
- Department of Nephrology The Fifth Affiliated Hospital of Sun Yat‐sun University Zhuhai China
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.) Max Delbrueck Center for Molecular Medicine Berlin Germany
| | - Jan Hentschel
- Berlin Ultrahigh Field Facility (B.U.F.F.) Max Delbrueck Center for Molecular Medicine Berlin Germany
| | - Karen Arakelyan
- Berlin Ultrahigh Field Facility (B.U.F.F.) Max Delbrueck Center for Molecular Medicine Berlin Germany
- Center for Cardiovascular Research, Institute of Physiology Charité‐Universitätsmedizin Berlin Berlin Germany
| | - Erdmann Seeliger
- Center for Cardiovascular Research, Institute of Physiology Charité‐Universitätsmedizin Berlin Berlin Germany
| | - Bert Flemming
- Center for Cardiovascular Research, Institute of Physiology Charité‐Universitätsmedizin Berlin Berlin Germany
| | - Dennis Gürgen
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
| | | | - Thoralf Niendorf
- Max Delbrueck Center for Molecular Medicine Berlin Germany
- Berlin Ultrahigh Field Facility (B.U.F.F.) Max Delbrueck Center for Molecular Medicine Berlin Germany
| | | | - John R. Falck
- Biochemistry Department UT Southwestern Dallas Texas
| | - Michael Haase
- Medical Faculty Otto‐von‐Guericke University Magdeburg Germany
- Diaverum Deutschland Potsdam Germany
| | | | - Duska Dragun
- Nephrology and Intensive Care Medicine, Center for Cardiovascular Research Charité‐Universitätsmedizin Berlin Berlin Germany
| |
Collapse
|
14
|
Ansari MN, Ganaie MA, Rehman NU, Alharthy KM, Khan TH, Imam F, Ansari MA, Al-Harbi NO, Jan BL, Sheikh IA, Hamad AM. Protective role of Roflumilast against cadmium-induced cardiotoxicity through inhibition of oxidative stress and NF-κB signaling in rats. Saudi Pharm J 2019; 27:673-681. [PMID: 31297022 PMCID: PMC6598217 DOI: 10.1016/j.jsps.2019.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022] Open
Abstract
Cadmium (Cd), a potent cardiotoxic environmental heavy metal, induces oxidative stress and membrane disturbances in cardiac myocytes. Phosphodiesterase (PDEs) retards the positive inotropic effects of β-adrenoceptor activation by decreasing levels of cAMP via degradation. Hence, PDE inhibitors sensitize the heart to catecholamine and are therefore, used as positive inotropic agents. The present study was designed to probe the potential attenuating effects of the selective PDE4 inhibitor (Roflumilast, ROF), on cardiac biomarkers, lipid profile, lipid peroxidation products, antioxidant status and histology of cardiac tissues against Cd-induced cardiotoxicity in rats. Rats were randomly distributed into four different groups: group 1, served as the normal control group. Group 2, served as the toxic control group and were administered Cd (3 mg/kg, i.p.) for next 7 days. Groups 3 and 4, served as treatment groups that received Cd with concomitant oral administration of ROF doses (0.5 and 1.5 mg/kg), respectively for 7 days. Serum samples of toxic control group rats resulted in significant (P < 0.001) increase in lactate dehydrogenase (LDH), creatine phosphokinase (CPK), total cholesterol (TC), triglycerides (TG) and low density lipoproteins (LDL) levels with concomitant decrease in high density lipoproteins (HDL) levels in serum which were found reversed with both of ROF treatment groups. Cd also causes significant increased (P < 0.001) in myocardial malondialdehyde (MDA) contents while cardiac glutathione (GSH) level, superoxide dismutase (SOD) and catalase (CAT) enzyme activities were found decreased whereas both doses of ROF, significantly reversed these oxidative stress markers and antioxidant enzymes. Cardiotoxicity induced by Cd also resulted in enhanced expression of non-phosphorylated and phosphorylated form of NF-κB p65 and decreased expression of glutathione-S-transferase (GST) and NQO1 which were found reversed with ROF treatments, comparable to normal control group. Histopathological changes were also improved by ROF administration as compared to Cd treated rats alone. In conclusion, Roflumilast exhibited attenuating effect against Cd-induced cardiac toxicity.
Collapse
Affiliation(s)
- Mohd Nazam Ansari
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Majid A. Ganaie
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Najeeb Ur Rehman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Khalid M. Alharthy
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Tajdar H. Khan
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A. Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Basit L. Jan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ishfaq A. Sheikh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abubaker M. Hamad
- Department of Basic Sciences, Preparatory Year Deanship, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Histopathology and Cytopathology, Faculty of Medical Laboratory Sciences, University of Gezira, Wad Madani, Sudan
| |
Collapse
|
15
|
Keck M, Flamant M, Mougenot N, Favier S, Atassi F, Barbier C, Nadaud S, Lompré AM, Hulot JS, Pavoine C. Cardiac inflammatory CD11b/c cells exert a protective role in hypertrophied cardiomyocyte by promoting TNFR 2- and Orai3- dependent signaling. Sci Rep 2019; 9:6047. [PMID: 30988334 PMCID: PMC6465256 DOI: 10.1038/s41598-019-42452-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/29/2019] [Indexed: 01/04/2023] Open
Abstract
Early adaptive cardiac hypertrophy (EACH) is initially a compensatory process to optimize pump function. We reported the emergence of Orai3 activity during EACH. This study aimed to characterize how inflammation regulates store-independent activation of Orai3-calcium influx and to evaluate the functional role of this influx. Isoproterenol infusion or abdominal aortic banding triggered EACH. TNFα or conditioned medium from cardiac CD11b/c cells activated either in vivo [isolated from rats displaying EACH], or in vitro [isolated from normal rats and activated with lipopolysaccharide], were added to adult cardiomyocytes before measuring calcium entry, cell hypertrophy and cell injury. Using intramyocardial injection of siRNA, Orai3 was in vivo knockdown during EACH to evaluate its protective activity in heart failure. Inflammatory CD11b/c cells trigger a store-independent calcium influx in hypertrophied cardiomyocytes, that is mimicked by TNFα. Pharmacological or molecular (siRNA) approaches demonstrate that this calcium influx, depends on TNFR2, is Orai3-driven, and elicits cardiomyocyte hypertrophy and resistance to oxidative stress. Neutralization of Orai3 inhibits protective GSK3β phosphorylation, impairs EACH and accelerates heart failure. Orai3 exerts a pathophysiological protective impact in EACH promoting hypertrophy and resistance to oxidative stress. We highlight inflammation arising from CD11b/c cells as a potential trigger of TNFR2- and Orai3-dependent signaling pathways.
Collapse
Affiliation(s)
- Mathilde Keck
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Mathilde Flamant
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Nathalie Mougenot
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
- UMS28, plateforme PECMV, F-75013, Paris, France
| | - Sophie Favier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Fabrice Atassi
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Camille Barbier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Sophie Nadaud
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Anne-Marie Lompré
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Jean-Sébastien Hulot
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France
| | - Catherine Pavoine
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Team 3, F-75013, Paris, France.
| |
Collapse
|
16
|
Liu C, Liu B, Liu L, Zhang EL, Sun BD, Xu G, Chen J, Gao YQ. Arachidonic Acid Metabolism Pathway Is Not Only Dominant in Metabolic Modulation but Associated With Phenotypic Variation After Acute Hypoxia Exposure. Front Physiol 2018; 9:236. [PMID: 29615930 PMCID: PMC5864929 DOI: 10.3389/fphys.2018.00236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Background: The modulation of arachidonic acid (AA) metabolism pathway is identified in metabolic alterations after hypoxia exposure, but its biological function is controversial. We aimed at integrating plasma metabolomic and transcriptomic approaches to systematically explore the roles of the AA metabolism pathway in response to acute hypoxia using an acute mountain sickness (AMS) model. Methods: Blood samples were obtained from 53 enrolled subjects before and after exposure to high altitude. Ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry and RNA sequencing were separately performed for metabolomic and transcriptomic profiling, respectively. Influential modules comprising essential metabolites and genes were identified by weighted gene co-expression network analysis (WGCNA) after integrating metabolic information with phenotypic and transcriptomic datasets, respectively. Results: Enrolled subjects exhibited diverse response manners to hypoxia. Combined with obviously altered heart rate, oxygen saturation, hemoglobin, and Lake Louise Score (LLS), metabolomic profiling detected that 36 metabolites were highly related to clinical features in hypoxia responses, out of which 27 were upregulated and nine were downregulated, and could be mapped to AA metabolism pathway significantly. Integrated analysis of metabolomic and transcriptomic data revealed that these dominant molecules showed remarkable association with genes in gas transport incapacitation and disorders of hemoglobin metabolism pathways, such as ALAS2, HEMGN. After detailed description of AA metabolism pathway, we found that the molecules of 15-d-PGJ2, PGA2, PGE2, 12-O-3-OH-LTB4, LTD4, LTE4 were significantly up-regulated after hypoxia stimuli, and increased in those with poor response manner to hypoxia particularly. Further analysis in another cohort showed that genes in AA metabolism pathway such as PTGES, PTGS1, GGT1, TBAS1 et al. were excessively elevated in subjects in maladaptation to hypoxia. Conclusion: This is the first study to construct the map of AA metabolism pathway in response to hypoxia and reveal the crosstalk between phenotypic variation under hypoxia and the AA metabolism pathway. These findings may improve our understanding of the advanced pathophysiological mechanisms in acute hypoxic diseases and provide new insights into critical roles of the AA metabolism pathway in the development and prevention of these diseases.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Bao Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,The 12th Hospital of Chinese People's Liberation Army, Kashi, China
| | - Lu Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Er-Long Zhang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Bind-da Sun
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Jian Chen
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Third Military Medical University, Chongqing, China.,Key Laboratory of High Altitude Environmental Medicine, Army Medical University, Third Military Medical University, Ministry of Education, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| |
Collapse
|
17
|
Shi P, Zhang L, Zhang M, Yang W, Wang K, Zhang J, Otsu K, Huang G, Fan X, Liu J. Platelet-Specific p38α Deficiency Improved Cardiac Function After Myocardial Infarction in Mice. Arterioscler Thromb Vasc Biol 2017; 37:e185-e196. [PMID: 28982666 DOI: 10.1161/atvbaha.117.309856] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE MAPKs (mitogen-activated protein kinases), especially p38, play detrimental roles in cardiac diseases and cardiac remodeling post-myocardial infarction. However, the activation and function of MAPKs in coronary thrombosis in vivo and its relationship with clinical outcomes remain poorly understood. APPROACH AND RESULTS Here, we showed that p38α was the major isoform expressed in human and mouse platelets. Platelet-specific p38α-deficient mice presented impaired thrombosis and hemostasis but had improved cardiac function, reduced infarct size, decreased inflammatory response, and microthrombus in a left anterior descending artery ligation model. Signaling analysis revealed that p38 activation was one of the earliest events in platelets after treatment with receptor agonists or reactive oxygen species. p38α/MAPK-activated protein kinase 2/heat shock protein 27 and p38α/cytosolic phospholipases A2 were the major pathways regulating receptor-mediated or hydrogen peroxide-induced platelet activation in an ischemic environment. Moreover, the distinct roles of ERK1/2 (extracellular signal-regulated kinase) in receptor- or reactive oxygen species-induced p38-mediated platelet activation reflected the complicated synergistic relationships among MAPKs. Analysis of clinical samples revealed that MAPKs were highly phosphorylated in platelets from preoperative patients with ST-segment-elevation myocardial infarction, and increased phosphorylation of p38 was associated with no-reflow outcomes. CONCLUSIONS We conclude that p38α serves as a critical regulator of platelet activation and potential indicator of highly thrombotic lesions and no-reflow, and inhibition of platelet p38α may improve clinical outcomes in subjects with ST-segment-elevation myocardial infarction.
Collapse
Affiliation(s)
- Panlai Shi
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.).
| | - Lin Zhang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.).
| | - Mingliang Zhang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.)
| | - Wenlong Yang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.)
| | - Kemin Wang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.)
| | - Junfeng Zhang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.)
| | - Kinya Otsu
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.)
| | - Gonghua Huang
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.).
| | - Xuemei Fan
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.).
| | - Junling Liu
- From the Department of Biochemistry and Molecular Cell Biology (P.S., L.Z., K.W., X.F., J.L.), Department of Cardiology, Ninth People's Hospital (M.Z., W.Y., J.Z.), and Shanghai Institute of Immunology (G.H.), Shanghai Jiao Tong University School of Medicine, China; and Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, United Kingdom (K.O.).
| |
Collapse
|
18
|
Míčová P, Klevstig M, Holzerová K, Vecka M, Žurmanová J, Neckář J, Kolář F, Nováková O, Novotný J, Hlaváčková M. Antioxidant tempol suppresses heart cytosolic phospholipase A2α stimulated by chronic intermittent hypoxia. Can J Physiol Pharmacol 2017; 95:920-927. [DOI: 10.1139/cjpp-2017-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adaptation to chronic intermittent hypoxia (CIH) is associated with reactive oxygen species (ROS) generation implicated in the improved cardiac tolerance against acute ischemia–reperfusion injury. Phospholipases A2(PLA2s) play an important role in cardiomyocyte phospholipid metabolism influencing membrane homeostasis. Here we aimed to determine the effect of CIH (7000 m, 8 h/day, 5 weeks) on the expression of cytosolic PLA2(cPLA2α), its phosphorylated form (p-cPLA2α), calcium-independent (iPLA2), and secretory (sPLA2IIA) at protein and mRNA levels, as well as fatty acids (FA) profile in left ventricular myocardium of adult male Wistar rats. Chronic administration of antioxidant tempol was used to verify the ROS involvement in CIH effect on PLA2s expression and phospholipid FA remodeling. While CIH did not affect PLA2s mRNA levels, it increased the total cPLA2α protein in cytosol and membranes (by 191% and 38%, respectively) and p-cPLA2α (by 23%) in membranes. On the contrary, both iPLA2and sPLA2IIA were downregulated by CIH. CIH further decreased phospholipid n-6 polyunsaturated FA (PUFA) and increased n-3 PUFA proportion. Tempol treatment prevented only CIH-induced cPLA2α up-regulation and its phosphorylation on Ser505. Our results show that CIH diversely affect myocardial PLA2s and suggest that ROS are responsible for the activation of cPLA2α under these conditions.
Collapse
Affiliation(s)
- Petra Míčová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martina Klevstig
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kristýna Holzerová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Marek Vecka
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University and General Teaching Hospital in Prague, Czech Republic
| | - Jitka Žurmanová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jan Neckář
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - František Kolář
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Nováková
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Novotný
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Markéta Hlaváčková
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
19
|
Astragalus polysaccharides exerts immunomodulatory effects via TLR4-mediated MyD88-dependent signaling pathway in vitro and in vivo. Sci Rep 2017; 7:44822. [PMID: 28303957 PMCID: PMC5355992 DOI: 10.1038/srep44822] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/15/2017] [Indexed: 12/18/2022] Open
Abstract
Astragalus polysaccharides (APS), which is widely used as a remedy to promote immunity of breast cancer patients, can enhance immune responses and exert anti-tumor effects. In this study, we investigated the effects and mechanisms of APS on macrophage RAW 264.7 and EAC tumor-bearing mice. Griess reaction and ELISA assays revealed that the concentrations of nitric oxide, TNF-α, IL-1β and IL-6 were increased by APS. However, this effect was diminished in the presence of TAK-242 (TLR4 inhibitor) or ST-2825(MyD88 inhibitor). In C57BL/10J (TLR4+/+wild-type) and C57BL/6J (MyD88+/+wild-type) tumor-bearing mice, the tumor apoptosis rate, immune organ indexes and the levels of TNF-α, IL-1β and IL-6 in blood increased and the tumor weight decreased by oral administration of APS for 25 days. APS had no obvious effects on IL-12p70. However, these effects were not significant in C57BL/10ScNJ (TLR4-deficient) and C57BL/B6.129P2(SJL)-Myd88m1.1Defr/J (MyD88-deficient) tumor-bearing mice. qRT-PCR and Western blot indicated that APS stimulated the key nodes in the TLR4-MyD88 dependent signaling pathway, including TLR4, MyD88, TRAF-6, NF-κB and AP-1, both in vitro and in vivo. However, TRAM was an exception. Moreover, TRAF-6 and NF-κB were not triggered by APS in gene-deficient tumor-bearing mice. Therefore, APS may modulate immunity of host organism through activation of TLR4-mediated MyD88-dependent signaling pathway.
Collapse
|
20
|
Kim YH, Bae JU, Kim IS, Chang CL, Oh SO, Kim CD. SIRT1 prevents pulmonary thrombus formation induced by arachidonic acid via downregulation of PAF receptor expression in platelets. Platelets 2016; 27:735-742. [DOI: 10.1080/09537104.2016.1190005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yun Hak Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Ung Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - In Suk Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine and Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, Republic of Korea
| | - Chulhun L. Chang
- Department of Laboratory Medicine, Pusan National University School of Medicine and Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, Republic of Korea
| | - Sae Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| |
Collapse
|
21
|
Lin CC, Yang CC, Wang CY, Tseng HC, Pan CS, Hsiao LD, Yang CM. NADPH Oxidase/ROS-Dependent VCAM-1 Induction on TNF-α-Challenged Human Cardiac Fibroblasts Enhances Monocyte Adhesion. Front Pharmacol 2016; 6:310. [PMID: 26858641 PMCID: PMC4729888 DOI: 10.3389/fphar.2015.00310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/16/2015] [Indexed: 01/23/2023] Open
Abstract
The inflammation-dependent adhesion molecule expressions are characterized in cardiovascular diseases and myocardial tissue infiltrations. Several pro-inflammatory cytokines are elevated in the acute myocardial injury and infarction. Tumor necrosis factor-α (TNF-α), a pro-inflammatory cytokine, is raised in the injury tissues and inflammatory regions and involved in the pathogenesis of cardiac injury, inflammation, and apoptosis. In fibroblasts, TNF-α-triggered expression of vascular cell adhesion molecule (VCAM)-1 aggravated the heart inflammation. However, the mechanisms underlying TNF-α-mediated VCAM-1 expression in cardiac fibroblasts remain unclear. Here, the primary cultured human cardiac fibroblasts (HCFs) were used to investigate the effects of TNF-α on VCAM-1 expression. The molecular evidence, including protein, mRNA, and promoter analyses, indicated that TNF-α-induced VCAM-1 gene expression is mediated through the TNFR-dependent manner. Activation of TNF-α/TNFR system triggered PKCα-dependent NADPH oxidase (Nox)/reactive oxygen species (ROS) signal linking to MAPK cascades, and then led to activation of the transcription factor, AP-1. Moreover, the results of mRNA and promoter assay demonstrated that c-Jun/AP-1 phosphorylated by TNF-α turns on VCAM-1 gene expression. Subsequently, up-regulated VCAM-1 on the cell surface of TNF-α-challenged HCFs increased the number of monocytes adhering to these cells. These results indicated that in HCFs, activation of AP-1 by PKCα-dependent Nox/ROS/MAPKs cascades is required for TNF-α-induced VCAM-1 expression. To clarify the mechanisms of TNF-α-induced VCAM-1 expression in HCFs may provide therapeutic strategies for heart injury and inflammatory diseases.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chien-Chung Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Lin-KouTao-Yuan, Taiwan
| | - Chen-Yu Wang
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Hui-Ching Tseng
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chih-Shuo Pan
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou, and College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan; Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and TechnologyTao-Yuan, Taiwan
| |
Collapse
|
22
|
Lin CC, Pan CS, Wang CY, Liu SW, Hsiao LD, Yang CM. Tumor necrosis factor-alpha induces VCAM-1-mediated inflammation via c-Src-dependent transactivation of EGF receptors in human cardiac fibroblasts. J Biomed Sci 2015; 22:53. [PMID: 26173590 PMCID: PMC4502472 DOI: 10.1186/s12929-015-0165-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 07/07/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Tumor necrosis factor-α (TNF-α) is a proinflammatory cytokine and elevated in the regions of tissue injury and inflammatory diseases. The deleterious effects of TNF-α on fibroblasts may aggravate heart inflammation mediated through the up-regulation of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1). However, the mechanisms underlying TNF-α-induced VCAM-1 expression in cardiac fibroblasts remain unknown. This study aimed to investigate the roles of TNF-α in VCAM-1 expression and its effects on human cardiac fibroblasts (HCFs). RESULTS The primary culture HCFs were used in this study. The results obtained with Western blotting, real time-quantitative PCR, and promoter activity analyses showed that TNF-α-induced VCAM-1 expression was mediated through TNF receptor (TNFR) 1-dependent gene up-regulation. Activation of TNFR1 by TNF-α transactivated c-Src-dependent EGF receptor (EGFR) linking to PI3K/Akt cascade, and then led to transcriptional activity of NF-κB. Moreover, the results of promoter reporter assay demonstrated that the phosphorylated p65 NF-κB turned on VCAM-1 gene expression. Subsequently, up-regulation of VCAM-1 promoted monocytes adhesion to HCFs challenged with TNF-α determined by cell adhesion assay. CONCLUSIONS Taken together, these results indicate that in HCFs, activation of NF-κB by c-Src-mediated transactivation of EGFR/PI3K/Akt cascade is required for TNF-α-induced VCAM-1 expression. Finally, increased VCAM-1 enhances monocytes adhering to HCFs challenged with TNF-α. Understanding the mechanisms of VCAM-1 up-regulated by TNF-α on HCFs may provide rationally therapeutic interventions for heart injury or inflammatory diseases.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo, Kwei-Shan, Tao-Yuan, Taiwan.,College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chih-Shuo Pan
- Department of Physiology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan.,Department of Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chen-Yu Wang
- Department of Physiology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan.,Department of Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan
| | - Shiau-Wen Liu
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo, Kwei-Shan, Tao-Yuan, Taiwan.,College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo, Kwei-Shan, Tao-Yuan, Taiwan.,College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan. .,Department of Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
23
|
Khan NS, Song CY, Jennings BL, Estes AM, Fang XR, Bonventre JV, Malik KU. Cytosolic phospholipase A2α is critical for angiotensin II-induced hypertension and associated cardiovascular pathophysiology. Hypertension 2015; 65:784-92. [PMID: 25667212 DOI: 10.1161/hypertensionaha.114.04803] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiotensin II activates cytosolic phospholipase A(2)α (cPLA2α) and releases arachidonic acid from tissue phospholipids, which mediate or modulate ≥1 cardiovascular effects of angiotensin II and has been implicated in hypertension. Because arachidonic acid release is the rate limiting step in eicosanoid production, cPLA2α might play a central role in the development of angiotensin II-induced hypertension. To test this hypothesis, we investigated the effect of angiotensin II infusion for 13 days by micro-osmotic pumps on systolic blood pressure and associated pathogenesis in wild type (cPLA2α(+/+)) and cPLA2α(-/-) mice. Angiotensin II-induced increase in systolic blood pressure in cPLA2α(+/+) mice was abolished in cPLA2α(-/-) mice; increased systolic blood pressure was also abolished by the arachidonic acid metabolism inhibitor, 5,8,11,14-eicosatetraynoic acid in cPLA2α(+/+) mice. Angiotensin II in cPLA2α(+/+) mice increased cardiac cPLA2 activity and urinary eicosanoid excretion, decreased cardiac output, caused cardiovascular remodeling with endothelial dysfunction, and increased vascular reactivity in cPLA2α(+/+) mice; these changes were diminished in cPLA2α(-/-) mice. Angiotensin II also increased cardiac infiltration of F4/80(+) macrophages and CD3(+) T lymphocytes, cardiovascular oxidative stress, expression of endoplasmic reticulum stress markers p58(IPK), and CHOP in cPLA2α(+/+) but not cPLA2α(-/-) mice. Angiotensin II increased cardiac activity of ERK1/2 and cSrc in cPLA2α(+/+) but not cPLA2α(-/-) mice. These data suggest that angiotensin II-induced hypertension and associated cardiovascular pathophysiological changes are mediated by cPLA2α activation, most likely through the release of arachidonic acid and generation of eicosanoids with predominant prohypertensive effects and activation of ≥1 signaling molecules, including ERK1/2 and cSrc.
Collapse
Affiliation(s)
- Nayaab S Khan
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Chi Young Song
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Brett L Jennings
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Anne M Estes
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Xiao R Fang
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Joseph V Bonventre
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Kafait U Malik
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN (N.S.K., C.Y.S., B.L.J., A.M.E., X.R.F., K.U.M.); and Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.).
| |
Collapse
|
24
|
CYP-13A12 of the nematode Caenorhabditis elegans is a PUFA-epoxygenase involved in behavioural response to reoxygenation. Biochem J 2014; 464:61-71. [DOI: 10.1042/bj20140848] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CYP-13A12 of the nematode Caenorhabditis elegans was characterized after heterologous expression in insect cells as a PUFA epoxygenase producing eicosanoids. These metabolites function as signalling molecules in the regulation of the O2-ON response, a rapid increase of locomotion in response to anoxia/reoxygenation.
Collapse
|
25
|
Abstract
This article outlines the therapeutic mechanisms of hyperbaric oxygenation in acute stroke, based on information obtained from peer-reviewed medical literature. Hyperbaric oxygen is an approved treatment modality for ischemia-reperfusion injury in several conditions. It maintains the viability of the marginal tissue, reduces the mitochondrial dysfunction, metabolic penumbra, and blocks inflammatory cascades observed in acute stroke. Basic and clinical data suggest that hyperbaric oxygen could be a safe and effective treatment option in the management of acute stroke. Further work is needed to clarify its clinical utility when applied within the treatment window of "gold standard" treatments (<3-5 hours).
Collapse
|
26
|
Takahashi S, Suzuki K, Watanabe Y, Watanabe K, Fujioka D, Nakamura T, Obata JE, Kawabata K, Mishina H, Kugiyama K. Phospholipase A2 expression in coronary thrombus is increased in patients with recurrent cardiac events after acute myocardial infarction. Int J Cardiol 2013; 168:4214-21. [PMID: 23948114 DOI: 10.1016/j.ijcard.2013.07.154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Intracoronary thrombus is a source of active lipid mediators including eicosanoids that play a critical role in the pathogenesis of acute myocardial infarction (AMI). Eicosanoids are derived from arachidonic acid generated by phospholipase A(2) (PLA(2)). This study examined whether PLA(2) is expressed in the aspirated coronary thrombus and whether PLA(2) expression in the thrombus may be related to recurrence of cardiac events and development of atherosclerosis in the culprit coronary artery after AMI. METHODS Intracoronary thrombus was obtained using an aspiration catheter from 48 patients with AMI, who had successful emergent treatment with percutaneous coronary intervention (PCI). Repeated intravascular ultrasound in the culprit coronary artery was performed at emergent PCI and 6 months later in a subgroup of 20 patients. RESULTS There was a higher prevalence of cells in the thrombus that were immunopositive to group IIA, IVA, V and X PLA2s in patients with (n = 11) than without (n = 37) cardiac events during 6 months of follow-up (P < 0.05 for all). The prevalence of the cells that were immunopositive to group IIA, IVA and V PLA2s in the thrombus was significantly associated with the percent increase in atheroma volume (r = 0.60, 0.55 and 0.45, respectively, P < 0.05 for all) after 6 months in the native coronary segment distal to the culprit coronary lesion. CONCLUSION PLA(2) expression in coronary thrombus is associated with recurrence of cardiac events and development of atherosclerosis in the culprit coronary artery in AMI survivors.
Collapse
Affiliation(s)
- Soichiro Takahashi
- Department of Internal Medicine II, University of Yamanashi, Faculty of Medicine, Chuo, Yamanashi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yu J, Deliu E, Zhang XQ, Hoffman NE, Carter RL, Grisanti LA, Brailoiu GC, Madesh M, Cheung JY, Force T, Abood ME, Koch WJ, Tilley DG, Brailoiu E. Differential activation of cultured neonatal cardiomyocytes by plasmalemmal versus intracellular G protein-coupled receptor 55. J Biol Chem 2013; 288:22481-92. [PMID: 23814062 DOI: 10.1074/jbc.m113.456178] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The L-α-lysophosphatidylinositol (LPI)-sensitive receptor GPR55 is coupled to Ca(2+) signaling. Low levels of GPR55 expression in the heart have been reported. Similar to other G protein-coupled receptors involved in cardiac function, GPR55 may be expressed both at the sarcolemma and intracellularly. Thus, to explore the role of GPR55 in cardiomyocytes, we used calcium and voltage imaging and extracellular administration or intracellular microinjection of GPR55 ligands. We provide the first evidence that, in cultured neonatal ventricular myocytes, LPI triggers distinct signaling pathways via GPR55, depending on receptor localization. GPR55 activation at the sarcolemma elicits, on one hand, Ca(2+) entry via L-type Ca(2+) channels and, on the other, inositol 1,4,5-trisphosphate-dependent Ca(2+) release. The latter signal is further amplified by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Conversely, activation of GPR55 at the membrane of intracellular organelles promotes Ca(2+) release from acidic-like Ca(2+) stores via the endolysosomal NAADP-sensitive two-pore channels. This response is similarly enhanced by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Extracellularly applied LPI produces Ca(2+)-independent membrane depolarization, whereas the Ca(2+) signal induced by intracellular microinjection of LPI converges to hyperpolarization of the sarcolemma. Collectively, our findings point to GPR55 as a novel G protein-coupled receptor regulating cardiac function at two cellular sites. This work may serve as a platform for future studies exploring the potential of GPR55 as a therapeutic target in cardiac disorders.
Collapse
Affiliation(s)
- Justine Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wang WZ, Jones AW, Wang M, Durante W, Korthuis RJ. Preconditioning with soluble guanylate cyclase activation prevents postischemic inflammation and reduces nitrate tolerance in heme oxygenase-1 knockout mice. Am J Physiol Heart Circ Physiol 2013; 305:H521-32. [PMID: 23771693 DOI: 10.1152/ajpheart.00810.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously we have shown that, unlike wild-type mice (WT), heme oxygenase-1 knockout (HO-1-/-) mice developed nitrate tolerance and were not protected from inflammation caused by ischemia-reperfusion (I/R) when preconditioned with a H2S donor. We hypothesized that stimulation (with BAY 41-2272) or activation (with BAY 60-2770) of soluble guanylate cyclase (sGC) would precondition HO-1-/- mice against an inflammatory effect of I/R and increase arterial nitrate responses. Intravital fluorescence microscopy was used to visualize leukocyte rolling and adhesion to postcapillary venules of the small intestine in anesthetized mice. Relaxation to ACh and BAY compounds was measured on superior mesenteric arteries isolated after I/R protocols. Preconditioning with either BAY compound 10 min (early phase) or 24 h (late phase) before I/R reduced postischemic leukocyte rolling and adhesion to sham control levels and increased superior mesenteric artery responses to ACh, sodium nitroprusside, and BAY 41-2272 in WT and HO-1-/- mice. Late-phase preconditioning with BAY 60-2770 was maintained in HO-1-/- and endothelial nitric oxide synthase knockout mice pretreated with an inhibitor (dl-propargylglycine) of enzymatically produced H2S. Pretreatment with BAY compounds also prevented the I/R increase in small intestinal TNF-α. We speculate that increasing sGC activity and related PKG acts downstream to H2S and disrupts signaling processes triggered by I/R in part by maintaining low cellular Ca²⁺. In addition, BAY preconditioning did not increase sGC levels, yet increased the response to agents that act on reduced heme-containing sGC. Collectively these actions would contribute to increased nitrate sensitivity and vascular function.
Collapse
Affiliation(s)
- Walter Z Wang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | | | | | | | | |
Collapse
|
29
|
Yang CM, Lee IT, Hsu RC, Chi PL, Hsiao LD. NADPH oxidase/ROS-dependent PYK2 activation is involved in TNF-α-induced matrix metalloproteinase-9 expression in rat heart-derived H9c2 cells. Toxicol Appl Pharmacol 2013; 272:431-42. [PMID: 23774252 DOI: 10.1016/j.taap.2013.05.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/16/2013] [Accepted: 05/30/2013] [Indexed: 11/29/2022]
Abstract
TNF-α plays a mediator role in the pathogenesis of chronic heart failure contributing to cardiac remodeling and peripheral vascular disturbances. The implication of TNF-α in inflammatory responses has been shown to be mediated through up-regulation of matrix metalloproteinase-9 (MMP-9). However, the detailed mechanisms of TNF-α-induced MMP-9 expression in rat embryonic-heart derived H9c2 cells are largely not defined. We demonstrated that in H9c2 cells, TNF-α induced MMP-9 mRNA and protein expression associated with an increase in the secretion of pro-MMP-9. TNF-α-mediated responses were attenuated by pretreatment with the inhibitor of ROS (N-acetyl-l-cysteine, NAC), NADPH oxidase [apocynin (APO) or diphenyleneiodonium chloride (DPI)], MEK1/2 (U0126), p38 MAPK (SB202190), JNK1/2 (SP600125), NF-κB (Bay11-7082), or PYK2 (PF-431396) and transfection with siRNA of TNFR1, p47(phox), p42, p38, JNK1, p65, or PYK2. Moreover, TNF-α markedly induced NADPH oxidase-derived ROS generation in these cells. TNF-α-enhanced p42/p44 MAPK, p38 MAPK, JNK1/2, and NF-κB (p65) phosphorylation and in vivo binding of p65 to the MMP-9 promoter were inhibited by U0126, SB202190, SP600125, NAC, DPI, or APO. In addition, TNF-α-mediated PYK2 phosphorylation was inhibited by NAC, DPI, or APO. PYK2 inhibition could reduce TNF-α-stimulated MAPKs and NF-κB activation. Thus, in H9c2 cells, we are the first to show that TNF-α-induced MMP-9 expression is mediated through a TNFR1/NADPH oxidase/ROS/PYK2/MAPKs/NF-κB cascade. We demonstrated that NADPH oxidase-derived ROS generation is involved in TNF-α-induced PYK2 activation in these cells. Understanding the regulation of MMP-9 expression and NADPH oxidase activation by TNF-α on H9c2 cells may provide potential therapeutic targets of chronic heart failure.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan.
| | | | | | | | | |
Collapse
|
30
|
Yang CM, Lee IT, Lin CC, Wang CH, Cherng WJ, Hsiao LD. c-Src-dependent MAPKs/AP-1 activation is involved in TNF-α-induced matrix metalloproteinase-9 expression in rat heart-derived H9c2 cells. Biochem Pharmacol 2013; 85:1115-23. [PMID: 23353699 DOI: 10.1016/j.bcp.2013.01.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
Abstract
TNF-α plays a critical mediator in the pathogenesis of chronic heart failure contributing to cardiac remodeling and peripheral vascular disturbances. The implication of TNF-α in inflammatory responses has been shown to be mediated through up-regulation of inflammatory genes, including matrix metalloproteinase-9 (MMP-9). However, the detailed mechanisms of TNF-α-induced MMP-9 expression are largely unclear in the heart cells. Here, we demonstrated that in rat embryonic-heart derived H9c2 cells, TNF-α could induce MMP-9 mRNA expression associated with an increase in the secretion of MMP-9, determined by real-time PCR, zymography, and promoter activity assays. TNF-α-mediated responses were attenuated by pretreatment with the inhibitor of c-Src (PP1), EGFR (AG1478), PDGFR (AG1296), PI3K (LY294002), Akt (SH-5), MEK1/2 (U0126), p38 MAPK (SB202190), JNK1/2 (SP600125), or AP-1 (Tanshinone IIA) and transfection with siRNA of c-Src, EGFR, PDGFR, p110, Akt, or c-Jun. TNF-α stimulated c-Src, PDGFR, and EGFR phosphorylation, which were reduced by PP1. In addition, TNF-α-stimulated Akt phosphorylation was inhibited by PP1, AG1478, AG1296, or LY294002. We further demonstrated that TNF-α markedly stimulated p38 MAPK, p42/p44 MAPK, and JNK1/2 phosphorylation via a c-Src/EGFR, PDGFR/PI3K/Akt pathway. Finally, we showed that, in H9c2 cells, TNF-α-stimulated AP-1 promoter activity, c-Jun mRNA expression, and c-Jun phosphorylation were attenuated by PP1, AG1478, AG1296, LY294002, SB202190, SP600125, or U0126. These results suggested that TNF-α-induced MMP-9 expression is mediated through a c-Src/EGFR, PDGFR/PI3K/Akt/MAPKs/AP-1 cascade in H9c2 cells. Consequently, MMP-9 induction may contribute to cell migration and cardiovascular inflammation.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.
| | | | | | | | | | | |
Collapse
|
31
|
Magrioti V, Kokotos G. Phospholipase A2inhibitors for the treatment of inflammatory diseases: a patent review (2010 – present). Expert Opin Ther Pat 2013; 23:333-44. [DOI: 10.1517/13543776.2013.754425] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|