1
|
Alharbi HO, Sugden PH, Clerk A. Mitogen-activated protein kinase signalling in rat hearts during postnatal development: MAPKs, MAP3Ks, MAP4Ks and DUSPs. Cell Signal 2024; 124:111397. [PMID: 39251052 DOI: 10.1016/j.cellsig.2024.111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Mammalian cardiomyocytes become terminally-differentiated during the perinatal period. In rodents, cytokinesis ceases after a final division cycle immediately after birth. Nuclear division continues and most cardiomyocytes become binucleated by ∼11 days. Subsequent growth results from an increase in cardiomyocyte size. The mechanisms involved remain under investigation. Mitogen-activated protein kinases (MAPKs) regulate cell growth/death: extracellular signal-regulated kinases 1/2 (ERK1/2) promote proliferation, whilst c-Jun N-terminal kinases (JNKs) and p38-MAPKs respond to cellular stresses. We assessed their regulation in rat hearts during postnatal development (2, 7, 14, and 28 days, 12 weeks) during which time there was rapid, substantial downregulation of mitosis/cytokinesis genes (Cenpa/e/f, Aurkb, Anln, Cdca8, Orc6) with lesser downregulation of DNA replication genes (Orcs1-5, Mcms2-7). MAPK activation was assessed by immunoblotting for total and phosphorylated (activated) kinases. Total ERK1/2 was downregulated, but not JNKs or p38-MAPKs, whilst phosphorylation of all MAPKs increased relative to total protein albeit transiently for JNKs. These profiles differed from activation of Akt (also involved in cardiomyocyte growth). Dual-specificity phosphatases, upstream MAPK kinase kinases (MAP3Ks), and MAP3K kinases (MAP4Ks) identified in neonatal rat cardiomyocytes by RNASeq were differentially regulated during postnatal cardiac development. The MAP3Ks that we could assess by immunoblotting (RAF kinases and Map3k3) showed greater downregulation of the protein than mRNA. MAP3K2/MAP3K3/MAP4K5 were upregulated in human failing heart samples and may be part of the "foetal gene programme" of re-expressed genes in disease. Thus, MAPKs, along with kinases and phosphatases that regulate them, potentially play a significant role in postnatal remodelling of the heart.
Collapse
Affiliation(s)
- Hajed O Alharbi
- Department of Medical Laboratory, College of Applied Medical Sciences, Quassim University, Buraydah, Saudi Arabia; School of Biological Sciences, University of Reading, Reading, UK
| | - Peter H Sugden
- School of Biological Sciences, University of Reading, Reading, UK
| | - Angela Clerk
- School of Biological Sciences, University of Reading, Reading, UK.
| |
Collapse
|
2
|
ElKhatib MAW, Gerges SH, Isse FA, El-Kadi AOS. Cytochrome P450 1B1 is critical in the development of TNF-α, IL-6, and LPS-induced cellular hypertrophy. Can J Physiol Pharmacol 2024; 102:408-421. [PMID: 38701513 DOI: 10.1139/cjpp-2024-0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Heart failure (HF) is preceded by cellular hypertrophy (CeH) which alters expression of cytochrome P450 enzymes (CYPs) and arachidonic acid (AA) metabolism. Inflammation is involved in CeH pathophysiology, but mechanisms remain elusive. This study investigates the impacts of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and lipopolysaccharides (LPS) on the development of CeH and the role of CYP1B1. AC16 cells were treated with TNF-α, IL-6, and LPS in the presence and absence of CYP1B1-siRNA or resveratrol. mRNA and protein expression levels of CYP1B1 and hypertrophic markers were determined using PCR and Western blot analysis, respectively. CYP1B1 enzyme activity was determined, and AA metabolites were analyzed using liquid chromatography-tandem mass spectrometry. Our results show that TNF-α, IL-6, and LPS induce expression of hypertrophic markers, induce CYP1B1 expression, and enantioselectively modulate CYP1B1-mediated AA metabolism in favor of mid-chain HETEs. CYP1B1-siRNA or resveratrol ameliorated these effects. In conclusion, our results demonstrate the crucial role of CYP1B1 in TNF-α, IL-6, and LPS-induced CeH.
Collapse
Affiliation(s)
- Mohammed A W ElKhatib
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Fadumo A Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
He J, Huang L, Sun K, Li J, Han S, Gao X, Wang QQ, Yang S, Sun W, Gao H. Oleuropein alleviates myocardial ischemia-reperfusion injury by suppressing oxidative stress and excessive autophagy via TLR4/MAPK signaling pathway. Chin Med 2024; 19:59. [PMID: 38589925 PMCID: PMC11003011 DOI: 10.1186/s13020-024-00925-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MIRI) is an important complication of reperfusion therapy, and has a lack of effective prevention and treatment methods. Oleuropein (OP) is a natural strong antioxidant with many protective effects on cardiovascular diseases, but its protective effect on MIRI has not yet been studied in depth. METHODS Tert-Butyl hydroperoxide (tBHP) was used to establish an in vitro oxidative stress model. Cell viability was detected by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and lactate dehydrogenase (LDH). Flow cytometry and fluorescence assays were performed for evaluating the ROS levels and mitochondrial membrane potential (MMP). Immunofluorescence analysis detected the NRF2 nuclear translocation and autophagy indicators. Further, Western blotting and quantitative real-time PCR were performed to evaluate the expression levels of proteins and mRNAs. Molecular docking, CETSA, and molecular interaction analysis explored the binding between OP and TLR4. The protective effects of OP in vivo were determined using a preclinical MIRI rat model. RESULTS OP protected against tBHP-treated injury, reduced ROS levels and reversed the damaged MMP. Mechanistically, OP activated NRF2-related antioxidant pathways, inhibited autophagy and attenuated the TLR4/MAPK signaling pathway in tBHP-treated H9C2 cells with a high binding affinity to TLR4 (KD = 37.5 µM). The TLR4 inhibitor TAK242 showed a similar effect as OP. In vivo, OP could alleviate cardiac ischemia/reperfusion injury and it ameliorated adverse cardiac remodeling. Consistent with in vitro studies, OP inhibited TLR4/MAPK and autophagy pathway and activated NRF2-dependent antioxidant pathways in vivo. CONCLUSION This study shows that OP binds to TLR4 to regulate oxidative stress and autophagy for protecting damaged cardiomyocytes, supporting that OP can be a potential therapeutic agent for MIRI.
Collapse
Affiliation(s)
- Jia He
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Liting Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Jilang Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Shan Han
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Research Center for Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Xiang Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Qin-Qin Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China
| | - Shilin Yang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China.
| | - Wen Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530020, China.
| |
Collapse
|
4
|
Zheng S, Tan W, Li X, Wang L, Zhu C, Pyle WG, Chen J, Wu J, Ren X, Chen H, Zou Y, Backx PH, Yang FH. Apelin receptor inhibition in ischemia-reperfused mouse hearts protected by endogenous n-3 polyunsaturated fatty acids. Front Pharmacol 2023; 14:1145413. [PMID: 37942483 PMCID: PMC10628527 DOI: 10.3389/fphar.2023.1145413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Background: While the protective effects of n-3 polyunsaturated fatty acids (PUFAs) on cardiac ischemia-reperfusion (IR) injury have been previously reported, limited data are available regarding how these fatty acids affect membrane receptors and their downstream signaling following IR injury. We aimed to identify potential receptors activated by n-3 PUFAs in IR hearts to understand the regulatory mechanisms of these receptors. Methods: We used fat-1 mice, which naturally have elevated levels of n-3 PUFAs, and C57BL/6J mice as a control group to create a myocardial IR injury model through Langendorff perfusion. We assessed the impact of endogenous n-3 PUFAs on left ventricular function, myocardial infarct size, myocardial apoptosis, and ATP production. RNA sequencing (RNA-seq) and bioinformatics analysis were conducted to identify molecular targets affected by n-3 PUFAs. Based on these analyses we then treated IR hearts of WT and fat-1 mice with an antagonist (ML221) or an agonist (apelin-13) for the predicted receptor to assess cardiac contractile function and intracellular signaling pathways. An in vitro hypoxia-reoxygenation (HR) model was also used to confirm the effects of n-3 PUFAs on the examined intracellular signaling pathways. Results: Endogenous n-3 PUFAs protected cardiac structure and function in post-IR hearts, and modulated phosphorylation patterns in the PI3K-AKT-mTOR signaling pathways. RNA-seq analysis revealed that n-3 PUFAs affected multiple biological processes as well as levels of the apelin receptor (APLNR). Consistent with a role for the PLNNR, ML221 synchronized the activation of the PI3K-AKT-mTOR signaling axis, suppressed the expression of PKCδ and phosphorylated p38α, upregulated PKCε expression, upregulated or restored the phosphorylation of myofilaments, and prevented myocardial injury and contractile dysfunction in WT IR hearts. By contrast, apelin-13 disrupted the PI3K-AKT-mTOR signaling axis in post-IR fat-1 hearts. The phosphorylation signaling targeted by APLNR inhibition in post-IR fat-1 hearts was also observed after treating HR cells with eicosatetraenoic acid (EPA). Conclusion: Endogenous n-3 PUFAs protect against post-IR injury and preserve cardiac contractile function possibly through APLNR inhibition. This inhibition synchronizes the PI3K-AKT-mTOR axis, suppresses detrimental phosphorylation signaling, and restores or increases myofilament phosphorylation in post-IR hearts. The beneficial effects observed in fat-1 transgenic mouse hearts can be attributed, at least in part, to elevated EPA levels. This study is the first to demonstrate that n-3 PUFAs protect hearts against IR injury through APLNR inhibition.
Collapse
Affiliation(s)
- Shuang Zheng
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Weijiang Tan
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiang Li
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lijing Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Caiyi Zhu
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - W. Glen Pyle
- IMPART Investigator Team, Dalhousie Medicine, Saint John, NB, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Jianxin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuecong Ren
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Honghua Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Yunzeng Zou
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peter H. Backx
- Department of Biology, York University, Toronto, ON, Canada
| | - Feng Hua Yang
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| |
Collapse
|
5
|
Chen Y, Liang C, Li J, Ma L, Wang B, Yuan Z, Yang S, Nong X. Effect of artesunate on cardiovascular complications in periodontitis in a type I diabetes rat model and related mechanisms. J Endocrinol Invest 2023; 46:2031-2053. [PMID: 36892740 DOI: 10.1007/s40618-023-02052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/24/2023] [Indexed: 03/10/2023]
Abstract
PURPOSE Both cardiovascular disease and periodontitis are complications of diabetes that have a great impact on human life and health. Our previous research found that artesunate can effectively improve cardiovascular disease in diabetes and has an inhibitory effect on periodontal disease. Therefore, the present study aimed to explore the potential therapeutic possibility of artesunate in the protection against cardiovascular complications in periodontitis with type I diabetes rats and to elucidate the possible underlying mechanisms. METHODS Sprague‒Dawley rats were randomly divided into the healthy, diabetic, periodontitis, diabetic with periodontitis, and artesunate treatment groups (10, 30, and 60 mg/kg, i.g.). After artesunate treatment, oral swabs were collected and used to determine changes in the oral flora. Micro-CT was performed to observe changes in alveolar bone. Blood samples were processed to measure various parameters, while cardiovascular tissues were evaluated by haematoxylin-eosin, Masson, Sirius red, and TUNEL staining to observe fibrosis and apoptosis. The protein and mRNA expression levels in the alveolar bone and cardiovascular tissues were detected using immunohistochemistry and RT‒PCR. RESULTS Diabetic rats with periodontitis and cardiovascular complications maintained heart and body weight but exhibited reduced blood glucose levels, and they were able to regulate blood lipid indicators at normal levels after artesunate treatment. The staining assays suggested that treatment with 60 mg/kg artesunate has a significant therapeutic effect on myocardial apoptotic fibrosis. The high expression of NF-κB, TLR4, VEGF, ICAM-1, p38 MAPK, TGF-β, Smad2, and MMP9 in the alveolar bone and cardiovascular tissue in the type I diabetes and type I diabetes with periodontitis rat models was reduced after treatment with artesunate in a concentration-dependent manner. Micro-CT showed that treatment with 60 mg/kg artesunate effectively alleviated alveolar bone resorption and density reduction. The sequencing results suggested that each model group of rats had vascular and oral flora dysbiosis, but artesunate treatment could correct the dysbacteriosis. CONCLUSIONS Periodontitis-related pathogenic bacteria cause dysbiosis of the oral and intravascular flora in type I diabetes and aggravate cardiovascular complications. The mechanism by which periodontitis aggravates cardiovascular complications involves the NF-κB pathway, which induces myocardial apoptosis, fibrosis, and vascular inflammation.
Collapse
Affiliation(s)
- Y Chen
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - C Liang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - J Li
- Life Science Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Medical Science Research Center, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - L Ma
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - B Wang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Z Yuan
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - S Yang
- School of Information and Management, Nanning, 530021, Guangxi, China
| | - X Nong
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
6
|
Repeated exposure to 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP) accelerates ligand-independent activation of estrogen receptors in long-term estradiol-deprived MCF-7 cells. Toxicol Lett 2023; 378:31-38. [PMID: 36863540 DOI: 10.1016/j.toxlet.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
It was previously identified that there may be an active metabolite of bisphenol A (BPA), 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP). An in vitro system was developed to detect MBP toxicity to the Michigan Cancer Foundation-7 (MCF-7) cells that had been repeatedly exposed to a low dose of the metabolite. MBP profoundly activated estrogen receptor (ER)-dependent transcription as a ligand, with an EC50 of 2.8 nM. Women are continuously exposed to numerous estrogenic environmental chemicals; but their susceptibility to these chemicals may be significantly altered after menopause. Long-term estrogen-deprived (LTED) cells, which display ligand-independent ER activation, are a postmenopausal breast cancer model derived from MCF-7 cells. In this study, we investigated the estrogenic effects of MBP on LTED cells in a repeated exposure in vitro model. The results suggest that i) nanomolar levels of MBP reciprocally disrupt the balanced expression of ERα and ERβ proteins, leading to the dominant expression of ERβ, ii) MBP stimulates ERs-mediated transcription without acting as an ERβ ligand, and iii) MBP utilizes mitogen-activated protein kinase and phosphatidylinositol-3 kinase signaling to evoke its estrogenic action. Moreover, the repeated exposure strategy was effective for detecting low-dose estrogenic-like effects caused by MBP in LTED cells.
Collapse
|
7
|
Hu S, Jiang L, Yan Q, Zhou C, Guo X, Chen T, Ma S, Luo Y, Hu C, Yang F, Yuan L, Ma X, Zeng J. Evidence construction of baicalin for treating myocardial ischemia diseases: A preclinical meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154476. [PMID: 36191551 DOI: 10.1016/j.phymed.2022.154476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/12/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Baicalin, a flavonoid glycoside isolated from Scutellaria baicalensis Georgi, has shown potential pharmacological effects on myocardial ischemia diseases. Nevertheless, systematic preclinical studies on baicalin in the treatment of ischemic diseases are scarce. PURPOSE To assess the efficacy and potential mechanisms of baicalin in myocardial ischemia (RI), myocardial ischemia-reperfusion (IR) injury and myocardial infarction (MI) animal models for future clinical research. METHODS Preclinical studies published prior to August 27th, 2022 were retrieved from PubMed, Embase, Web of Science and Cochrane Library. CAMARADES list was used to evaluate the quality of included researches. Meta-analyses of cardiac pathology and function parameters, myocardial injury markers and other indicators were performed by STATA 15.0 software. Potential mechanisms are categorized and summarized. Dose-response interval analyses were used to analyze the dose-response relationship between baicalin and myocardial ischemia disease. RESULTS Fourteen studies and 222 animals were included in the analysis. The results showed that compared with the control group, baicalin could reduce myocardial infarction size associated with cardiac pathological condition and the corresponding cardiac pathological index containing CK-MB, CK and cTnT. Additionally, heart function indicators including LVSP, LVFS, LVEF, -dp/dt max, dp/dt max were increased by baicalin. As for subgroup analyses, baicalin also demonstrated certain effect on CK-MB and LVSP by administration method or stage. Furthermore, it displayed obvious effect on myocardial ischemia diseases when the dose is maintained at 100-150 mg/kg based on dosage analyses. CONCLUSION Based on the relevant literature retrieved, this is the first meta-analysis on baicalin in treating myocardial ischemia diseases. Notably, we linked the dynamic development of the disease and discussed it pertinently, from RI, IR injury to MI. Baicalin exhibits positive effects on myocardial ischemia diseases (especially when the dose is 100-150 mg/kg), which is achieved by regulating key pathological indicators and various signaling pathways.
Collapse
Affiliation(s)
- Sihan Hu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Lan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qi Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chenyang Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaochuan Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Tong Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Siting Ma
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yimiao Luo
- Department of Integrated Traditional Chinese and Western Medicine of Peking University Health Science Center, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China
| | - Caiyu Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Fumin Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Lishan Yuan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jinhao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
8
|
Perry F, Lahaye L, Santin E, Johnson C, Korver D, Kogut M, Arsenault R. Protected Biofactors and Antioxidants Reduce the Negative Consequences of Virus and Cold Challenge while Enhancing Performance by Modulating Immunometabolism through Cytoskeletal and Immune Signaling in the Jejunum. Poult Sci 2022; 101:102172. [PMID: 36240637 PMCID: PMC9573920 DOI: 10.1016/j.psj.2022.102172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/19/2022] [Accepted: 08/30/2022] [Indexed: 11/27/2022] Open
|
9
|
Dent P. Cell Signaling and Translational Developmental Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC7538147 DOI: 10.1016/b978-0-12-820472-6.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The relationships between drug pharmacodynamics and subsequent changes in cellular signaling processes are complex. Many in vitro cell signaling studies often use drug concentrations above physiologically safe drug levels achievable in a patient's plasma. Drug companies develop agents to inhibit or modify the activities of specific target enzymes, often without a full consideration that their compounds have additional unknown targets. These two negative sequelae, when published together, become impediments against successful developmental therapeutics and translation because this data distorts our understanding of signaling mechanisms and reduces the probability of successfully translating drug-based concepts from the bench to the bedside. This article will discuss cellular signaling in isolation and as it relates to extant single and combined therapeutic drug interventions. This will lead to a hypothetical series standardized sequential approaches describing a rigorous concept to drug development and clinical translation.
Collapse
|
10
|
Tang XH, Gambardella J, Jankauskas S, Wang X, Santulli G, Gudas LJ, Levi R. A Retinoic Acid Receptor β 2 Agonist Improves Cardiac Function in a Heart Failure Model. J Pharmacol Exp Ther 2021; 379:182-190. [PMID: 34389654 PMCID: PMC8626778 DOI: 10.1124/jpet.121.000806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
We previously demonstrated that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. We hypothesized that by decreasing oxidative stress and consequent fibrogenesis, AC261066 could attenuate the development of contractile dysfunction in post-ischemic heart failure (HF). We tested this hypothesis in vivo using an established murine model of myocardial infarction (MI), obtained by permanent occlusion of the left anterior descending coronary artery. Treating mice with AC261066 in drinking water significantly attenuated the post-MI deterioration of echocardiographic indices of cardiac function, diminished remodeling, and reduced oxidative stress, as evidenced by a decrease in malondialdehyde level and p38 mitogen-activated protein kinase expression in cardiomyocytes. The effects of AC261066 were also associated with a decrease in interstitial fibrosis, as shown by a marked reduction in collagen deposition and α-smooth muscle actin expression. In cardiac murine fibroblasts subjected to hypoxia, AC261066 reversed hypoxia-induced decreases in superoxide dismutase 2 and angiopoietin-like 4 transcriptional levels as well as the increase in NADPH oxidase 2 mRNA, demonstrating that the post-MI cardioprotective effects of AC261066 are associated with an action at the fibroblast level. Thus, AC261066 alleviates post-MI cardiac dysfunction by modulating a set of genes involved in the oxidant/antioxidant balance. These AC261066 responsive genes diminish interstitial fibrogenesis and remodeling. Since MI is a recognized major cause of HF, our data identify RARβ 2 as a potential pharmacological target in the treatment of HF. SIGNIFICANCE STATEMENT: A previous report showed that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. This study shows that AC261066 attenuates the development of contractile dysfunction and maladaptive remodeling in post-ischemic heart failure (HF) by modulating a set of genes involved in oxidant/antioxidant balance. Since myocardial infarction is a recognized major cause of HF, these data identify RARβ 2 as a potential pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Jessica Gambardella
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Stanislovas Jankauskas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Xujun Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Gaetano Santulli
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| |
Collapse
|
11
|
Miyake T, McDermott JC. Nucleolar localization of c-Jun. FEBS J 2021; 289:748-765. [PMID: 34499807 DOI: 10.1111/febs.16187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 01/20/2023]
Abstract
Nucleoli are well defined for their function in ribosome biogenesis, but only a small fraction of the nucleolar proteome has been characterized. Here, we report that the proto-oncogene, c-Jun, is targeted to the nucleolus. Using live cell imaging in myogenic cells, we document that the c-Jun basic domain contains a unique, evolutionarily conserved motif that determines nucleolar targeting. Fos family Jun dimer partners, such as Fra2, while nuclear, do not co-localize with c-Jun in the nucleolus. A point mutation in c-Jun that mimics Fra2 (M260E) in its Nucleolar Localization sequence (NoLS) results in loss of c-Jun nucleolar targeting while still preserving nuclear localization. Fra2 can sequester c-Jun in the nucleoplasm, indicating that the stoichiometric ratio of heterodimeric partners regulates c-Jun nucleolar targeting. Finally, nucleolar localization of c-Jun modulates nucleolar architecture and ribosomal RNA accumulation. These studies highlight a novel role for Jun family proteins in the nucleolus, having potential implications for a diverse array of AP-1-regulated cellular processes.
Collapse
Affiliation(s)
- Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada.,Centre for Research in Mass Spectrometry (CRMS), York University, Toronto, ON, Canada
| |
Collapse
|
12
|
The insulin receptor family and protein kinase B (Akt) are activated in the heart by alkaline pH and α1-adrenergic receptors. Biochem J 2021; 478:2059-2079. [PMID: 34002209 PMCID: PMC8203208 DOI: 10.1042/bcj20210144] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
Insulin and insulin-like growth factor stimulate protein synthesis and cardioprotection in the heart, acting through their receptors (INSRs, IGF1Rs) and signalling via protein kinase B (PKB, also known as Akt). Protein synthesis is increased in hearts perfused at alkaline pHo to the same extent as with insulin. Moreover, α1-adrenergic receptor (α1-AR) agonists (e.g. phenylephrine) increase protein synthesis in cardiomyocytes, activating PKB/Akt. In both cases, the mechanisms are not understood. Our aim was to determine if insulin receptor-related receptors (INSRRs, activated in kidney by alkaline pH) may account for the effects of alkaline pHo on cardiac protein synthesis, and establish if α1-ARs signal through the insulin receptor family. Alkaline pHo activated PKB/Akt signalling to the same degree as insulin in perfused adult male rat hearts. INSRRs were expressed in rat hearts and, by immunoblotting for phosphorylation (activation) of INSRRs/INSRs/IGF1Rs, we established that INSRRs, together with INSRs/IGF1Rs, are activated by alkaline pHo. The INSRR/INSR/IGF1R kinase inhibitor, linsitinib, prevented PKB/Akt activation by alkaline pHo, indicating that INSRRs/INSRs/IGF1Rs are required. Activation of PKB/Akt in cardiomyocytes by α1-AR agonists was also inhibited by linsitinib. Furthermore, linsitinib inhibited cardiomyocyte hypertrophy induced by α1-ARs in cultured cells, reduced the initial cardiac adaptation (24 h) to phenylephrine in vivo (assessed by echocardiography) and increased cardiac fibrosis over 4 days. We conclude that INSRRs are expressed in the heart and, together with INSRs/IGF1Rs, the insulin receptor family provide a potent system for promoting protein synthesis and cardioprotection. Moreover, this system is required for adaptive hypertrophy induced by α1-ARs.
Collapse
|
13
|
Farouk SM, Gad FAM, Emam MA. Comparative immuno-modulatory effects of basil and sesame seed oils against diazinon-induced toxicity in rats; a focus on TNF-α immunolocalization. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:5332-5346. [PMID: 32964385 DOI: 10.1007/s11356-020-10840-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/13/2020] [Indexed: 06/11/2023]
Abstract
Diazinon (DZN), a common organophosphorus insecticide (OPI), has hazardous effect to human and animals with its ubiquitous use. Considering the implication of reactive oxygen species (ROS) in the OPIs toxicity, the present study was aimed to evaluate the ameliorative properties of basil (BO) and sesame (SO) seed oils against the toxic effect of DZN. Forty adult male albino rats were divided into four experimental groups (n = 10 rats/group); control, DZN (10 mg/kg b.w/day), DZN + BO (5 ml/kg b.w/day), and DZN + SO (8 ml/kg b.w/day) groups, treated for a period of 4 weeks. DZN-exposed animals showed significant elevation in serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and creatinine (Cr) with a significant decline in testosterone level compared with control. On the other hand, DZN + BO and DZN + SO groups revealed significant decreases in ALT, AST, BUN, and Cr with a significant increase in testosterone level when compared with DZN-exposed animals. Oxidative/antioxidant indices revealed significant increases of malondialdehyde (MDA) levels along with significant decreases of superoxide dismutase (SOD), glutathione peroxidase (Gpx), and catalase (CAT) activities among DZN-treated rats compared with control. Distinctly lower levels of MDA and increased activities of SOD, Gpx, and CAT were evident in both DZN + BO and DZN + SO groups when compared with DZN-exposed animals. Inflammatory and immuno-modulatory markers assessment showed a significant increase in TNF-α with a significant decline in IL-10 level in DZN group; meanwhile, both DZN + BO and DZN + SO groups revealed significant declines in levels of TNF-α with significant increases in IL-10. Corresponds immunohistochemistry, the total scores (TS) of TNF-α immunostainings in hepatorenal, testicular, and epididymal tissues of control, DZN + BO and DZN + SO groups were significantly lower than those values of DZN group. Additionally, the examined tissues of DZN + BO group revealed significant lower TS of TNF-α immunostaining compared with DZN + SO group. The overall data suggested that both BO and SO can be efficiently used as preventive herbal compounds against DZN-induced oxidative stress with special reference to their possible antioxidant, anti-inflammatory, and free radical activities. However, BO has more potent protective effect against DZN-induced tissue injury at both immunohistochemical and molecular levels.
Collapse
Affiliation(s)
- Sameh Mohamed Farouk
- Cytology and Histology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Fatma Abdel-Monem Gad
- Clinical Pathology Department, Faculty of Veterinary Medicine, Benha University, Banha, 13736, Egypt
| | | |
Collapse
|
14
|
Singh R, Kaundal RK, Zhao B, Bouchareb R, Lebeche D. Resistin induces cardiac fibroblast-myofibroblast differentiation through JAK/STAT3 and JNK/c-Jun signaling. Pharmacol Res 2021; 167:105414. [PMID: 33524540 DOI: 10.1016/j.phrs.2020.105414] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/06/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022]
Abstract
Cardiac fibrosis is characterized by excessive deposition of extracellular matrix proteins and myofibroblast differentiation. Our previous findings have implicated resistin in cardiac fibrosis; however, the molecular mechanisms underlying this process are still unclear. Here we investigated the role of resistin in fibroblast-to-myofibroblast differentiation and elucidated the pathways involved in this process. Fibroblast-to-myofibroblast transdifferentiation was induced with resistin or TGFβ1 in NIH-3T3 and adult cardiac fibroblasts. mRNA and protein expression of fibrotic markers were analyzed by qPCR and immunoblotting. Resistin-knockout mice, challenged with a high-fat diet (HFD) for 20 weeks to stimulate cardiac impairment, were analyzed for cardiac function and fibrosis using histologic and molecular methods. Cardiac fibroblasts stimulated with resistin displayed increased fibroblast-to-myofibroblast conversion, with increased levels of αSma, col1a1, Fn, Ccn2 and Mmp9, with remarkable differences in the actin network appearance. Mechanistically, resistin promotes fibroblast-to-myofibroblast transdifferentiation and fibrogenesis via JAK2/STAT3 and JNK/c-Jun signaling pathways, independent of TGFβ1. Resistin-null mice challenged with HFD showed an improvement in cardiac function and a decrease in tissue fibrosis and reduced mRNA levels of fibrogenic markers. These findings are the first to delineate the role of resistin in the process of cardiac fibroblast-to-myofibroblast differentiation via JAK/STAT3 and JNK/c-Jun pathways, potentially leading to stimulation of cardiac fibrosis.
Collapse
Affiliation(s)
- Rajvir Singh
- Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ravinder K Kaundal
- Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Baoyin Zhao
- Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rihab Bouchareb
- Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Diabetes, Obesity and Metabolism Institute, Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA; Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
15
|
Meijles DN, Cull JJ, Markou T, Cooper STE, Haines ZHR, Fuller SJ, O'Gara P, Sheppard MN, Harding SE, Sugden PH, Clerk A. Redox Regulation of Cardiac ASK1 (Apoptosis Signal-Regulating Kinase 1) Controls p38-MAPK (Mitogen-Activated Protein Kinase) and Orchestrates Cardiac Remodeling to Hypertension. Hypertension 2020; 76:1208-1218. [PMID: 32903101 PMCID: PMC7480944 DOI: 10.1161/hypertensionaha.119.14556] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Supplemental Digital Content is available in the text. Systemic hypertension increases cardiac workload causing cardiomyocyte hypertrophy and increased cardiac fibrosis. An underlying feature is increased production of reactive oxygen species. Redox-sensitive ASK1 (apoptosis signal-regulating kinase 1) activates stress-regulated protein kinases (p38-MAPK [mitogen-activated protein kinases] and JNKs [c-Jun N-terminal kinases]) and promotes fibrosis in various tissues. Here, we determined the specificity of ASK1 signaling in the heart, with the hypothesis that ASK1 inhibitors may be used to manage fibrosis in hypertensive heart disease. Using immunoblotting, we established that moderate levels of H2O2 activate ASK1 in neonatal rat cardiomyocytes and perfused rat hearts. ASK1 was activated during ischemia in adult rat hearts, but not on reperfusion, consistent with activation by moderate (not high) reactive oxygen species levels. In contrast, IL (interleukin)-1β activated an alternative kinase, TAK1 (transforming growth factor–activated kinase 1). ASK1 was not activated by IL1β in cardiomyocytes and activation in perfused hearts was due to increased reactive oxygen species. Selonsertib (ASK1 inhibitor) prevented activation of p38-MAPKs (but not JNKs) by oxidative stresses in cultured cardiomyocytes and perfused hearts. In vivo (C57Bl/6J mice with osmotic minipumps for drug delivery), selonsertib (4 mg/[kg·d]) alone did not affect cardiac function/dimensions (assessed by echocardiography). However, it suppressed hypertension-induced cardiac hypertrophy resulting from angiotensin II (0.8 mg/[kg·d], 7d), with inhibition of Nppa/Nppb mRNA upregulation, reduced cardiomyocyte hypertrophy and, notably, significant reductions in interstitial and perivascular fibrosis. Our data identify a specific reactive oxygen species→ASK1→p38-MAPK pathway in the heart and establish that ASK1 inhibitors protect the heart from hypertension-induced cardiac remodeling. Thus, targeting the ASK1→p38-MAPK nexus has potential therapeutic viability as a treatment for hypertensive heart disease.
Collapse
Affiliation(s)
- Daniel N Meijles
- From the Molecular and Clinical Sciences Institute (D.N.M., S.T.E.C., Z.H.R.H.), St George's University of London, United Kingdom.,School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| | - Joshua J Cull
- School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| | - Thomais Markou
- School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| | - Susanna T E Cooper
- From the Molecular and Clinical Sciences Institute (D.N.M., S.T.E.C., Z.H.R.H.), St George's University of London, United Kingdom
| | | | - Stephen J Fuller
- From the Molecular and Clinical Sciences Institute (D.N.M., S.T.E.C., Z.H.R.H.), St George's University of London, United Kingdom.,School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| | - Peter O'Gara
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom (P.O., S.E.H.)
| | - Mary N Sheppard
- CRY Cardiovascular Pathology Department (M.N.S.), St George's University of London, United Kingdom
| | - Sian E Harding
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom (P.O., S.E.H.)
| | - Peter H Sugden
- School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| | - Angela Clerk
- School of Biological Sciences, University of Reading, United Kingdom (D.N.M., J.J.C., T.M., S.J.F., P.H.S., A.C.), St. George's Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
16
|
Heat Shock Protein 70 Protects the Heart from Ischemia/Reperfusion Injury through Inhibition of p38 MAPK Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3908641. [PMID: 32308802 PMCID: PMC7142395 DOI: 10.1155/2020/3908641] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/14/2020] [Accepted: 03/10/2020] [Indexed: 12/18/2022]
Abstract
Background Heat shock protein 70 (Hsp70) has been shown to exert cardioprotection. Intracellular calcium ([Ca2+]i) overload induced by p38 mitogen-activated protein kinase (p38 MAPK) activation contributes to cardiac ischemia/reperfusion (I/R) injury. However, whether Hsp70 interacts with p38 MAPK signaling is unclear. Therefore, this study investigated the regulation of p38 MAPK by Hsp70 in I/R-induced cardiac injury. Methods Neonatal rat cardiomyocytes were subjected to oxygen-glucose deprivation for 6 h followed by 2 h reoxygenation (OGD/R), and rats underwent left anterior artery ligation for 30 min followed by 30 min of reperfusion. The p38 MAPK inhibitor (SB203580), Hsp70 inhibitor (Quercetin), and Hsp70 short hairpin RNA (shRNA) were used prior to OGD/R or I/R. Cell viability, lactate dehydrogenase (LDH) release, serum cardiac troponin I (cTnI), [Ca2+]i levels, cell apoptosis, myocardial infarct size, mRNA level of IL-1β and IL-6, and protein expression of Hsp70, phosphorylated p38 MAPK (p-p38 MAPK), sarcoplasmic/endoplasmic reticulum Ca2+-ATPase2 (SERCA2), phosphorylated signal transducer and activator of transcription3 (p-STAT3), and cleaved caspase3 were assessed. Results Pretreatment with a p38 MAPK inhibitor, SB203580, significantly attenuated OGD/R-induced cell injury or I/R-induced myocardial injury, as evidenced by improved cell viability and lower LDH release, resulted in lower serum cTnI and myocardial infarct size, alleviation of [Ca2+]i overload and cell apoptosis, inhibition of IL-1β and IL-6, and modulation of protein expressions of p-p38 MAPK, SERCA2, p-STAT3, and cleaved-caspase3. Knockdown of Hsp70 by shRNA exacerbated OGD/R-induced cell injury, which was effectively abolished by SB203580. Moreover, inhibition of Hsp70 by quercetin enhanced I/R-induced myocardial injury, while SB203580 pretreatment reversed the harmful effects caused by quercetin. Conclusions Inhibition of Hsp70 aggravates [Ca2+]i overload, inflammation, and apoptosis through regulating p38 MAPK signaling during cardiac I/R injury, which may help provide novel insight into cardioprotective strategies.
Collapse
|
17
|
Zhou Y, Gao G, Li Z, Jiang L. Protective Effect of Mitogen- and Stress-Activated Protein Kinase on the Rats with Focal Ischemia-Reperfusion Injury. Inflammation 2020; 42:2159-2169. [PMID: 31529230 DOI: 10.1007/s10753-019-01080-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitogen- and stress-activated protein kinase (MSK) is a recently identified nuclear cAMP-regulated enhancer B (CREB) and histone H3 kinase that responds to both mitogen- and stress-activated protein kinases. This study was designed to investigate the protective effect of MSK on the rats with focal ischemia-reperfusion injury. The rat model was established by inserting thread into the middle cerebral artery. The protein expression was measured by immunoblotting. The localization of MSK was measured by immunofluorescence assay. Highly-differentiated pheochromocytoma 12 (PC12) is used as a sympathetic neuron-like cell line and treated with glutamate to induce neurotoxicity. MSK was knocked down and overexpressed by siRNA and MSK over-expressing vector, respectively. The cell viability was measured by cell counting kit (CCK-8) assay. The coronal sections were isolated and stained with 2, 3, 5-triphenyltetrazolium chloride (TTC) to determine infarct volume. Finally, astrocytes were separated from cerebral cortexes of normal rats to analyze the effects of MSK on inflammatory response. In the rats with focal ischemia-reperfusion injury, the expression of MSK was reduced, reaching the lowest level at 3 d after ischemia-reperfusion, and then recovered gradually. MSK was found mainly localized in neurons and astrocytes. The expression levels of caspase-3, caspase-8, caspase-9, and INOS showed the opposite trend with respect to MSK. Further analysis showed that overexpression of MSK exerted a protective effect on glutamate-induced neurotoxicity through inhibiting apoptosis of PC12 cells, as well as decreased the infarct size in rat with focal ischemia-reperfusion injury. On the contrary, knockdown of MSK showed opposite results. Finally, MSK suppressed LPS-induced inflammatory response by decreasing the expression of inducible nitric oxide synthase (INOS) and increasing the expression of interleukin-10 (IL-10) in astrocytes from cerebral cortexes of normal rats. In conclusion, MSK exerted a protective effect on rat with focal ischemia-reperfusion injury through its anti-apoptotic effect on neurons and anti-inflammatory effect on astrocytes.
Collapse
Affiliation(s)
- Yanfeng Zhou
- Departments of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow, 215006, China.,Departments of Neurosurgery, The First People's Hospital of Taizhou, Taizhou, 225300, China
| | - Guangzhong Gao
- Departments of Neurosurgery, The First People's Hospital of Taizhou, Taizhou, 225300, China
| | - Zhen Li
- Departments of Neurosurgery, The First People's Hospital of Taizhou, Taizhou, 225300, China
| | - Lin Jiang
- Departments of Neurosurgery, The First People's Hospital of Taizhou, Taizhou, 225300, China.
| |
Collapse
|
18
|
Qiu L, Liu X. Identification of key genes involved in myocardial infarction. Eur J Med Res 2019; 24:22. [PMID: 31269974 PMCID: PMC6607516 DOI: 10.1186/s40001-019-0381-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
Background This study focuses on the identification of conserved genes involved in myocardial infarction (MI), and then analyzed the differentially expressed genes (DEGs) between the incident and recurrent events to identify MI-recurrent biomarkers. Methods Gene expression data of MI peripheral blood were downloaded from GSE97320 and GSE66360 datasets. We identified the common DEGs in these two datasets by functional enrichment analysis and protein–protein interaction (PPI) network analysis. GSE48060 was further analyzed to validate the conserved genes in MI and to compare the DEGs between the incident and recurrent MI. Results A total of 477 conserved genes were identified in the comparison between MI and control. Protein–protein interaction (PPI) network showed hub genes, such as MAPK14, STAT3, and MAPKAPK2. Part of those conserved genes was validated in the analysis of GSE48060. The DEGs in the incident and recurrent MI showed significant differences, including RNASE2 and A2M-AS1 as the potential biomarkers of MI recurrence. Conclusions The conserved genes in the pathogenesis of MI were identified, benefit for target therapy. Meanwhile, some specific genes may be used as markers for the prediction of recurrent MI. Electronic supplementary material The online version of this article (10.1186/s40001-019-0381-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Linlin Qiu
- Danyang People's Hospital of Jiangsu Province, Danyang, China
| | - Xueqing Liu
- Danyang People's Hospital of Jiangsu Province, Danyang, China.
| |
Collapse
|
19
|
Hirata M, Yao T, Fujimura S, Kanai Y, Yoshimoto M, Sato T, Ohmomo Y, Temma T. Development of a p38α-selective radioactive probe for qualitative diagnosis of cancer using SPECT. Ann Nucl Med 2019; 33:333-343. [PMID: 30953245 DOI: 10.1007/s12149-019-01341-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/23/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVE p38 mitogen-activated protein (MAP) kinase (p38α) has drawn attention as a new target molecule for the treatment and diagnosis of cancer, and its overexpression and activation have been reported in various types of cancer. In this study, a single photon emission computed tomography (SPECT) imaging probe of p38α was developed to noninvasively image p38α activity for effective qualitative diagnosis of cancer. METHODS Pyrrolepyridine derivatives, m-YTM and p-YTM, were designed and synthesized based on the structure of the p38α-selective inhibitor. Radioactive iodine-labeled m-YTM, [125I]m-YTM, was synthesized because m-YTM greatly inhibited the phosphorylation of p38α upon examining the inhibitory effects of the compounds. After investigating the binding affinity of [125I]m-YTM to the recombinant p38α, a saturation binding experiment using activated p38α and inactive p38α was performed to determine the binding site. Uptake of [125I]m-YTM into various cancer cell lines was investigated, and the pharmacokinetics was evaluated using tumor-bearing mice. RESULTS The inhibitory activity of m-YTM was approximately 13 times higher than that of SB203580, a p38α-selective inhibitor. The binding site of [125I]m-YTM was estimated to be the p38α activating site, similar to that of SB203580, because the [125I]m-YTM bound strongly to both activated p38α and inactive p38α. Various different cancer cells incorporated [125I]m-YTM; however, its accumulation was significantly reduced by treatment with SB203580. Pharmacokinetics study of [125I]m-YTM in B-16 tumor-bearing mice was examined which revealed high accumulation of radioactivity in tumor tissues. The ratios of radioactivity in the B-16 tumor to that in blood were 3.1 and 50 after 1 and 24 h, respectively. The ratio of radioactivity in the tumor to that in blood in the tumor-bearing mice generated using other cancer cell lines was also ≥ 1 at 1 h after the administration of the probe. CONCLUSIONS This study suggests that [123I]m-YTM has potential as a p38α imaging probe effective for various cancer types.
Collapse
Affiliation(s)
- Masahiko Hirata
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Tatsuma Yao
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Shigeaki Fujimura
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Yasukazu Kanai
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Mitsuyoshi Yoshimoto
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.,Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Takaji Sato
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Yoshiro Ohmomo
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Takashi Temma
- Department of Biofunctional Analysis, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| |
Collapse
|
20
|
Reyes L, Hawkins CL, Rayner BS. Characterization of the cellular effects of myeloperoxidase-derived oxidants on H9c2 cardiac myoblasts. Arch Biochem Biophys 2019; 665:132-142. [DOI: 10.1016/j.abb.2019.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
|
21
|
Nawaito SA, Sahadevan P, Clavet-Lanthier MÉ, Pouliot P, Sahmi F, Shi Y, Gillis MA, Lesage F, Gaestel M, Sirois MG, Calderone A, Tardif JC, Allen BG. MK5 haplodeficiency decreases collagen deposition and scar size during post-myocardial infarction wound repair. Am J Physiol Heart Circ Physiol 2019; 316:H1281-H1296. [PMID: 30901279 DOI: 10.1152/ajpheart.00532.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
MK5 is a protein serine/threonine kinase activated by p38, ERK3, and ERK4 MAPKs. MK5 mRNA and immunoreactivity are detected in mouse cardiac fibroblasts, and MK5 haplodeficiency attenuates the increase in collagen 1-α1 mRNA evoked by pressure overload. The present study examined the effect of MK5 haplodeficiency on reparative fibrosis following myocardial infarction (MI). Twelve-week-old MK5+/- and wild-type littermate (MK5+/+) mice underwent ligation of the left anterior descending coronary artery (LADL). Surviving mice were euthanized 8 or 21 days post-MI. Survival rates did not differ significantly between MK5+/+ and MK5+/- mice, with rupture of the LV wall being the primary cause of death. Echocardiographic imaging revealed similar increases in LV end-diastolic diameter, myocardial performance index, and wall motion score index in LADL-MK5+/+ and LADL-MK5+/- mice. Area at risk did not differ between LADL-MK5+/+ and LADL-MK5+/- hearts. In contrast, infarct size, scar area, and scar collagen content were reduced in LADL-MK5+/- hearts. Immunohistochemical analysis of mice experiencing heart rupture revealed increased MMP-9 immunoreactivity in the infarct border zone of LADL-MK5+/- hearts compared with LADL-MK5+/+. Although inflammatory cell infiltration was similar in LADL-MK5+/+ and LADL-MK5+/- hearts, angiogenesis was more pronounced in the infarct border zone of LADL-MK5+/- mice. Characterization of ventricular fibroblasts revealed reduced motility and proliferation in fibroblasts isolated from MK5-/- mice compared with those from both wild-type and haplodeficient mice. siRNA-mediated knockdown of MK5 in fibroblasts from wild-type mice also impaired motility. Hence, reduced MK5 expression alters fibroblast function and scar morphology but not mortality post-MI. NEW & NOTEWORTHY MK5/PRAK is a protein serine/threonine kinase activated by p38 MAPK and/or atypical MAPKs ERK3/4. MK5 haplodeficiency reduced infarct size, scar area, and scar collagen content post-myocardial infarction. Motility and proliferation were reduced in cultured MK5-null cardiac myofibroblasts.
Collapse
Affiliation(s)
- Sherin Ali Nawaito
- Department of Pharmacology and Physiology, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada.,Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Pramod Sahadevan
- Department of Biochemistry and Molecular Medicine, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| | | | | | - Fatiha Sahmi
- Montreal Heart Institute , Montreal, Quebec, Canada
| | - Yanfen Shi
- Montreal Heart Institute , Montreal, Quebec, Canada
| | | | - Frederic Lesage
- Department of Electrical Engineering, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| | - Matthias Gaestel
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany
| | - Martin G Sirois
- Department of Pharmacology and Physiology, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| | - Angelo Calderone
- Department of Pharmacology and Physiology, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| | - Bruce G Allen
- Department of Biochemistry and Molecular Medicine, Université de Montréal , Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal , Montreal, Quebec, Canada.,Montreal Heart Institute , Montreal, Quebec, Canada
| |
Collapse
|
22
|
Connexin43 dephosphorylation at serine 282 is associated with connexin43-mediated cardiomyocyte apoptosis. Cell Death Differ 2019; 26:1332-1345. [PMID: 30770876 DOI: 10.1038/s41418-019-0277-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
Gap junction protein connexin 43 (Cx43) plays an important role in regulating cardiomyocyte survival in addition to regulating electrical coordination. Cx43 dephosphorylation, found in severe cardiac pathologies, is thought to contribute to myocardial injury. However, the mechanisms underlying Cx43 mediation of cell survival and myocardial lesions remain unknown. Here, we found that transfecting an adenovirus carrying a mutant gene of Cx43-serine 282 substituted with alanine (S282A) into neonatal rat ventricular myocytes (NRVMs) induced cell apoptosis and Ca2+ transient desynchronization, whereas using gap junction inhibitor or knocking down Cx43 expression with Cx43-miRNA caused uncoupled Ca2+ signaling without cell death. Similarly, while Cx43-S282A+/+ failed in generation, Cx43-S282A+/- mice exhibited cardiomyocyte apoptosis and ventricular arrhythmias dependent on S282 dephosphorylation. Further, Cx43 dephosphorylation at S282 activated p38 mitogen-activated protein kinase (p38 MAPK), factor-associated suicide and the caspase-8 apoptotic pathway by physically interacting with p38 MAPK. These findings uncovered a specific Cx43 phosphorylation residue involved in regulating cardiomyocyte homeostasis. S282 phosphorylation deficiency acts as a trigger inducing cardiomyocyte apoptosis and cardiac arrhythmias, providing a potential mechanism for Cx43-mediated myocardial injury in severe cardiac diseases.
Collapse
|
23
|
|
24
|
Effects of Anti-Inflammatory Medications in Patients With Coronary Artery Disease: A Focus on Losmapimod. Cardiol Rev 2018; 26:152-156. [DOI: 10.1097/crd.0000000000000176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Transcriptional and Post-Translational Targeting of Myocyte Stress Protein 1 (MS1) by the JNK Pathway in Cardiac Myocytes. J Mol Signal 2017; 12:3. [PMID: 30210579 PMCID: PMC5853832 DOI: 10.5334/1750-2187-12-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myocyte Stress Protein 1 (MS1) is a muscle-specific, stress-responsive, regulator of gene expression. It was originally identified in embryonic mouse heart which showed increased expression in a rat model of left ventricular hypertrophy. To determine if MS1 was responsive to other stresses relevant to cardiac myocyte function, we tested if it could be induced by the metabolic stresses associated with ischaemia/reperfusion injury in cardiac myocytes. We found that metabolic stress increased MS1 expression, both at the mRNA and protein level, concurrent with activation of the c-Jun N-terminal Kinase (JNK) signalling pathway. MS1 induction by metabolic stress was blocked by both the transcription inhibitor actinomycin D and a JNK inhibitor, suggesting that activation of the JNK pathway during metabolic stress in cardiac myocytes leads to transcriptional induction of MS1. MS1 was also found to be an efficient JNK substrate in vitro, with a major JNK phosphorylation site identified at Thr-62. In addition, MS1 was found to co-precipitate with JNK, and inspection of the amino acid sequence upstream of the phosphorylation site, at Thr-62, revealed a putative Mitogen-Activated Protein Kinase (MAPK) binding site. Taken together, these data identify MS1 as a likely transcriptional and post-translational target for the JNK pathway in cardiac myocytes subjected to metabolic stress.
Collapse
|
26
|
Luo Y, Shang P, Li D. Luteolin: A Flavonoid that Has Multiple Cardio-Protective Effects and Its Molecular Mechanisms. Front Pharmacol 2017; 8:692. [PMID: 29056912 PMCID: PMC5635727 DOI: 10.3389/fphar.2017.00692] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 09/19/2017] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease (CVD) has become the leading cause of morbidity and mortality worldwide. A well-monitored diet with a sufficient intake of fruits and vegetables has been confirmed as a primary prevention of CVD. Plant constituents such as flavonoids have been shown to confer healthy benefits. Luteolin (Lut), a kind of flavonoid, possesses anti-oxidative, anti-tumor, and anti-inflammatory properties. Recent scientific literature has reported the cardiac protective effects of Lut in vitro and in vivo. Therefore, the aim of this review is to provide an update and detailed overview with cardio-protective molecular mechanisms of Lut with a focus on multiple intrinsic and extrinsic effectors. We further explore how these mechanisms participate in ischemia/reperfusion (I/R) injury, heart failure (HF) and atherosclerosis (AS). A proper understanding of the cardiovascular protective effects and the relative mechanisms of Lut may provide the possibility of new drug design and development for CVD. With the previous studies mainly focused on basic research, we need to advance the prospects of its further clinical utilization against CVD, large prospective clinical trials of Lut are needed to observe its therapeutic effects on patients with I/R injury, HF and AS, especially on the effective therapeutic dosage, and safety of long-term administration.
Collapse
Affiliation(s)
- Yuanyuan Luo
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Pingping Shang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Dongye Li
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
27
|
Lee SM, Hutchinson M, Saint DA. The role of Toll-like receptor 4 (TLR4) in cardiac ischaemic-reperfusion injury, cardioprotection and preconditioning. Clin Exp Pharmacol Physiol 2017; 43:864-71. [PMID: 27249055 DOI: 10.1111/1440-1681.12602] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/26/2016] [Accepted: 05/30/2016] [Indexed: 01/04/2023]
Abstract
Cardiac ischaemic-reperfusion injury (IRI) remains the primary cause of mortality throughout the developed world. Molecular mechanisms underlying IRI are complex and are often interlinked with each other driving a synergistic response. Toll-like receptor 4 (TLR4), an immunosurveillance receptor, is known to enhance tissue injury during IRI by enhancing the inflammatory response. The release of endogenous components during IRI bind onto TLR4 leading to the activation of multiple signalling kinases. Once this event occurs these proteins are defined as danger associated molecular patterns molecules (DAMPs) or alarmins. Examples include heat shock proteins, high mobility group box one (HMGB1) and extracellular matrix proteins, all of which are involved in IRI. However, literature in the last two decades suggests that transient stimulation of TLR4 may suppress IRI and thus improve cardiac recovery. Furthermore, it remains to be seen what role TLR4 plays during ischaemic-preconditioning where acute bouts of ischaemia, preceding a harmful bout of ischaemic-reperfusion, is cardioprotective. The other question which also needs to be considered is that if transient TLR4 signalling drives a preconditioning response then what are the ligands which drive this? Hence the second part of this review explores the possible TLR4 ligands which may promote cardioprotection against IRI.
Collapse
Affiliation(s)
- Sam Man Lee
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Mark Hutchinson
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA, Australia
| | - David A Saint
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
28
|
Wen SY, Tamilselvi S, Shen CY, Day CH, Chun LC, Cheng LY, Ou HC, Chen RJ, Viswanadha VP, Kuo WW, Huang CY. Protective effect of HDL on NADPH oxidase-derived super oxide anion mediates hypoxia-induced cardiomyocyte apoptosis. PLoS One 2017; 12:e0179492. [PMID: 28617849 PMCID: PMC5472312 DOI: 10.1371/journal.pone.0179492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/31/2017] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death of death in Taiwan. Atherosclerosis can lead to serious problems, including heart attack, stroke, or even death. Coronary heart disease (CHD) occurs when plaque builds up in the coronary arteries to cause the ischemic heart disease which will enhance myocardial remodeling and also induce myocardial hypoxia. High density lipoprotein (HDL) has been proposed to have cardio-protective effects. Under hypoxic conditions (1%O2 for 24hr), in H9c2 cells, reactive oxygen species (ROS) is induced which leads to cardiomyocyte apoptosis and cardiac dysfunction. Therefore, the present study described the protective effect of HDL on hypoxia-induced cardiomyocyte damage. We investigated the NADPH oxidase-produced ROS-related signaling pathways and apoptosis in cardiomyocytes under hypoxia conditions. Results showed that the ROS mediated cardiac damage might occur via AT1 and PKC activation. Furthermore, hypoxia downregulated the survival protein (p-AKTser473) and anti-apoptotic protein (BCL2), whereas pro-apoptotic protein, Bax and caspase 3 were upregulated. These detrimental effects by ROS and apoptosis were prevented by HDL pretreatment. Our findings revealed the underlying molecular mechanism by which HDL suppresses the hypoxia-induced cardiomyocyte dysfunction. Further, we elucidated the role of HDL on preventing hypoxia induced cardiomyocyte apoptosis is mediated through the inhibition of NADPH oxidase-derived ROS.
Collapse
Affiliation(s)
- Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- Center for General Education, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Shanmugam Tamilselvi
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chia-Yao Shen
- Department of Nursing, MeiHo University, Pingtung, Taiwan
| | | | - Li-Chin Chun
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy & Science, Tainan County, Taiwan
| | - Li-Yi Cheng
- Department of Biological Science and Technology, Asia University, Taichung, Taiwan
| | - Hsiu-Chung Ou
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biological Science and Technology, Asia University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
29
|
Corre I, Paris F, Huot J. The p38 pathway, a major pleiotropic cascade that transduces stress and metastatic signals in endothelial cells. Oncotarget 2017; 8:55684-55714. [PMID: 28903453 PMCID: PMC5589692 DOI: 10.18632/oncotarget.18264] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022] Open
Abstract
By gating the traffic of molecules and cells across the vessel wall, endothelial cells play a central role in regulating cardiovascular functions and systemic homeostasis and in modulating pathophysiological processes such as inflammation and immunity. Accordingly, the loss of endothelial cell integrity is associated with pathological disorders that include atherosclerosis and cancer. The p38 mitogen-activated protein kinase (MAPK) cascades are major signaling pathways that regulate several functions of endothelial cells in response to exogenous and endogenous stimuli including growth factors, stress and cytokines. The p38 MAPK family contains four isoforms p38α, p38β, p38γ and p38δ that are encoded by four different genes. They are all widely expressed although to different levels in almost all human tissues. p38α/MAPK14, that is ubiquitously expressed is the prototype member of the family and is referred here as p38. It regulates the production of inflammatory mediators, and controls cell proliferation, differentiation, migration and survival. Its activation in endothelial cells leads to actin remodeling, angiogenesis, DNA damage response and thereby has major impact on cardiovascular homeostasis, and on cancer progression. In this manuscript, we review the biology of p38 in regulating endothelial functions especially in response to oxidative stress and during the metastatic process.
Collapse
Affiliation(s)
- Isabelle Corre
- CRCINA, INSERM, CNRS, Université de Nantes, Nantes, France
| | - François Paris
- CRCINA, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Jacques Huot
- Le Centre de Recherche du CHU de Québec-Université Laval et le Centre de Recherche sur le Cancer de l'Université Laval, Québec, Canada
| |
Collapse
|
30
|
Zhu S, Xu T, Luo Y, Zhang Y, Xuan H, Ma Y, Pan D, Li D, Zhu H. Luteolin Enhances Sarcoplasmic Reticulum Ca2+-ATPase Activity through p38 MAPK Signaling thus Improving Rat Cardiac Function after Ischemia/Reperfusion. Cell Physiol Biochem 2017; 41:999-1010. [DOI: 10.1159/000460837] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/01/2016] [Indexed: 01/15/2023] Open
Abstract
Background/Aims: A major challenge for current therapeutic strategies against ischemia/reperfusion (I/R) is the lack of effective drugs. Considering luteolin enhances the activity of sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) to improve the systolic/diastolic function of rat hearts and cardiomyocytes during the I/R process, we studied the regulatory function of the p38 MAPK pathway in this protective mechanism. Methods: Isolated cardiomyocytes and perfused hearts were separately divided into five groups and used to investigate I/R. The phosphorylation of p38 and phospholamban (p-PLB), the levels and activity of SERCA2a and the levels of proteins related to apoptosis were measured. Apoptotic cells were assessed using the TUNEL assay. Single-cell shortening, Ca2+ transients, and the decay of the mitochondrial membrane potential (Δψm) were detected. Results: The p38 MAPK pathway was activated during the I/R process, and inhibiting it with SB203580 promoted p-PLB, which enhanced the activity of SERCA2a and relieved the calcium overload to promote the recovery of the Δψm and reduce cardiomyocyte apoptosis in I/R. Luteolin also suppressed the activation of the p38 MAPK pathway and showed cardioprotective effects during I/R injury. Conclusions: We conclude that luteolin enhances SERCA2a activity to improve systolic/diastolic function during I/R in rat hearts and cardiomyocytes by attenuating the inhibitive effects of the p38 pathway on p-PLB.
Collapse
|
31
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
32
|
Su F, Myers VD, Knezevic T, Wang J, Gao E, Madesh M, Tahrir FG, Gupta MK, Gordon J, Rabinowitz J, Ramsey FV, Tilley DG, Khalili K, Cheung JY, Feldman AM. Bcl-2-associated athanogene 3 protects the heart from ischemia/reperfusion injury. JCI Insight 2016; 1:e90931. [PMID: 27882354 DOI: 10.1172/jci.insight.90931] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bcl-2-associated athanogene 3 (BAG3) is an evolutionarily conserved protein expressed at high levels in the heart and the vasculature and in many cancers. While altered BAG3 expression has been associated with cardiac dysfunction, its role in ischemia/reperfusion (I/R) is unknown. To test the hypothesis that BAG3 protects the heart from reperfusion injury, in vivo cardiac function was measured in hearts infected with either recombinant adeno-associated virus serotype 9-expressing (rAAV9-expressing) BAG3 or GFP and subjected to I/R. To elucidate molecular mechanisms by which BAG3 protects against I/R injury, neonatal mouse ventricular cardiomyocytes (NMVCs) in which BAG3 levels were modified by adenovirus expressing (Ad-expressing) BAG3 or siBAG3 were exposed to hypoxia/reoxygenation (H/R). H/R significantly reduced NMVC BAG3 levels, which were associated with enhanced expression of apoptosis markers, decreased expression of autophagy markers, and reduced autophagy flux. The deleterious effects of H/R on apoptosis and autophagy were recapitulated by knockdown of BAG3 with Ad-siBAG3 and were rescued by Ad-BAG3. In vivo, treatment of mice with rAAV9-BAG3 prior to I/R significantly decreased infarct size and improved left ventricular function when compared with mice receiving rAAV9-GFP and improved markers of autophagy and apoptosis. These findings suggest that BAG3 may provide a therapeutic target in patients undergoing reperfusion after myocardial infarction.
Collapse
Affiliation(s)
- Feifei Su
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania USA.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Valerie D Myers
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania USA
| | | | | | - Erhe Gao
- Center for Translational Medicine, and
| | | | | | | | | | | | - Frederick V Ramsey
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania USA
| | | | | | - Joseph Y Cheung
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania USA.,Center for Translational Medicine, and
| | - Arthur M Feldman
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania USA
| |
Collapse
|
33
|
Hackfort BT, Mishra PK. Emerging role of hydrogen sulfide-microRNA crosstalk in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2016; 310:H802-12. [PMID: 26801305 PMCID: PMC4867357 DOI: 10.1152/ajpheart.00660.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/18/2016] [Indexed: 12/15/2022]
Abstract
Despite an obnoxious smell and toxicity at a high dose, hydrogen sulfide (H2S) is emerging as a cardioprotective gasotransmitter. H2S mitigates pathological cardiac remodeling by regulating several cellular processes including fibrosis, hypertrophy, apoptosis, and inflammation. These encouraging findings in rodents led to initiation of a clinical trial using a H2S donor in heart failure patients. However, the underlying molecular mechanisms by which H2S mitigates cardiac remodeling are not completely understood. Empirical evidence suggest that H2S may regulate signaling pathways either by directly influencing a gene in the cascade or interacting with nitric oxide (another cardioprotective gasotransmitter) or both. Recent studies revealed that H2S may ameliorate cardiac dysfunction by up- or downregulating specific microRNAs. MicroRNAs are noncoding, conserved, regulatory RNAs that modulate gene expression mostly by translational inhibition and are emerging as a therapeutic target for cardiovascular disease (CVD). Few microRNAs also regulate H2S biosynthesis. The inter-regulation of microRNAs and H2S opens a new avenue for exploring the H2S-microRNA crosstalk in CVD. This review embodies regulatory mechanisms that maintain the physiological level of H2S, exogenous H2S donors used for increasing the tissue levels of H2S, H2S-mediated regulation of CVD, H2S-microRNAs crosstalk in relation to the pathophysiology of heart disease, clinical trials on H2S, and future perspectives for H2S as a therapeutic agent for heart failure.
Collapse
Affiliation(s)
- Bryan T Hackfort
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
34
|
Bi QR, Hou JJ, Qi P, Ma CH, Feng RH, Yan BP, Wang JW, Shi XJ, Zheng YY, Wu WY, Guo DA. TXNIP/TRX/NF-κB and MAPK/NF-κB pathways involved in the cardiotoxicity induced by Venenum Bufonis in rats. Sci Rep 2016; 6:22759. [PMID: 26961717 PMCID: PMC4785358 DOI: 10.1038/srep22759] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/04/2016] [Indexed: 01/24/2023] Open
Abstract
Venenum Bufonis (VB) is a widely used traditional medicine with serious cardiotoxic effects. The inflammatory response has been studied to clarify the mechanism of the cardiotoxicity induced by VB for the first time. In the present study, Sprague Dawley (SD) rats, were administered VB (100, 200, and 400 mg/kg) intragastrically, experienced disturbed ECGs (lowered heart rate and elevated ST-segment), increased levels of serum indicators (creatine kinase (CK), creatine kinase isoenzyme-MB (CK-MB), alanine aminotransferase (ALT), aspartate aminotransferase (AST)) and serum interleukin (IL-6, IL-1β, TNF-α) at 2 h, 4 h, 6 h, 8 h, 24 h, and 48 h, which reflected that an inflammatory response, together with cardiotoxicity, were involved in VB-treated rats. In addition, the elevated serum level of MDA and the down-regulated SOD, CAT, GSH, and GPx levels indicated the appearance of oxidative stress in the VB-treated group. Furthermore, based on the enhanced expression levels of TXNIP, p-NF-κBp65, p-IκBα, p-IKKα, p-IKKβ, p-ERK, p-JNK, and p-P38 and the obvious myocardial degeneration, it is proposed that VB-induced cardiotoxicity may promote an inflammatory response through the TXNIP/TRX/NF-κB and MAPK/NF-κB pathways. The observed inflammatory mechanism induced by VB may provide a theoretical reference for the toxic effects and clinical application of VB.
Collapse
Affiliation(s)
- Qi-Rui Bi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China.,College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Jin-Jun Hou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| | - Peng Qi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| | - Chun-Hua Ma
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Rui-Hong Feng
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| | - Bing-Peng Yan
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China.,College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Jian-Wei Wang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China.,College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Jian Shi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China.,College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan-Yuan Zheng
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| | - Wan-Ying Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Haike Road 501, Shanghai 201203, China
| |
Collapse
|
35
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
36
|
Song X, Wei Z, Shaikh ZA. Requirement of ERα and basal activities of EGFR and Src kinase in Cd-induced activation of MAPK/ERK pathway in human breast cancer MCF-7 cells. Toxicol Appl Pharmacol 2015; 287:26-34. [PMID: 26006730 PMCID: PMC4490940 DOI: 10.1016/j.taap.2015.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/26/2015] [Accepted: 05/13/2015] [Indexed: 12/29/2022]
Abstract
Cadmium (Cd) is a common environmental toxicant and an established carcinogen. Epidemiological studies implicate Cd with human breast cancer. Low micromolar concentrations of Cd promote proliferation of human breast cancer cells in vitro. The growth promotion of breast cancer cells is associated with the activation of MAPK/ERK pathway. This study explores the mechanism of Cd-induced activation of MAPK/ERK pathway. Specifically, the role of cell surface receptors ERα, EGFR, and Src kinase was evaluated in human breast cancer MCF-7 cells treated with 1-3μM Cd. The activation of ERK was studied using a serum response element (SRE) luciferase reporter assay. Receptor phosphorylation was detected by Western blot analyses. Cd treatment increased both the SRE reporter activity and ERK1/2 phosphorylation in a concentration-dependent manner. Cd treatment had no effect on reactive oxygen species (ROS) generation. Also, blocking the entry of Cd into the cells with manganese did not diminish Cd-induced activation of MAPK/ERK. These results suggest that the effect of Cd was likely not caused by intracellular ROS generation, but through interaction with the membrane receptors. While Cd did not appear to activate either EGFR or Src kinase, their inhibition completely blocked the Cd-induced activation of ERK as well as cell proliferation. Similarly, silencing ERα with siRNA or use of ERα antagonist blocked the effects of Cd. Based on these results, it is concluded that not only ERα, but also basal activities of EGFR and Src kinase are essential for Cd-induced signal transduction and activation of MAPK/ERK pathway for breast cancer cell proliferation.
Collapse
Affiliation(s)
- Xiulong Song
- Center for Molecular Toxicology, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Zhengxi Wei
- Center for Molecular Toxicology, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Zahir A Shaikh
- Center for Molecular Toxicology, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
37
|
Abstract
It is well established that cardiac remodeling plays a pivotal role in the development of heart failure, a leading cause of death worldwide. Meanwhile, sympathetic hyperactivity is an important factor in inducing cardiac remodeling. Therefore, an in-depth understanding of beta-adrenoceptor signaling pathways would help to find better ways to reverse the adverse remodeling. Here, we reviewed five pathways, namely mitogen-activated protein kinase signaling, Gs-AC-cAMP signaling, Ca(2+)-calcineurin-NFAT/CaMKII-HDACs signaling, PI3K signaling and beta-3 adrenergic signaling, in cardiac remodeling. Furthermore, we constructed a cardiac-remodeling-specific regulatory network including miRNA, transcription factors and target genes within the five pathways. Both experimental and clinical studies have documented beneficial effects of beta blockers in cardiac remodeling; nevertheless, different blockers show different extent of therapeutic effect. Exploration of the underlying mechanisms could help developing more effective drugs. Current evidence of treatment effect of beta blockers in remodeling was also reviewed based upon information from experimental data and clinical trials. We further discussed the mechanism of how beta blockers work and why some beta blockers are more potent than others in treating cardiac remodeling within the framework of cardiac remodeling network.
Collapse
|
38
|
Lee S, Hutchinson M, Staikopoulos V, Saint D. Amitriptyline pharmacologically preconditions rat hearts against cardiac ischemic–reperfusion injury. Int J Cardiol 2015; 190:353-9. [DOI: 10.1016/j.ijcard.2015.04.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/16/2015] [Accepted: 04/03/2015] [Indexed: 11/16/2022]
|
39
|
Activated regulatory T-cells attenuate myocardial ischaemia/reperfusion injury through a CD39-dependent mechanism. Clin Sci (Lond) 2015; 128:679-93. [PMID: 25558978 DOI: 10.1042/cs20140672] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Regulatory T-cells (Tregs) are generally regarded as key immunomodulators that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. However, its role in myocardial ischaemia/reperfusion injury (MIRI) remains unknown. The purpose of the present study was to determine whether Tregs exert a beneficial effect on mouse MIRI. We examined the role of Tregs in murine MIRI by depletion using 'depletion of regulatory T-cell' (DEREG) mice and adoptive transfer using Forkhead box P3 (Foxp3)-GFP knockin mice and the mechanisms of cardio protection were further studied in vivo and in vitro. Tregs rapidly accumulated in murine hearts following MIRI. Selective depletion of Tregs in the DEREG mouse model resulted in aggravated MIRI. In contrast, the adoptive transfer of in vitro-activated Tregs suppressed MIRI, whereas freshly isolated Tregs had no effect. Mechanistically, activated Treg-mediated protection against MIRI was not abrogated by interleukin (IL)-10 or transforming growth factor (TGF)-β1 inhibition but was impaired by the genetic deletion of cluster of differentiation 39 (CD39). Moreover, adoptive transfer of in vitro-activated Tregs attenuated cardiomyocyte apoptosis, activated a pro-survival pathway involving Akt and extracellular-signal-regulated kinase (ERK) and inhibited neutrophil infiltration, which was compromised by CD39 deficiency. Finally, the peripheral blood mononuclear cells of acute myocardial infarction (AMI) patients after primary percutaneous coronary intervention (PCI) revealed a decrease in CD4+CD25+CD127low Tregs and a relative increase in CD39+ cells within the Treg population. In conclusion, our data validated a protective role for Tregs in MIRI. Moreover, in vitro-activated Tregs ameliorated MIRI via a CD39-dependent mechanism, representing a putative therapeutic strategy.
Collapse
|
40
|
Kang YJ, Bang BR, Otsuka M, Otsu K. Tissue-Specific Regulation of p38α-Mediated Inflammation in Con A-Induced Acute Liver Damage. THE JOURNAL OF IMMUNOLOGY 2015; 194:4759-66. [PMID: 25888643 DOI: 10.4049/jimmunol.1402954] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/16/2015] [Indexed: 01/19/2023]
Abstract
Because p38α plays a critical role in inflammation, it has been an attractive target for the development of anti-inflammation therapeutics. However, p38α inhibitors showed side effects, including severe liver toxicity, that often prevailed over the benefits in clinical studies, and the mechanism of toxicity is not clear. In this study, we demonstrate that p38α regulates the inflammatory responses in acute liver inflammation in a tissue-specific manner, and liver toxicity by p38α inhibitors may be a result of the inhibition of protective activity of p38α in the liver. Genetic ablation of p38α in T and NKT cells protected mice from liver injury in Con A-induced liver inflammation, whereas liver-specific deletion of p38α aggravated liver pathology. We found that p38α deficiency in the liver increased the expression of chemokines to recruit more inflammatory cells, indicating that p38α in the liver plays a protective anti-inflammatory role during acute liver inflammation. Therefore, our results suggest that p38α regulates the inflammatory responses in a tissue-specific manner, and that the tissue-specific p38α targeting strategies can be used for the development of an effective anti-inflammation treatment with an improved side-effect profile.
Collapse
Affiliation(s)
- Young Jun Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037;
| | - Bo-Ram Bang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Kinya Otsu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; and Cardiovascular Division, King's College London, London SE5 9NU, United Kingdom
| |
Collapse
|
41
|
Kumphune S, Surinkaew S, Chattipakorn SC, Chattipakorn N. Inhibition of p38 MAPK activation protects cardiac mitochondria from ischemia/reperfusion injury. PHARMACEUTICAL BIOLOGY 2015; 53:1831-1841. [PMID: 25880145 DOI: 10.3109/13880209.2015.1014569] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Cardiac cell death and fatal arrhythmias during myocardial ischemia/reperfusion (I/R) can be reduced by p38 MAPK inhibition. However, the effects of p38 MAPK inhibition on cardiac mitochondria have not been investigated. OBJECTIVE We tested the hypothesis that p38 MAPK inhibition at different times during I/R protects cardiac mitochondrial functions. MATERIALS AND METHODS Adult Wistar rats were subjected to 30 min of left anterior descending coronary artery (LAD) occlusion, followed by 120 min of reperfusion. A 2 mg/kg bolus infusion of p38 MAPK inhibitor, SB203580, was given before or during ischemia, or at reperfusion. Mitochondrial function and ultrastructure were assessed and Western blots were performed. RESULTS Administration of SB203580 at any time point of I/R significantly attenuated the mitochondrial ultrastructure change, mitochondrial swelling, by increasing the absorbance at 540 nm (I/R control 0.42 ± 0.03; pretreatment 0.58 ± 0.04; during ischemia 0.49 ± 0.02; at reperfusion 0.51 ± 0.02, p < 0.05), similar to reactive oxygen species (ROS) generation (I/R control 1300 ± 48; pretreatment 1150 ± 30; during ischemia 1000 ± 50; at reperfusion 1050 ± 55, p < 0.05). Only SB203580 given before or during ischemia attenuated mitochondrial membrane depolarization (I/R control 0.78 ± 0.04; pretreatment 1.02 ± 0.03; during ischemia 1.05 ± 0.12, p < 0.05). In addition, pre-treatment of SB203580 significantly reduced the phosphorylation of p53, CREB, Bax, cytochrome c, and cleaved caspase 3. DISCUSSION AND CONCLUSION The results from this study showed for the first time that p38 MAPK inhibition protects mitochondria from I/R injury.
Collapse
Affiliation(s)
- Sarawut Kumphune
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University , Chiang Mai , Thailand
| | | | | | | |
Collapse
|
42
|
Jeon S, Hur J, Kim J. DHEA Alleviates Oxidative Stress of Muscle Cells via Activation of Nrf2 Pathway. Appl Biochem Biotechnol 2015; 176:22-32. [DOI: 10.1007/s12010-015-1500-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/19/2015] [Indexed: 11/24/2022]
|
43
|
Blood-brain barrier Na transporters in ischemic stroke. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:113-46. [PMID: 25307215 DOI: 10.1016/bs.apha.2014.06.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood-brain barrier (BBB) endothelial cells form a barrier that is highly restrictive to passage of solutes between blood and brain. Many BBB transport mechanisms have been described that mediate transcellular movement of solutes across the barrier either into or out of the brain. One class of BBB transporters that is all too often overlooked is that of the ion transporters. The BBB has a rich array of ion transporters and channels that carry Na, K, Cl, HCO3, Ca, and other ions. Many of these are asymmetrically distributed between the luminal and abluminal membranes, giving BBB endothelial cells the ability to perform vectorial transport of ions across the barrier between blood and brain. In this manner, the BBB performs the important function of regulating the volume and composition of brain interstitial fluid. Through functional coupling of luminal and abluminal transporters and channels, the BBB carries Na, Cl, and other ions from blood into brain, producing up to 30% of brain interstitial fluid in healthy brain. During ischemic stroke cerebral edema forms by processes involving increased activity of BBB luminal Na transporters, resulting in "hypersecretion" of Na, Cl, and water into the brain interstitium. This review discusses the roles of luminal BBB Na transporters in edema formation in stroke, with an emphasis on Na-K-Cl cotransport and Na/H exchange. Evidence that these transporters provide effective therapeutic targets for reduction of edema in stroke is also discussed, as are recent findings regarding signaling pathways responsible for ischemia stimulation of the BBB Na transporters.
Collapse
|
44
|
Aceros H, Farah G, Noiseux N, Mukaddam-Daher S. Moxonidine modulates cytokine signalling and effects on cardiac cell viability. Eur J Pharmacol 2014; 740:168-82. [PMID: 25036265 DOI: 10.1016/j.ejphar.2014.06.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
Regression of left ventricular hypertrophy and improved cardiac function in SHR by the centrally acting imidazoline I1-receptor agonist, moxonidine, are associated with differential actions on circulating and cardiac cytokines. Herein, we investigated cell-type specific I1-receptor (also known as nischarin) signalling and the mechanisms through which moxonidine may interfere with cytokines to affect cardiac cell viability. Studies were performed on neonatal rat cardiomyocytes and fibroblasts incubated with interleukin (IL)-1β (5 ng/ml), tumor necrosis factor (TNF)-α (10 ng/ml), and moxonidine (10(-7) and 10(-5) M), separately and in combination, for 15 min, and 24 and 48 h for the measurement of MAPKs (ERK1/2, JNK, and p38) and Akt activation and inducible NOS (iNOS) expression, by Western blotting, and cardiac cell viability/proliferation and apoptosis by flow cytometry, MTT assay, and Live/Dead assay. Participation of imidazoline I1-receptors and the signalling proteins in the detected effects was identified using imidazoline I1-receptor antagonist and signalling protein inhibitors. The results show that IL-1β, and to a lower extent, TNF-α, causes cell death and that moxonidine protects against starvation- as well as IL-1β -induced mortality, mainly by maintaining membrane integrity, and in part, by improving mitochondrial activity. The protection involves activation of Akt, ERK1/2, p38, JNK, and iNOS. In contrast, moxonidine stimulates basal and IL-1β-induced fibroblast mortality by mechanisms that include inhibition of JNK and iNOS. Thus, apart from their actions on the central nervous system, imidazoline I1-receptors are directly involved in cardiac cell growth and death, and may play an important role in cardiovascular diseases associated with inflammation.
Collapse
Affiliation(s)
- Henry Aceros
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada
| | - Georges Farah
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada
| | - Nicolas Noiseux
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Suhayla Mukaddam-Daher
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada; Department of Medicine, Université de Montréal, Montreal, Québec, Canada.
| |
Collapse
|
45
|
The beneficial effect of melatonin in brain endothelial cells against oxygen-glucose deprivation followed by reperfusion-induced injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:639531. [PMID: 25126203 PMCID: PMC4122057 DOI: 10.1155/2014/639531] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/07/2014] [Accepted: 06/20/2014] [Indexed: 12/31/2022]
Abstract
Melatonin has a cellular protective effect in cerebrovascular and neurodegenerative diseases. Protection of brain endothelial cells against hypoxia and oxidative stress is important for treatment of central nervous system (CNS) diseases, since brain endothelial cells constitute the blood brain barrier (BBB). In the present study, we investigated the protective effect of melatonin against oxygen-glucose deprivation, followed by reperfusion- (OGD/R-) induced injury, in bEnd.3 cells. The effect of melatonin was examined by western blot analysis, cell viability assays, measurement of intracellular reactive oxygen species (ROS), and immunocytochemistry (ICC). Our results showed that treatment with melatonin prevents cell death and degradation of tight junction protein in the setting of OGD/R-induced injury. In response to OGD/R injury of bEnd.3 cells, melatonin activates Akt, which promotes cell survival, and attenuates phosphorylation of JNK, which triggers apoptosis. Thus, melatonin protects bEnd.3 cells against OGD/R-induced injury.
Collapse
|
46
|
Roles of MAPKAPK-2 and HSP27 in the reduction of renal ischemia-reperfusion injury by ischemic postconditioning in rats. Int Urol Nephrol 2014; 46:1455-64. [PMID: 24927932 DOI: 10.1007/s11255-014-0748-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Ischemic postconditioning is a procedure during which intermittent reperfusions are performed in the early phase of reperfusion to protect organs from ischemia/reperfusion injury. And in this study, we mainly investigated the injury-alleviative role of mitogen-activated protein kinase-activating protein kinase-2 (MAPKAPK-2) and heat shock protein 27 (HSP27) in renal ischemic reperfusion injury during the procedure of ischemic postconditioning. METHODS Sprague-Dawley rats were randomly divided into four groups. The injury models were prepared by clipping the left renal pedicle of rats after ligating the right renal pedicle for 60 min. In the ischemic postconditioning group, sequential reperfusions were done for 10 s and another ischemia for 10 s for six cycles after kidney ischemia for 60 min. In addition, the specific inhibitor SB203580 was injected through caudal vein before ischemia. Serum creatinine, blood urea nitrogen and the expression of HSP27 and MAPKAPK-2 were detected 1, 3, 6 and 24 h later after reperfusion. Furthermore, phosphorylation of HSP27 and MAPKAPK-2 protein contents, histological changes and apoptosis were compared 24 h later after reperfusion. RESULTS Our data showed that ischemic postconditioning attenuated the renal dysfunction and cell apoptosis induced by I/R and increased phosphorylation of MAPKAPK-2 and HSP27. The results indicated that ischemic postconditioning decreased apoptosis and improved renal function. CONCLUSIONS Taken together, it is suggested that the renal protective effect may be related to the levels of HSP27 and MAPKAPK-2 activation.
Collapse
|
47
|
Javadov S, Jang S, Agostini B. Crosstalk between mitogen-activated protein kinases and mitochondria in cardiac diseases: therapeutic perspectives. Pharmacol Ther 2014; 144:202-25. [PMID: 24924700 DOI: 10.1016/j.pharmthera.2014.05.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases cause more mortality and morbidity worldwide than any other diseases. Although many intracellular signaling pathways influence cardiac physiology and pathology, the mitogen-activated protein kinase (MAPK) family has garnered significant attention because of its vast implications in signaling and crosstalk with other signaling networks. The extensively studied MAPKs ERK1/2, p38, JNK, and ERK5, demonstrate unique intracellular signaling mechanisms, responding to a myriad of mitogens and stressors and influencing the signaling of cardiac development, metabolism, performance, and pathogenesis. Definitive relationships between MAPK signaling and cardiac dysfunction remain elusive, despite 30 years of extensive clinical studies and basic research of various animal/cell models, severities of stress, and types of stimuli. Still, several studies have proven the importance of MAPK crosstalk with mitochondria, powerhouses of the cell that provide over 80% of ATP for normal cardiomyocyte function and play a crucial role in cell death. Although many questions remain unanswered, there exists enough evidence to consider the possibility of targeting MAPK-mitochondria interactions in the prevention and treatment of heart disease. The goal of this review is to integrate previous studies into a discussion of MAPKs and MAPK-mitochondria signaling in cardiac diseases, such as myocardial infarction (ischemia), hypertrophy and heart failure. A comprehensive understanding of relevant molecular mechanisms, as well as challenges for studies in this area, will facilitate the development of new pharmacological agents and genetic manipulations for therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| | - Bryan Agostini
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| |
Collapse
|
48
|
Mitra A, Ray A, Datta R, Sengupta S, Sarkar S. Cardioprotective Role of P38 MAPK During Myocardial Infarction Via Parallel Activation of α-Crystallin B and Nrf2. J Cell Physiol 2014; 229:1272-82. [DOI: 10.1002/jcp.24565] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/22/2014] [Indexed: 01/25/2023]
Affiliation(s)
- Arkadeep Mitra
- Genetics and Molecular Cardiology Laboratory; Department of Zoology; University of Calcutta; Kolkata India
| | - Aramita Ray
- Genetics and Molecular Cardiology Laboratory; Department of Zoology; University of Calcutta; Kolkata India
| | - Ritwik Datta
- Genetics and Molecular Cardiology Laboratory; Department of Zoology; University of Calcutta; Kolkata India
| | - Shantanu Sengupta
- Genomics and Molecular Medicine Unit; CSIR-Institute of Genomics and Integrative Biology; New Delhi India
| | - Sagartirtha Sarkar
- Genetics and Molecular Cardiology Laboratory; Department of Zoology; University of Calcutta; Kolkata India
| |
Collapse
|
49
|
Akaboshi T, Yamanishi R. Certain carotenoids enhance the intracellular glutathione level in a murine cultured macrophage cell line by inducing glutamate-cysteine-ligase. Mol Nutr Food Res 2014; 58:1291-300. [PMID: 24668641 DOI: 10.1002/mnfr.201300753] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/12/2013] [Accepted: 12/20/2013] [Indexed: 02/05/2023]
Abstract
SCOPE Glutathione (GSH) increases in RAW264 murine macrophage cells exposed to β-carotene or β-cryptoxanthin, however, the underlying mechanism has not been clarified. In the present study, we investigated the expression of glutamate-cysteine-ligase (GCL), the rate-limiting enzyme in GSH synthesis, in these cells. METHODS AND RESULTS Both the protein and mRNA expression of GCL increased in a β-carotene concentration-dependent manner. Buthionine sulfoximine, a GCL inhibitor, abolished the β-carotene-induced GSH increase without affecting the β-carotene-induced GCL protein expression. Both cycloheximide, a translation inhibitor, and actinomycin D, a transcription inhibitor, completely suppressed the β-carotene-induced GCL protein expression and the concomitant GSH increase. Actinomycin D inhibited the β-carotene-induced Gcl mRNA expression as well. Similarly to β-carotene, β-cryptoxanthin upregulated the GCL protein expression, but lutein did not. The c-Jun N-terminal kinase (JNK) inhibitor, SP600125, suppressed the β-carotene-induced GSH increase, whereas a p38 mitogen-activated protein kinase inhibitor or an extracellular signal-regulated kinase 1/2 inhibitor did not. The JNK inhibitor also suppressed the β-carotene-induced GCL protein expression, and consistently β-carotene induced JNK phosphorylation. CONCLUSION These findings revealed that certain carotenoids induce the Gcl mRNA expression in RAW264 cells and subsequently the GCL protein expression, which concomitantly enhances the intracellular GSH level, in a JNK pathway-related manner.
Collapse
Affiliation(s)
- Teppei Akaboshi
- Department of Food Science, Graduate School of Nutrition and Biosciences, The University of Tokushima, Tokushima, Japan
| | | |
Collapse
|
50
|
Saleem MTS, Chetty MC, Kavimani S. Antioxidants and tumor necrosis factor alpha-inhibiting activity of sesame oil against doxorubicin-induced cardiotoxicity. Ther Adv Cardiovasc Dis 2014; 8:4-11. [PMID: 24441175 DOI: 10.1177/1753944713516532] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Oxidative stress is currently considered to be the key factor in doxorubicin-induced cardiotoxicity. Comparatively small quantity of the endogenous antioxidant content of the heart is assumed to be the predisposing factor for doxorubicin-induced cardiotoxicity. The present research was designed to evaluate the antioxidant potential and tumor necrosis factor alpha-(TNF-α) inhibiting activity of sesame oil against acute doxorubicin-induced cardiotoxicity. METHODS Male Wistar albino rats (180-200 g) were administered sesame oil in two dissimilar doses (5 and 10 ml/kg body weight, orally) for 30 days, followed by a single dose of doxorubicin (30 mg/kg s.c.). RESULTS In the doxorubicin-treated group, increased oxidative stress was proven by a significant rise of thiobarbituric acid reactive substances level and a decrease of myocardial superoxide dismutase, catalase and reduced glutathione content. Histopathological studies showed myocardial necrosis with accumulation of inflammatory cells, vacuolization and overall enlargement of the myocardium. Western blot analysis showed marked expression of TNF-α in the myocardium. Alteration in biochemical parameters by doxorubicin administration was prevented significantly (p < 0.0001) in the 5 and 10 ml/kg sesame oil treated rat hearts. Treatment with 5 and 10 ml/kg of sesame oil reduced the doxorubicin-induced TNF-α expression in the myocardium, which was associated with reduced myocyte injury. The overall effect of sesame oil was comparable with probucol, which shows similar protection. CONCLUSION The chronic oral administration of sesame oil prevents acute doxorubicin-induced cardiotoxicity by enhancing cardiac endogenous antioxidants and decreasing myocardial TNF-α expression.
Collapse
Affiliation(s)
- Mohamed T S Saleem
- Research Scholar, Jawaharlal Nehru Technological University, Kakinada & Department of Pharmacology, Annamacharya College of Pharmacy, Rajampet 516126, Andhrapradesh, India
| | | | | |
Collapse
|