1
|
Xuan Y, Yu C, Ni K, Congcong L, Lixin Q, Qingxian L. Protective effects of tanshinone IIA on Porphyromonas gingivalis-induced atherosclerosis via the downregulation of the NOX2/NOX4-ROS mediation of NF-κB signaling pathway. Microbes Infect 2023; 25:105177. [PMID: 37392987 DOI: 10.1016/j.micinf.2023.105177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/07/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Tanshinone IIA (TSA), an active component isolated from Danshen, possess high medicinal values against atherosclerosis by reducing vascular oxidative stress, inhibiting platelet aggregation, and protecting the endothelium from damage. The periodontal pathogen Porphyromonas gingivalis (P. gingivalis) has been proven to accelerate the development of atherosclerosis. We aim to determine the effects of TSA on P. gingivalis-induced atherosclerosis in ApoE-knockout (ApoE-/-) mice. After feeding with a high-lipid diet and infected with P. gingivalis three times per week for four weeks, TSA-treated (60 mg/kg/d) mice greatly inhibited atherosclerotic lesions both morphologically and biochemically and exhibited significantly reduction ROS, 8-OHdG, and ox-LDL levels in serum compared with P. gingivalis-infected mice. Additionally, TSA-treated mice were observed a marked reduction of ROS, 8-OHdG and ox-LDL in the serum, mRNA levels of COX-2, LOX-1, NOX2 and NOX4 in the aorta, as well as the levels of NOX2, NOX4, and NF-κB. These results suggest that TSA attenuates oxidative stress by decreasing NOX2 and NOX4 and downregulating NF-κB signaling pathway, which might be contributed to the amelioration of atherosclerosis.
Collapse
Affiliation(s)
- Yan Xuan
- Department of the Fourth Division, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Cai Yu
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Kang Ni
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Lou Congcong
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Qiu Lixin
- Department of the Fourth Division, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| | - Luan Qingxian
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| |
Collapse
|
2
|
Wang Z, Choi K. Pharmacological modulation of chloride channels as a therapeutic strategy for neurological disorders. Front Physiol 2023; 14:1122444. [PMID: 36935741 PMCID: PMC10017882 DOI: 10.3389/fphys.2023.1122444] [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: 12/12/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Chloride homeostasis is critical in the physiological functions of the central nervous system (CNS). Its concentration is precisely regulated by multiple ion-transporting proteins such as chloride channels and transporters that are widely distributed in the brain cells, including neurons and glia. Unlike ion transporters, chloride channels provide rapid responses to efficiently regulate ion flux. Some of chloride channels are also permeable to selected organic anions such as glutamate and γ-aminobutyric acid, suggesting neuroexcitatory and neuroinhibitory functions while gating. Dysregulated chloride channels are implicated in neurological disorders, e.g., ischemia and neuroinflammation. Modulation of chloride homeostasis through chloride channels has been suggested as a potential therapeutic approach for neurological disorders. The drug design for CNS diseases is challenging because it requires the therapeutics to traverse the blood-brain-barrier. Small molecules are a well-established modality with better cell permeability due to their lower molecular weight and flexibility for structure optimization compared to biologics. In this article, we describe the important roles of chloride homeostasis in each type of brain cells and introduce selected chloride channels identified in the CNS. We then discuss the contribution of their dysregulations towards the pathogenesis of neurological disorders, emphasizing the potential of targeting chloride channels as a therapeutic strategy for CNS disease treatment. Along with this literature survey, we summarize the small molecules that modulate chloride channels and propose the potential strategy of optimizing existing drugs to brain-penetrants to support future CNS drug discovery.
Collapse
|
3
|
Discovery of novel dihydropyrazole-stilbene derivatives for ameliorating heart failure through modulation of p38/NF-κB signaling pathway. Bioorg Chem 2022; 129:106206. [DOI: 10.1016/j.bioorg.2022.106206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/28/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
4
|
Shishikura D, Octavia Y, Hayat U, Thondapu V, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
5
|
Lycium barbarum polysaccharide antagonizes cardiomyocyte apoptosis by inhibiting the upregulation of GRK2 induced by I/R injury, and salvage mitochondrial fission/fusion imbalance and AKT/eNOS signaling. Cell Signal 2022; 92:110252. [DOI: 10.1016/j.cellsig.2022.110252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/20/2021] [Accepted: 01/14/2022] [Indexed: 12/24/2022]
|
6
|
D’Errico S, Russa RL, Maiese A, Santurro A, Scopetti M, Romano S, Zanon M, Frati P, Fineschi V. Atypical antipsychotics and oxidative cardiotoxicity: review of literature and future perspectives to prevent sudden cardiac death. J Geriatr Cardiol 2021; 18:663-685. [PMID: 34527032 PMCID: PMC8390928 DOI: 10.11909/j.issn.1671-5411.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress is considered the principal mediator of myocardial injury under pathological conditions. It is well known that reactive oxygen (ROS) or nitrogen species (RNS) are involved in myocardial injury and repair at the same time and that cellular damage is generally due to an unbalance between generation and elimination of the free radicals due to an inadequate mechanism of antioxidant defense or to an increase in ROS and RNS. Major adverse cardiovascular events are often associated with drugs with associated findings such as fibrosis or inflammation of the myocardium. Despite efforts in the preclinical phase of the development of drugs, cardiotoxicity still remains a great concern. Cardiac toxicity due to second-generation antipsychotics (clozapine, olanzapine, quetiapine) has been observed in preclinical studies and described in patients affected with mental disorders. A role of oxidative stress has been hypothesized but more evidence is needed to confirm a causal relationship. A better knowledge of cardiotoxicity mechanisms should address in the future to establish the right dose and length of treatment without impacting the physical health of the patients.
Collapse
Affiliation(s)
- Stefano D’Errico
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Raffaele La Russa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
| | - Aniello Maiese
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Surgical Pathology, Medical, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Santurro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Scopetti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Romano
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Martina Zanon
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Paola Frati
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Vittorio Fineschi
- IRCSS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
7
|
Souza LM, Okoshi MP, Gomes MJ, Gatto M, Rodrigues EA, Pontes THD, Damatto FC, Oliveira LRS, Borim PA, Lima ARR, Zornoff LAM, Okoshi K, Pagan LU. Effects of Late Aerobic Exercise on Cardiac Remodeling of Rats with Small-Sized Myocardial Infarction. Arq Bras Cardiol 2021; 116:784-792. [PMID: 33886729 PMCID: PMC8121407 DOI: 10.36660/abc.20190813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/16/2020] [Indexed: 12/30/2022] Open
Abstract
Fundamento: O exercício físico tem sido considerado uma importante terapia não farmacológica para a prevenção e tratamento das doenças cardiovasculares. No entanto, seus efeitos na remodelação cardíaca leve não são claros. Objetivo: Avaliar a influência do exercício aeróbico sobre a capacidade funcional, estrutura cardíaca, função ventricular esquerda (VE) e expressão gênica das subunidades da NADPH oxidase em ratos com infarto do miocárdio pequeno (IM). Métodos: Três meses após a indução do IM, ratos Wistar foram divididos em três grupos: Sham; IM sedentário (IM-SED); e IM exercício aeróbico (IM-EA). Os ratos se exercitaram em uma esteira três vezes por semana durante 12 semanas. Um ecocardiograma foi realizado antes e após o treinamento. O tamanho do infarto foi avaliado por histologia e a expressão gênica por RT-PCR. O nível de significância para análise estatística foi estabelecido em 5%. Resultados: Ratos com IM menor que 30% da área total do VE foram incluídos no estudo. A capacidade funcional foi maior no IM-EA do que nos ratos Sham e IM-SED. O tamanho do infarto não diferiu entre os grupos. Ratos infartados apresentaram aumento do diâmetro diastólico e sistólico do VE, diâmetro do átrio esquerdo e massa do VE, com disfunção sistólica. A espessura relativa da parede foi menor no grupo IM-SED do que nos grupos IM-EA e Sham. A expressão gênica das subunidades NADPH oxidase NOX2, NOX4, p22phox e p47phox não diferiu entre os grupos. Conclusão: Infarto do miocárdio pequeno altera a estrutura cardíaca e a função sistólica do VE. O exercício aeróbico tardio pode melhorar a capacidade funcional e a remodelação cardíaca por meio da preservação da geometria ventricular esquerda. A expressão gênica das subunidades da NADPH oxidase não está envolvida na remodelação cardíaca, nem é modulada pelo exercício aeróbico em ratos com infarto do miocárdio pequeno.
Collapse
Affiliation(s)
- Lidiane M Souza
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Marina P Okoshi
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Mariana J Gomes
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Mariana Gatto
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Eder A Rodrigues
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Thierres H D Pontes
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Felipe C Damatto
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Leiliane R S Oliveira
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Patrícia Aparecida Borim
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Aline R R Lima
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Leonardo A M Zornoff
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Katashi Okoshi
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| | - Luana U Pagan
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Botucatu, SP - Brasil
| |
Collapse
|
8
|
Chazelas P, Steichen C, Favreau F, Trouillas P, Hannaert P, Thuillier R, Giraud S, Hauet T, Guillard J. Oxidative Stress Evaluation in Ischemia Reperfusion Models: Characteristics, Limits and Perspectives. Int J Mol Sci 2021; 22:ijms22052366. [PMID: 33673423 PMCID: PMC7956779 DOI: 10.3390/ijms22052366] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemia reperfusion injury is a complex process consisting of a seemingly chaotic but actually organized and compartmentalized shutdown of cell function, of which oxidative stress is a key component. Studying oxidative stress, which results in an imbalance between reactive oxygen species (ROS) production and antioxidant defense activity, is a multi-faceted issue, particularly considering the double function of ROS, assuming roles as physiological intracellular signals and as mediators of cellular component damage. Herein, we propose a comprehensive overview of the tools available to explore oxidative stress, particularly in the study of ischemia reperfusion. Applying chemistry as well as biology, we present the different models currently developed to study oxidative stress, spanning the vitro and the silico, discussing the advantages and the drawbacks of each set-up, including the issues relating to the use of in vitro hypoxia as a surrogate for ischemia. Having identified the limitations of historical models, we shall study new paradigms, including the use of stem cell-derived organoids, as a bridge between the in vitro and the in vivo comprising 3D intercellular interactions in vivo and versatile pathway investigations in vitro. We shall conclude this review by distancing ourselves from "wet" biology and reviewing the in silico, computer-based, mathematical modeling, and numerical simulation options: (a) molecular modeling with quantum chemistry and molecular dynamic algorithms, which facilitates the study of molecule-to-molecule interactions, and the integration of a compound in a dynamic environment (the plasma membrane...); (b) integrative systemic models, which can include many facets of complex mechanisms such as oxidative stress or ischemia reperfusion and help to formulate integrated predictions and to enhance understanding of dynamic interaction between pathways.
Collapse
Affiliation(s)
- Pauline Chazelas
- Maintenance Myélinique et Neuropathies Périphériques, Université de Limoges, EA 6309, 87032 Limoges, France; (P.C.); (F.F.)
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Limoges, 87042 Limoges, France
| | - Clara Steichen
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
| | - Frédéric Favreau
- Maintenance Myélinique et Neuropathies Périphériques, Université de Limoges, EA 6309, 87032 Limoges, France; (P.C.); (F.F.)
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Limoges, 87042 Limoges, France
| | - Patrick Trouillas
- INSERM U1248, IPPRITT, Université de Limoges, 87032 Limoges, France;
- RCPTM, University Palacký of Olomouc, 771 47 Olomouc, Czech Republic
| | - Patrick Hannaert
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
| | - Raphaël Thuillier
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
| | - Sébastien Giraud
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
| | - Thierry Hauet
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
- FHU SUPORT Survival Optimization in Organ Transplantation, 86021 Poitiers, France
- IBiSA Plateforme Modélisation Préclinique-Innovations Chirurgicale et Technologique (MOPICT), Do-maine Expérimental du Magneraud, 17700 Surgères, France
| | - Jérôme Guillard
- UMR CNRS 7285 IC2MP, Team 5 Chemistry, Université de Poitiers, 86073 Poitiers, France
- Correspondence: ; Tel.: +33-5-49-44-38-59
| |
Collapse
|
9
|
Daiber A, Andreadou I, Oelze M, Davidson SM, Hausenloy DJ. Discovery of new therapeutic redox targets for cardioprotection against ischemia/reperfusion injury and heart failure. Free Radic Biol Med 2021; 163:325-343. [PMID: 33359685 DOI: 10.1016/j.freeradbiomed.2020.12.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
Global epidemiological studies reported a shift from maternal/infectious communicable diseases to chronic non-communicable diseases and a major part is attributable to atherosclerosis and metabolic disorders. Accordingly, ischemic heart disease was identified as a leading risk factor for global mortality and morbidity with a prevalence of 128 million people. Almost 9 million premature deaths can be attributed to ischemic heart disease and subsequent acute myocardial infarction and heart failure, also representing a substantial socioeconomic burden. As evidenced by typical oxidative stress markers such as lipid peroxidation products or oxidized DNA/RNA bases, the formation of reactive oxygen species by various sources (NADPH oxidases, xanthine oxidase and mitochondrial resperatory chain) plays a central role for the severity of ischemia/reperfusion damage. The underlying mechanisms comprise direct oxidative damage but also adverse redox-regulation of kinase and calcium signaling, inflammation and cardiac remodeling among others. These processes and the role of reactive oxygen species are discussed in the present review. We also present and discuss potential targets for redox-based therapies that are either already established in the clinics (e.g. guanylyl cyclase activators and stimulators) or at least successfully tested in preclinical models of myocardial infarction and heart failure (mitochondria-targeted antioxidants). However, reactive oxygen species have not only detrimental effects but are also involved in essential cellular signaling and may even act protective as seen by ischemic pre- and post-conditioning or eustress - which makes redox therapy quite challenging.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Matthias Oelze
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan.
| |
Collapse
|
10
|
Mollace V, Rosano GMC, Anker SD, Coats AJS, Seferovic P, Mollace R, Tavernese A, Gliozzi M, Musolino V, Carresi C, Maiuolo J, Macrì R, Bosco F, Chiocchi M, Romeo F, Metra M, Volterrani M. Pathophysiological Basis for Nutraceutical Supplementation in Heart Failure: A Comprehensive Review. Nutrients 2021; 13:257. [PMID: 33477388 PMCID: PMC7829856 DOI: 10.3390/nu13010257] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/25/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
There is evidence demonstrating that heart failure (HF) occurs in 1-2% of the global population and is often accompanied by comorbidities which contribute to increasing the prevalence of the disease, the rate of hospitalization and the mortality. Although recent advances in both pharmacological and non-pharmacological approaches have led to a significant improvement in clinical outcomes in patients affected by HF, residual unmet needs remain, mostly related to the occurrence of poorly defined strategies in the early stages of myocardial dysfunction. Nutritional support in patients developing HF and nutraceutical supplementation have recently been shown to possibly contribute to protection of the failing myocardium, although their place in the treatment of HF requires further assessment, in order to find better therapeutic solutions. In this context, the Optimal Nutraceutical Supplementation in Heart Failure (ONUS-HF) working group aimed to assess the optimal nutraceutical approach to HF in the early phases of the disease, in order to counteract selected pathways that are imbalanced in the failing myocardium. In particular, we reviewed several of the most relevant pathophysiological and molecular changes occurring during the early stages of myocardial dysfunction. These include mitochondrial and sarcoplasmic reticulum stress, insufficient nitric oxide (NO) release, impaired cardiac stem cell mobilization and an imbalanced regulation of metalloproteinases. Moreover, we reviewed the potential of the nutraceutical supplementation of several natural products, such as coenzyme Q10 (CoQ10), a grape seed extract, Olea Europea L.-related antioxidants, a sodium-glucose cotransporter (SGLT2) inhibitor-rich apple extract and a bergamot polyphenolic fraction, in addition to their support in cardiomyocyte protection, in HF. Such an approach should contribute to optimising the use of nutraceuticals in HF, and the effect needs to be confirmed by means of more targeted clinical trials exploring the efficacy and safety of these compounds.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Giuseppe M. C. Rosano
- Cardiology Clinical Academic Group, St George’s Hospitals NHS Trust University of London, London SW17 0QT, UK;
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Stefan D. Anker
- Department of Cardiology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Andrew J. S. Coats
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Petar Seferovic
- Faculty of Medicine, Belgrade University, 11000 Belgrade, Serbia;
| | - Rocco Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Annamaria Tavernese
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Micaela Gliozzi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Vincenzo Musolino
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Cristina Carresi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Jessica Maiuolo
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Roberta Macrì
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Francesca Bosco
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Marcello Chiocchi
- Department of Diagnostic Imaging and Interventional Radiology, Policlinico Tor Vergata, 00199 Rome, Italy;
| | - Francesco Romeo
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00199 Rome, Italy;
| | - Marco Metra
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Maurizio Volterrani
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| |
Collapse
|
11
|
Nox2 Upregulation and p38α MAPK Activation in Right Ventricular Hypertrophy of Rats Exposed to Long-Term Chronic Intermittent Hypobaric Hypoxia. Int J Mol Sci 2020; 21:ijms21228576. [PMID: 33202984 PMCID: PMC7698046 DOI: 10.3390/ijms21228576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
One of the consequences of high altitude (hypobaric hypoxia) exposure is the development of right ventricular hypertrophy (RVH). One particular type of exposure is long-term chronic intermittent hypobaric hypoxia (CIH); the molecular alterations in RVH in this particular condition are less known. Studies show an important role of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex-induced oxidative stress and protein kinase activation in different models of cardiac hypertrophy. The aim was to determine the oxidative level, NADPH oxidase expression and MAPK activation in rats with RVH induced by CIH. Male Wistar rats were randomly subjected to CIH (2 days hypoxia/2 days normoxia; n = 10) and normoxia (NX; n = 10) for 30 days. Hypoxia was simulated with a hypobaric chamber. Measurements in the RV included the following: hypertrophy, Nox2, Nox4, p22phox, LOX-1 and HIF-1α expression, lipid peroxidation and H2O2 concentration, and p38α and Akt activation. All CIH rats developed RVH and showed an upregulation of LOX-1, Nox2 and p22phox and an increase in lipid peroxidation, HIF-1α stabilization and p38α activation. Rats with long-term CIH-induced RVH clearly showed Nox2, p22phox and LOX-1 upregulation and increased lipid peroxidation, HIF-1α stabilization and p38α activation. Therefore, these molecules may be considered new targets in CIH-induced RVH.
Collapse
|
12
|
Targeting the Nrf2/ARE Signalling Pathway to Mitigate Isoproterenol-Induced Cardiac Hypertrophy: Plausible Role of Hesperetin in Redox Homeostasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9568278. [PMID: 32952852 PMCID: PMC7482027 DOI: 10.1155/2020/9568278] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/27/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
Cardiac hypertrophy is the underlying cause of heart failure and is characterized by excessive oxidative stress leading to collagen deposition. Therefore, understanding the signalling mechanisms involved in excessive extracellular matrix deposition is necessary to prevent cardiac remodelling and heart failure. In this study, we hypothesized that hesperetin, a flavanone that elicits the activation of Nrf2 signalling and thereby suppresses oxidative stress, mediated pathological cardiac hypertrophy progression. A cardiac hypertrophy model was established with subcutaneous injection of isoproterenol in male Wistar rats. Oxidative stress markers, antioxidant defense status, and its upstream signalling molecules were evaluated to discover the impacts of hesperetin in ameliorating cardiac hypertrophy. Our results implicate that hesperetin pretreatment resulted in the mitigation of oxidative stress by upregulating antioxidant capacity of the heart. This curative effect might be owing to the activation of the master regulator of antioxidant defense system, known as Nrf2. Further, analysis of Nrf2 revealed that hesperetin enhances its nuclear translocation as well as the expression of its downstream targets (GCLC, NQO1, and HO-1) to boost the antioxidative status of the cells. To support this notion, in vitro studies were carried out in isoproterenol-treated H9c2 cells. Immunocytochemical analysis showed augmented nuclear localization of Nrf2 implicating the action of hesperetin at the molecular level to maintain the cellular redox homeostasis. Thus, it is conceivable that hesperetin could be a potential therapeutic candidate that enhances Nrf2 signalling and thereby ameliorates pathological cardiac remodelling.
Collapse
|
13
|
Bilgic S, Ozgocmen M, Ozer MK, Asci H. Misoprostol ameliorates doxorubicin induced cardiac damage by decreasing oxidative stress and apoptosis in rats. Biotech Histochem 2020; 95:514-521. [PMID: 32180467 DOI: 10.1080/10520295.2020.1727013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We investigated the potential cardioprotective effects of misoprostol (MP) on doxorubicin (DOX) induced cardiac damage using histologic and biochemical assessment of rat heart. We used 21 male rats divided randomly into three groups: group 1, control; group 2, DOX; group 3, DOX + MP. The control group was given 0.5 ml 0.9% NaCl intraperitoneally (i.p.) and 1 ml 0.9% NaCl orally for 6 days. DOX was administered as a single dose of 20 mg/kg i.p. on day 3. MP was administered orally for 6 days. We found that treatment with MP decreased significantly serum cardiac troponin-I, brain natriuretic peptide levels, and lactate dehydrogenase, aspartate aminotransferase, alanine transaminase and creatine kinase isoenzyme-MB activities. DOX increased the malondialdehyde level and decreased the catalase, superoxide dismutase activities and glutathione levels; MP prevented these alterations. MP also decreased NADPH oxidase-4 and caspase-3 levels. In the DOX + MP group, oxidative stress was decreased, antioxidant activity was increased and histopathological changes were decreased compared to the DOX group. Cardiac damage caused by DOX was attenuated by MP treatment owing to the antioxidative and anti-apoptotic effects of MP. MP may be useful for reducing the severity of DOX induced damage.
Collapse
Affiliation(s)
- S Bilgic
- Department of Medical Biochemistry, Vocational School of Health Services, University of Adıyaman , Adıyaman, Turkey
| | - M Ozgocmen
- Department of Histology, and Embryology, Faculty of Medicine, Suleyman Demirel University , Isparta, Turkey
| | - M K Ozer
- Department of Pharmacology, Faculty of Medicine, Adıyaman University , Adıyaman, Turkey
| | - H Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University , Isparta, Turkey
| |
Collapse
|
14
|
Wu Z, Zhao X, Miyamoto A, Zhao S, Liu C, Zheng W, Wang H. Effects of steroidal saponins extract from Ophiopogon japonicus root ameliorates doxorubicin-induced chronic heart failure by inhibiting oxidative stress and inflammatory response. PHARMACEUTICAL BIOLOGY 2019; 57:176-183. [PMID: 30860934 PMCID: PMC6419681 DOI: 10.1080/13880209.2019.1577467] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/08/2018] [Accepted: 01/28/2019] [Indexed: 05/25/2023]
Abstract
CONTEXT Ophiopogonis Radix, the root of Ophiopogon japonicus (Thunb.) Ker-Gawl (Liliaceae), is a Traditional Chinese Medicine, which has been investigated to possess effective treatment of cardiovascular diseases. OBJECTIVE This study evaluates the cardioprotective effects of steroidal saponins extract from Ophiopogon japonicus (SOJ) root against doxorubicin-induced chronic heart failure (CHF) through the amelioration of oxidative stress and inflammation. MATERIALS AND METHODS A Sprague-Dawley rat model of CHF was established by intraperitoneally injected with DOX. All rats were randomly divided into four groups: Control group, CHF group, CHF + SOJ (100 mg/kg) treatment group, SOJ (100 mg/kg) treatment group (n = 8/group). After six weeks administration, biometric and echocardiography were measured. The levels of biochemical parameters were measured using commercial kits. RESULTS The values of LVESP, +dP/dtmax, -dP/dtmax, EF and FS increased to 116.20 ± 1.68 mmHg, 2978.71 ± 168.26 mmHg/s, 3452.61 ± 286.09 mmHg/s, 68.26 ± 5.28% and 31.97 ± 3.79%, respectively; the values of LVEDP, LVESD and LVEDD decreased to 8.85 ± 0.84 mmHg, 8.39 ± 0.45 mm and 12.36 ± 0.87 mm in CHF + SOJ group. In addition, the levels of IL-6, TNF-α and IL-1β decreased to 154.41 ± 7.72 pg/mg protein, 110.02 ± 6.96 pg/mg protein and 39.39 ± 5.27 pg/mg protein, respectively; the relative activity of p38 MAPK decreased to 2.60 ± 0.40 in CHF + SOJ group. Furthermore, the activities of SOD, CAT and GSH-Px increased to 268.77 ± 6.20 U/mg protein, 13.68 ± 0.68 U/mg protein and 316.90 ± 8.08 µmol/mg protein, and the content of MDA decreased to 4.03 ± 0.43 nmol/mg protein in CHF + SOJ group. CONCLUSIONS SOJ exerts the cardioprotective effect against DOX-induced CHF through suppressing inflammatory and oxidative stress. These results provide evidence that SOJ might be an effective treatment for CHF.
Collapse
Affiliation(s)
- Zhongwei Wu
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Xuekai Zhao
- Department of Cardiac Surgery, Zibo Central Hospital, Zibo, China
| | - Akira Miyamoto
- Department of Rehabilitation, Kobe International University, Kobe, Japan
| | - Shengji Zhao
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Chaoquan Liu
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Weimin Zheng
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - HongTao Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an JiaoTong University, Xian, China
| |
Collapse
|
15
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
16
|
Ortiz VD, Türck P, Teixeira R, Lima-Seolin BG, Lacerda D, Fraga SF, Hickmann A, Gatelli Fernandes TR, Belló-Klein A, Luz de Castro A, da Rosa Araujo AS. Carvedilol and thyroid hormones co-administration mitigates oxidative stress and improves cardiac function after acute myocardial infarction. Eur J Pharmacol 2019; 854:159-166. [DOI: 10.1016/j.ejphar.2019.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 10/27/2022]
|
17
|
Sakai T, Imai J, Takagaki H, Ui M, Hatta S. Cytoplasmic OH scavenger TA293 attenuates cellular senescence and fibrosis by activating macrophages through oxidized phospholipids/TLR4. Life Sci 2019; 221:284-292. [DOI: 10.1016/j.lfs.2019.02.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/05/2019] [Accepted: 02/17/2019] [Indexed: 01/19/2023]
|
18
|
Activated glycine receptors may decrease endosomal NADPH oxidase activity by opposing ClC-3-mediated efflux of chloride from endosomes. Med Hypotheses 2019; 123:125-129. [PMID: 30696582 DOI: 10.1016/j.mehy.2019.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/15/2019] [Indexed: 12/25/2022]
|
19
|
Fibroblast Growth Factor Receptor 4 Targeting in Cancer: New Insights into Mechanisms and Therapeutic Strategies. Cells 2019; 8:cells8010031. [PMID: 30634399 PMCID: PMC6356571 DOI: 10.3390/cells8010031] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/21/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4), a tyrosine kinase receptor for FGFs, is involved in diverse cellular processes, including the regulation of cell proliferation, differentiation, migration, metabolism, and bile acid biosynthesis. High activation of FGFR4 is strongly associated with the amplification of its specific ligand FGF19 in many types of solid tumors and hematologic malignancies, where it acts as an oncogene driving the cancer development and progression. Currently, the development and therapeutic evaluation of FGFR4-specific inhibitors, such as BLU9931 and H3B-6527, in animal models and cancer patients, are paving the way to suppress hyperactive FGFR4 signaling in cancer. This comprehensive review not only covers the recent discoveries in understanding FGFR4 regulation and function in cancer, but also reveals the therapeutic implications and applications regarding emerging anti-FGFR4 agents. Our aim is to pinpoint the potential of FGFR4 as a therapeutic target and identify new avenues for advancing future research in the field.
Collapse
|
20
|
Reyes ME, Ma J, Grove ML, Ater JL, Morrison AC, Hildebrandt MAT. RNA sequence analysis of inducible pluripotent stem cell-derived cardiomyocytes reveals altered expression of DNA damage and cell cycle genes in response to doxorubicin. Toxicol Appl Pharmacol 2018; 356:44-53. [PMID: 30031762 DOI: 10.1016/j.taap.2018.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/29/2018] [Accepted: 07/18/2018] [Indexed: 02/03/2023]
Abstract
Anthracyclines, such as doxorubicin, are highly effective chemotherapeutic agents, yet are associated with increased risk of cardiotoxicity. The genes and pathways involved in the development of heart damage following doxorubicin exposure in humans remain elusive. Our objective was to explore time- and dose-dependent changes in gene expression via RNA sequence (RNAseq) that mediate doxorubicin response in human iPSC-cardiomyocytes following 50, 150, or 450 nM exposure for 2, 7, or 12 days. Clustering and differential expression analyses were conducted to identify genes with altered expression. Samples clustered in dose and time-dependent manners, and MCM5, PRC1, NUSAP1, CENPF, CCNB1, MELK, AURKB, and RACGAP1 were consistently significantly differentially expressed between untreated and treated conditions. These genes were also significantly downregulated in pairwise analyses, which was validated by reverse transcription polymerase chain reaction (RT-PCR). Pathway analysis identified the top canonical pathways involved in response, implicating DNA damage repair response and the cell cycle as having roles in the development of doxorubicin-induced cardiotoxicity in the human cardiomyocyte.
Collapse
Affiliation(s)
- Monica E Reyes
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianzhong Ma
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joann L Ater
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michelle A T Hildebrandt
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
21
|
Bai Y, Chen Q, Sun YP, Wang X, Lv L, Zhang LP, Liu JS, Zhao S, Wang XL. Sulforaphane protection against the development of doxorubicin-induced chronic heart failure is associated with Nrf2 Upregulation. Cardiovasc Ther 2018. [PMID: 28636290 DOI: 10.1111/1755-5922.12277] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an anthracycline antitumor drug. However, its clinical use is limited by dose-dependent cardiotoxicity and even progresses to chronic heart failure (CHF). OBJECTIVE This study aims to investigate whether the Nrf2 activator, sulforaphane (SFN), can prevent DOX-induced CHF. METHODS Male Sprague-Dawley rats which received treatment for 6 weeks were divided into four groups (n=30 per group): control, SFN, DOX and DOX plus SFN group. RESULTS Results revealed that DOX induced progressive cardiac damage as indicated by increased cardiac injury markers, cardiac inflammation, fibrosis and oxidative stress. SFN significantly prevented DOX-induced progressive cardiac dysfunction between 2-6 weeks and prevented DOX-induced cardiac function deterioration. Furthermore, it significantly decreased ejection fraction and increased the expression of brain natriuretic peptide. SFN also almost completely prevented DOX-induced cardiac oxidative stress, inflammation and fibrosis. SFN upregulated NF-E2-related factor 2 (Nrf2) expression and transcription activity, which was reflected by the increased mRNA expression of Nrf2 and its downstream genes. Furthermore, in cultured H9c2 cardiomyocytes, the protective effect of SFN against DOX-induced fibrotic and inflammatory responses was abolished by Nrf2 silencing. CONCLUSION We arrived at the conclusion that DOX-induced CHF can be prevented by SFN through the upregulation of Nrf2 expression and transcriptional function.
Collapse
Affiliation(s)
- Yang Bai
- The Cardiac Surgery Department, The First Hospital of Jilin University, Changchun, China
| | - Qiang Chen
- School of Public Health, Jilin University, Changchun, China
| | - Yun-Peng Sun
- The Cardiac Surgery Department, The First Hospital of Jilin University, Changchun, China
| | - Xuan Wang
- Department of Pharmacology, The College of Basic Medicine of Jilin University, Changchun, China
| | - Li Lv
- The Jilin Province People's Hospital, Changchun, China
| | - Li-Ping Zhang
- The Cardiovascular Department, The First Hospital of Jilin University, Changchun, China
| | - Jin-Sha Liu
- The Cardiovascular Department, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Song Zhao
- The Spine Surgery Department, The First Hospital of Jilin University, Changchun, China
| | - Xiao-Lu Wang
- The Jilin Province People's Hospital, Changchun, China
| |
Collapse
|
22
|
Essential role for smooth muscle cell stromal interaction molecule-1 in myocardial infarction. J Hypertens 2018; 36:377-386. [PMID: 29611835 DOI: 10.1097/hjh.0000000000001518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Stromal interacting molecule-1 (STIM1) plays a role in coordinating calcium signaling in different cell types. The increase or deletion of STIM1 expression in cardiomyocyte causes cardiac complication. Moreover, the deletion of STIM1 in endothelial cell causes vascular endothelial dysfunction. However, the disruption of STIM1 in smooth muscle cells (SMC) has no effect on endothelial function but protects vascular function when mice are infused with angiotensin-II. Nevertheless, the role of SMC-STIM1 in acute and chronic myocardial infarction (MI) induced by acute ischemia-reperfusion injury and permanent coronary artery occlusion is unknown. METHODS AND RESULTS Stim1 were generated and crossed into the SM22α-Cre backgrounds. SM22α-Cre causes deletion of STIM1 floxed genes in adult SMC (Stim1). Control and Stim1 mice were subjected to acute ischemia-reperfusion injury. Hearts were then harvested and incubated with triphenyltetrazolium chloride to determine the infarct size. In control mice which are subjected to ischemia-reperfusion, the heart developed a significant infarct associated with an increase in STIM1 expression. Interestingly, the infarct size was substantially reduced in Stim1 mice. The protection in Stim1 mice against ischemia-reperfusion injury involves the modulation of endoplasmic reticulum stress, apoptosis, oxidative stress, protein kinase B, and mitogen-activated protein (MAP) kinase (ERK1/2 and p38) signaling, and inflammation. Furthermore, in another model of chronic MI induced by permanent coronary artery occlusion, SMC-STIM1 disruption significantly reduced myocardial infarct size and improved cardiac function. CONCLUSION Our results provide new evidence that SMC-STIM1 disruption is a novel mechanism that protects the heart from MI through reduction of endoplasmic reticulum stress, oxidative stress, MAP-Kinase, apoptosis, and inflammation.
Collapse
|
23
|
Hu W, Wu J, Jiang W, Tang J. MicroRNAs and Presbycusis. Aging Dis 2018; 9:133-142. [PMID: 29392088 PMCID: PMC5772851 DOI: 10.14336/ad.2017.0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Presbycusis (age-related hearing loss) is the most universal sensory degenerative disease in elderly people caused by the degeneration of cochlear cells. Non-coding microRNAs (miRNAs) play a fundamental role in gene regulation in almost every multicellular organism, and control the aging processes. It has been identified that various miRNAs are up- or down-regulated during mammalian aging processes in tissue-specific manners. Most miRNAs bind to specific sites on their target messenger-RNAs (mRNAs) and decrease their expression. Germline mutation may lead to dysregulation of potential miRNAs expression, causing progressive hair cell degeneration and age-related hearing loss. Therapeutic innovations could emerge from a better understanding of diverse function of miRNAs in presbycusis. This review summarizes the relationship between miRNAs and presbycusis, and presents novel miRNAs-targeted strategies against presbycusis.
Collapse
Affiliation(s)
- Weiming Hu
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Junwu Wu
- 2Department of Otolaryngology-Head and Neck Surgery, Yiwu traditional Chinese Medicine Hospital, Yiwu 322000, China.,3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| | - Wenjing Jiang
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jianguo Tang
- 3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
24
|
Gao L, Wang X, Tang Y, Huang S, Hu CAA, Teng Y. FGF19/FGFR4 signaling contributes to the resistance of hepatocellular carcinoma to sorafenib. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:8. [PMID: 28069043 PMCID: PMC5223586 DOI: 10.1186/s13046-016-0478-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/16/2016] [Indexed: 12/16/2022]
Abstract
Background Sorafenib, a multi-kinase inhibitor, is used as a standard therapy for advanced hepatocellular carcinoma (HCC). However, complete remission has not been achieved and the molecular basis of HCC resistance to sorafenib remains largely unknown. Previous studies have shown that fibroblast growth factor 19 (FGF19) expression correlates with tumor progression and poor prognosis of HCC. Here, we demonstrate the novel role of FGF19 in HCC resistance to sorafenib therapy. Methods FGF19 Knockdown cells were achieved by lentiviral-mediated interference, and FGFR4 knockout cells were achieved by CRISPR-Cas9. Protein levels of FGF19, FGFR4 and c-PARP in various HCC cell lines were measured by Western blotting analysis. Cell viability was determined by MTS assay, apoptosis was determined by DAPI nuclear staining and Western blot of c-PRAP, and ROS generation was determined by DCFH-DA staining and electrochemical biosensor. Results We showed that FGF19, when overexpressed, inhibited the effect of sorafenib on ROS generation and apoptosis in HCC. In contrast, loss of FGF19 or its receptor FGFR4 led to a remarkable increase in sorafenib-induced ROS generation and apoptosis. In addition, knockdown of FGF19 in sorafenib-resistant HCC cells significantly enhanced the sensitivity to sorafenib. Importantly, targeting FGF19/FGFR4 axis by ponatinib, a third-generation inhibitor of chronic myeloid leukemia, overcomes HCC resistance of sorafenib by enhancing ROS-associated apoptosis in sorafenib-treated HCC. Conclusion Our results provide the first evidence that inhibition of FGF19/FGFR4 signaling significantly overcomes sorafenib resistance in HCC. Co-treatment of ponatinib and sorafinib may represent an effective therapeutic approach for eradicating HCC. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0478-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lixia Gao
- Department of Oral Biology, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Xuli Wang
- Department of Radiology and Imaging Sciences, School of Medicine, University of Utah, Salt Lake City, UT, USA.,Experimental Therapeutics Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Yaoliang Tang
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shuang Huang
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Chien-An Andy Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Yong Teng
- Department of Oral Biology, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA. .,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, USA.
| |
Collapse
|
25
|
Seke M, Petrovic D, Djordjevic A, Jovic D, Borovic ML, Kanacki Z, Jankovic M. Fullerenol/doxorubicin nanocomposite mitigates acute oxidative stress and modulates apoptosis in myocardial tissue. NANOTECHNOLOGY 2016; 27:485101. [PMID: 27811390 DOI: 10.1088/0957-4484/27/48/485101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Fullerenol (C60(OH)24) is present in aqueous solutions in the form of polyanion nanoparticles with particles' size distribution within the range from 15 to 42 nm. In this research it is assumed that these features could enable fullerenol nanoparticles (FNPs) to bind positively charged molecules like doxorubicin (DOX) and serve as drug carriers. Considering this, fullerenol/doxorubicin nanocomposite (FNP/DOX) is formed and characterized by ultra-performance liquid chromatography tandem mass spectrometry, dynamic light scattering, atomic force microscopy and transmission electron microscopy. Measurements have shown that DOX did not significantly affect particle size (23 nm). It is also assumed that FNP/DOX could reduce the acute cardiotoxic effects of DOX in vivo (Wistar rats treated i.p.). In this study, quantitative real time polymerase chain reaction results have shown that treatment with DOX alone caused significant increase in mRNA levels of catalase (p < 0.05) enzyme indicating the presence of oxidative stress. This effect is significantly reduced by the treatment with FNP/DOX (p < 0.05). Furthermore, mRNA levels of antiapoptotic enzyme (Bcl-2) are significantly increased (p < 0.05) in all treated groups, particularly where FNP/DOX was applied, suggesting cell resistance to apoptosis. Moreover, ultrastructural analysis has shown the absence of myelin figures within the mitochondria in the heart tissue with FNP/DOX treatment, indicating reduction of oxidative stress. Hence, our results have implied that FNP/DOX is generally less harmful to the heart compared to DOX.
Collapse
Affiliation(s)
- Mariana Seke
- Institute of Nuclear Sciences 'Vinca', University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
26
|
Monoamine Oxidase Is Overactivated in Left and Right Ventricles from Ischemic Hearts: An Intriguing Therapeutic Target. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4375418. [PMID: 28044091 PMCID: PMC5156804 DOI: 10.1155/2016/4375418] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/19/2016] [Accepted: 10/09/2016] [Indexed: 12/21/2022]
Abstract
Growing evidence indicates that reactive oxygen species (ROS) may play a key role in human heart failure (HF). Monoamine oxidase (MAO) is emerging as a major ROS source in several cardiomyopathies. However, little is known about MAO activity in human failing heart and its relationship with redox imbalance. Therefore, we measured MAO activity in the left (LV) and in the right (RV) ventricle of human nonfailing (NF) and in end-stage ischemic (IHD) and nonischemic failing hearts. We found that both MAO isoforms (MAO-A/B) significantly increased in terms of activity and expression levels only in IHD ventricles. Catalase and aldehyde dehydrogenase-2 activities (ALDH-2), both implicated in MAO-catalyzed catecholamine catabolism, were significantly elevated in the failing LV, whereas, in the RV, statistical significance was observed only for ALDH-2. Oxidative stress markers levels were significantly increased only in the failing RV. Actin oxidation was significantly elevated in both failing ventricles and related to MAO-A activity and to functional parameters. These data suggest a close association between MAO-A-dependent ROS generation, actin oxidation, and ventricular dysfunction. This latter finding points to a possible pathogenic role of MAO-A in human myocardial failure supporting the idea that MAO-A could be a new therapeutic target in HF.
Collapse
|
27
|
Hou J, Wan B, Yang Y, Ren XM, Guo LH, Liu JF. Biodegradation of Single-Walled Carbon Nanotubes in Macrophages through Respiratory Burst Modulation. Int J Mol Sci 2016; 17:409. [PMID: 27011169 PMCID: PMC4813264 DOI: 10.3390/ijms17030409] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 12/29/2022] Open
Abstract
The biodegradation of carbon nanotubes (CNTs) may be one of major determinants of the toxic outcomes in exposed individuals. In this study, we employed a macrophage/monocyte model, Raw264.7, to investigate the feasibility of regulating the biodegradation of three types of single-walled carbon nanotubes (SWCNTs) (pristine, ox-, and OH-SWCNTs) by respiratory burst modulation. An artificial fluid mimicking the enzymatic reactions of respiratory burst was constituted to reveal the role of respiratory burst played in SWCNT biodegradation. The biodegradation of SWCNTs were characterized by Raman, ultraviolet-visible-near-infrared spectroscopy, and transmission electron microscopy. Our results showed significantly accelerated biodegradation of ox-SWCNTs and OH-SWCNTs in macrophages activated by phorbol myristate acetate (PMA), which could be prevented by N-acetyl-l-cysteine (NAC), whereas p-SWCNTs were resistant to biodegradation. Similar tendencies were observed by using the in vitro enzymatic system, and the degradation rates of these SWCNTs are in the order of OH-SWCNTs > ox-SWCNTs >> p-SWCNTs, suggesting a pivotal role of respiratory burst in accelerating the biodegradation of SWCNTs and that defect sites on SWCNTs might be a prerequisite for the biodegradation to occur. Our findings might provide invaluable clues on the development of intervention measurements for relieving the side effects of SWCNTs and would help to design safer SWCNT products with higher biodegradability and less toxicity.
Collapse
Affiliation(s)
- Jie Hou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Bin Wan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Yu Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Xiao-Min Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Liang-Hong Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China.
| | - Jing-Fu Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
28
|
Otaki Y, Takahashi H, Watanabe T, Funayama A, Netsu S, Honda Y, Narumi T, Kadowaki S, Hasegawa H, Honda S, Arimoto T, Shishido T, Miyamoto T, Kamata H, Nakajima O, Kubota I. HECT-Type Ubiquitin E3 Ligase ITCH Interacts With Thioredoxin-Interacting Protein and Ameliorates Reactive Oxygen Species-Induced Cardiotoxicity. J Am Heart Assoc 2016; 5:JAHA.115.002485. [PMID: 26796253 PMCID: PMC4859366 DOI: 10.1161/jaha.115.002485] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background The homologous to the E6‐AP carboxyl terminus (HECT)–type ubiquitin E3 ligase ITCH is an enzyme that plays a pivotal role in posttranslational modification by ubiquitin proteasomal protein degradation. Thioredoxin‐interacting protein (TXNIP) is a negative regulator of the thioredoxin system and an endogenous reactive oxygen species scavenger. In the present study, we focused on the functional role of ubiquitin E3 ligase ITCH and its interaction with TXNIP to elucidate the mechanism of cardiotoxicity induced by reactive oxygen species, such as doxorubicin and hydrogen peroxide. Methods and Results Protein interaction between TXNIP and ITCH in cardiomyocyte was confirmed by immunoprecipitation assays. Overexpression of ITCH increased proteasomal TXNIP degradation and augmented thioredoxin activity, leading to inhibition of reactive oxygen species generation, p38 MAPK, p53, and subsequent intrinsic pathway cardiomyocyte apoptosis in reactive oxygen species–induced cardiotoxicity. Conversely, knockdown of ITCH using small interfering RNA inhibited TXNIP degradation and resulted in a subsequent increase in cardiomyocyte apoptosis. Next, we generated a transgenic mouse with cardiac‐specific overexpression of ITCH, called the ITCH‐Tg mouse. The expression level of TXNIP in the myocardium in ITCH‐Tg mice was significantly lower than WT littermates. In ITCH‐Tg mice, cardiac dysfunction and remodeling were restored compared with WT littermates after doxorubicin injection and myocardial infarction surgery. Kaplan–Meier analysis revealed that ITCH‐Tg mice had a higher survival rate than WT littermates after doxorubicin injection and myocardial infarction surgery. Conclusion We demonstrated, for the first time, that ITCH targets TXNIP for ubiquitin‐proteasome degradation in cardiomyocytes and ameliorates reactive oxygen species–induced cardiotoxicity through the thioredoxin system.
Collapse
Affiliation(s)
- Yoichiro Otaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hiroki Takahashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Akira Funayama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shunsuke Netsu
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Yuki Honda
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Taro Narumi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shinpei Kadowaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hiromasa Hasegawa
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shintaro Honda
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Takanori Arimoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Tetsuro Shishido
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Takuya Miyamoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hideaki Kamata
- Laboratory of Biomedical Chemistry, Department of Molecular Medical Science, Graduate School of Medicine, University of Hiroshima, Japan (H.K.)
| | - Osamu Nakajima
- Research Laboratory for Molecular Genetics, Yamagata University School of Medicine, Yamagata, Japan (O.N.)
| | - Isao Kubota
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| |
Collapse
|
29
|
Statins and oxidative stress in chronic heart failure. Rev Port Cardiol 2016; 35:41-57. [PMID: 26763895 DOI: 10.1016/j.repc.2015.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/13/2015] [Indexed: 11/23/2022] Open
Abstract
Statins are the most commonly prescribed drugs for the treatment of dyslipidemia. They are also recommended in primary and secondary prevention of cardiovascular disease. In addition to decreasing cholesterol synthesis, statins interfere with the synthesis of isoprenoid intermediates, which may explain many of their pleiotropic properties, including their antioxidant effects. Oxidative stress is defined as an imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems, with a prevailing pro-oxidant status that results in macromolecular damage and disruption of cellular redox signaling. Reactive oxygen species interfere with various processes that affect cardiac structure and function, contributing to the contractile dysfunction, myocardial hypertrophy and fibrosis observed in the pathophysiology of heart failure. By regulating several molecular pathways that control nicotinamide adenine dinucleotide phosphate oxidase and endothelial nitric oxide synthase activity, statins help restore redox homeostasis. These drugs also contribute to the control of inflammation and appear to have a protective role in various diseases. The results of observational studies and clinical trials with statins in heart failure have not been consensual. This review aims to analyze the role of oxidative stress in heart failure and the molecular mechanisms underlying statins' antioxidant properties. It also examines current scientific evidence on the use of these drugs as a specific treatment for heart failure.
Collapse
|
30
|
Statins and oxidative stress in chronic heart failure. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.repce.2015.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
31
|
Regulation of cellular oxidative stress and apoptosis by G protein-coupled receptor kinase-2; The role of NADPH oxidase 4. Cell Signal 2015; 28:190-203. [PMID: 26631573 DOI: 10.1016/j.cellsig.2015.11.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/13/2015] [Accepted: 11/25/2015] [Indexed: 11/23/2022]
Abstract
Cardiac myocyte oxidative stress and apoptosis are considered important mechanisms for the development of heart failure (HF). Chronic HF is characterized by increased circulating catecholamines to augment cardiac output. Long-term stimulation of myocardial β-adrenergic receptors (β-ARs) is deleterious in cardiac myocytes, however, the potential mechanisms underlying increased cell death are unclear. We hypothesize that GRK2, a critical regulator of myocardial β-AR signaling, plays an important role in mediating cellular oxidative stress and apoptotic cell death in response to β-agonist stimulation. Stimulation of H9c2 cells with a non-selective β-agonist, isoproterenol (Iso) caused increased oxidative stress and apoptosis. There was also increased Nox4 expression, but no change in Nox2, the primary NADPH isoforms and major sources of ROS generation in cardiac myocytes. Adenoviral-mediated overexpression of GRK2 led to similar increases in ROS production and apoptosis as seen with Iso stimulation. These increases in oxidative stress were abolished by pre-treatment with the non-specific Nox inhibitor, apocynin, or siRNA knockdown of Nox4. Adenoviral-mediated expression of a GRK2 inhibitor prevented ROS production and apoptosis in response to Iso stimulation. β-Arrestins are signaling proteins that function downstream of GRK2 in β-AR uncoupling. Adenoviral-mediated overexpression of β-arrestins increased ROS production and Nox4 expression. Chronic β-agonist stimulation in mice increased Nox4 expression and apoptosis compared to PBS or AngII treatment. These data demonstrate that GRK2 may play an important role in regulating oxidative stress and apoptosis in cardiac myocytes and provides an additional novel mechanism for the beneficial effects of cardiac-targeted GRK2 inhibition to prevent the development of HF.
Collapse
|
32
|
Philip JL, Razzaque MA, Han M, Li J, Theccanat T, Xu X, Akhter SA. Regulation of mitochondrial oxidative stress by β-arrestins in cultured human cardiac fibroblasts. Dis Model Mech 2015; 8:1579-89. [PMID: 26449263 PMCID: PMC4728312 DOI: 10.1242/dmm.019968] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/24/2015] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress in cardiac fibroblasts (CFs) promotes transformation to myofibroblasts and collagen synthesis leading to myocardial fibrosis, a precursor to heart failure (HF). NADPH oxidase 4 (Nox4) is a major source of cardiac reactive oxygen species (ROS); however, mechanisms of Nox4 regulation are unclear. β-arrestins are scaffold proteins that signal in G-protein-dependent and -independent pathways; for example, in ERK activation. We hypothesize that β-arrestins regulate oxidative stress in a Nox4-dependent manner and increase fibrosis in HF. CFs were isolated from normal and failing adult human left ventricles. Mitochondrial ROS/superoxide production was quantitated using MitoSox. β-arrestin and Nox4 expressions were manipulated using adenoviral overexpression or short interfering RNA (siRNA)-mediated knockdown. Mitochondrial oxidative stress and Nox4 expression in CFs were significantly increased in HF. Nox4 knockdown resulted in inhibition of mitochondrial superoxide production and decreased basal and TGF-β-stimulated collagen and α-SMA expression. CF β-arrestin expression was upregulated fourfold in HF. β-arrestin knockdown in failing CFs decreased ROS and Nox4 expression by 50%. β-arrestin overexpression in normal CFs increased mitochondrial superoxide production twofold. These effects were prevented by inhibition of either Nox or ERK. Upregulation of Nox4 seemed to be a primary mechanism for increased ROS production in failing CFs, which stimulates collagen deposition. β-arrestin expression was upregulated in HF and plays an important and newly identified role in regulating mitochondrial superoxide production via Nox4. The mechanism for this effect seems to be ERK-mediated. Targeted inhibition of β-arrestins in CFs might decrease oxidative stress as well as pathological cardiac fibrosis. Summary: β-arrestins regulate oxidative stress in a Nox4-dependent manner leading to increased extracellular-matrix protein synthesis by cardiac fibroblasts (CFs). Targeted inhibition of β-arrestins in CFs might decrease pathological fibrosis.
Collapse
Affiliation(s)
- Jennifer L Philip
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Md Abdur Razzaque
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Mei Han
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Jinju Li
- Section of Cardiac and Thoracic Surgery, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tiju Theccanat
- Section of Cardiac and Thoracic Surgery, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xianyao Xu
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Shahab A Akhter
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| |
Collapse
|
33
|
Huang L, Zheng M, Zhou Y, Zhu J, Zhu M, Zhao F, Cui S. Tanshinone IIA attenuates cardiac dysfunction in endotoxin-induced septic mice via inhibition of NADPH oxidase 2-related signaling pathway. Int Immunopharmacol 2015. [DOI: 10.1016/j.intimp.2015.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Oxidative stress and inflammatory markers - the future of heart failure diagnostics? POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2015; 12:145-9. [PMID: 26336497 PMCID: PMC4550016 DOI: 10.5114/kitp.2015.52856] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 04/30/2015] [Accepted: 05/18/2015] [Indexed: 01/14/2023]
Abstract
Heart failure remains one of the most important problems in cardiology despite the progress in its treatment. A number of recent studies have demonstrated the relationship between the intensification of oxidative stress and chronic inflammation and the severity of left ventricular dysfunction, development of heart failure symptoms, and prediction of future cardiac events. Early detection of changes developing in the heart is key in improving the treatment's effectiveness. It appears that determining specific, sensitive biomarkers reflecting the complex pathophysiology of heart failure and using them to detect asymptomatic cardiac alterations may become a crucial screening tool, assisting in the identification of patients requiring further diagnostic examinations. This article presents an overview of the current knowledge of the role of oxidative stress and inflammation in heart failure; it also discusses the potential role of oxidative stress and inflammatory markers as prognostic factors in heart failure that may be used in screening tests.
Collapse
|
35
|
Chia KKM, Liu CC, Hamilton EJ, Garcia A, Fry NA, Hannam W, Figtree GA, Rasmussen HH. Stimulation of the cardiac myocyte Na+-K+ pump due to reversal of its constitutive oxidative inhibition. Am J Physiol Cell Physiol 2015; 309:C239-50. [PMID: 26084308 DOI: 10.1152/ajpcell.00392.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 06/09/2015] [Indexed: 11/22/2022]
Abstract
Protein kinase C can activate NADPH oxidase and induce glutathionylation of the β1-Na(+)-K(+) pump subunit, inhibiting activity of the catalytic α-subunit. To examine if signaling of nitric oxide-induced soluble guanylyl cyclase (sGC)/cGMP/protein kinase G can cause Na(+)-K(+) pump stimulation by counteracting PKC/NADPH oxidase-dependent inhibition, cardiac myocytes were exposed to ANG II to activate NADPH oxidase and inhibit Na(+)-K(+) pump current (Ip). Coexposure to 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) to stimulate sGC prevented the decrease of Ip. Prevention of the decrease was abolished by inhibition of protein phosphatases (PP) 2A but not by inhibition of PP1, and it was reproduced by an activator of PP2A. Consistent with a reciprocal relationship between β1-Na(+)-K(+) pump subunit glutathionylation and pump activity, YC-1 decreased ANG II-induced β1-subunit glutathionylation. The decrease induced by YC-1 was abolished by a PP2A inhibitor. YC-1 decreased phosphorylation of the cytosolic p47(phox) NADPH oxidase subunit and its coimmunoprecipitation with the membranous p22(phox) subunit, and it decreased O2 (·-)-sensitive dihydroethidium fluorescence of myocytes. Addition of recombinant PP2A to myocyte lysate decreased phosphorylation of p47(phox) indicating the subunit could be a substrate for PP2A. The effects of YC-1 to decrease coimmunoprecipitation of p22(phox) and p47(phox) NADPH oxidase subunits and decrease β1-Na(+)-K(+) pump subunit glutathionylation were reproduced by activation of nitric oxide-dependent receptor signaling. We conclude that sGC activation in cardiac myocytes causes a PP2A-dependent decrease in NADPH oxidase activity and a decrease in β1 pump subunit glutathionylation. This could account for pump stimulation with neurohormonal oxidative stress expected in vivo.
Collapse
Affiliation(s)
- Karin K M Chia
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia; Royal Brisbane and Women's Hospital, The University of Queensland, Queensland, Australia; and
| | - Chia-Chi Liu
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia
| | - Elisha J Hamilton
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia
| | - Alvaro Garcia
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia
| | - Natasha A Fry
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia
| | - William Hannam
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, St. Leonards, Australia
| | - Helge H Rasmussen
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, St. Leonards, Australia
| |
Collapse
|
36
|
Thakur A, Alam MJ, Ajayakumar MR, Ghaskadbi S, Sharma M, Goswami SK. Norepinephrine-induced apoptotic and hypertrophic responses in H9c2 cardiac myoblasts are characterized by different repertoire of reactive oxygen species generation. Redox Biol 2015; 5:243-252. [PMID: 26070033 PMCID: PMC4477046 DOI: 10.1016/j.redox.2015.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 01/04/2023] Open
Abstract
Despite recent advances, the role of ROS in mediating hypertrophic and apoptotic responses in cardiac myocytes elicited by norepinephrine (NE) is rather poorly understood. We demonstrate through our experiments that H9c2 cardiac myoblasts treated with 2 µM NE (hypertrophic dose) generate DCFH-DA positive ROS only for 2 h; while those treated with 100 µM NE (apoptotic dose) sustains generation for 48 h, followed by apoptosis. Though the levels of DCFH fluorescence were comparable at early time points in the two treatment sets, its quenching by DPI, catalase and MnTmPyP suggested the existence of a different repertoire of ROS. Both doses of NE also induced moderate levels of H2O2 but with different kinetics. Sustained but intermittent generation of highly reactive species detectable by HPF was seen in both treatment sets but no peroxynitrite was generated in either conditions. Sustained generation of hydroxyl radicals with no appreciable differences were noticed in both treatment sets. Nevertheless, despite similar profile of ROS generation between the two conditions, extensive DNA damage as evident from the increase in 8-OH-dG content, formation of γ-H2AX and PARP cleavage was seen only in cells treated with the higher dose of NE. We therefore conclude that hypertrophic and apoptotic doses of NE generate distinct but comparable repertoire of ROS/RNS leading to two very distinct downstream responses. H9c2 myoblasts upon treatment with 2 and 100 µM NE induces hypertrophy and apoptosis. Both treatments show comparable levels of DCFH fluorescence with different kinetics. Both treatments show comparable levels of HPF fluorescence in an oscillating manner. More hydroxyl radical was generated in 100 µM NE treated set. DNA damage and apoptosis occurs only in 100 µM NE treated sets.
Collapse
Affiliation(s)
- Anita Thakur
- Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Md Jahangir Alam
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - M R Ajayakumar
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | | - Manish Sharma
- Defence Institute of Physiology & Allied Sciences, New Delhi 110054, India
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
37
|
Monoamine Oxidases as Potential Contributors to Oxidative Stress in Diabetes: Time for a Study in Patients Undergoing Heart Surgery. BIOMED RESEARCH INTERNATIONAL 2015; 2015:515437. [PMID: 26101773 PMCID: PMC4458524 DOI: 10.1155/2015/515437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/01/2014] [Accepted: 09/17/2014] [Indexed: 12/19/2022]
Abstract
Oxidative stress is a pathomechanism causally linked to the progression of chronic cardiovascular diseases and diabetes. Mitochondria have emerged as the most relevant source of reactive oxygen species, the major culprit being classically considered the respiratory chain at the inner mitochondrial membrane. In the past decade, several experimental studies unequivocally demonstrated the contribution of monoamine oxidases (MAOs) at the outer mitochondrial membrane to the maladaptative ventricular hypertrophy and endothelial dysfunction. This paper addresses the contribution of mitochondrial dysfunction to the pathogenesis of heart failure and diabetes together with the mounting evidence for an emerging role of MAO inhibition as putative cardioprotective strategy in both conditions.
Collapse
|
38
|
Li B, Tian J, Sun Y, Xu TR, Chi RF, Zhang XL, Hu XL, Zhang YA, Qin FZ, Zhang WF. Activation of NADPH oxidase mediates increased endoplasmic reticulum stress and left ventricular remodeling after myocardial infarction in rabbits. Biochim Biophys Acta Mol Basis Dis 2015; 1852:805-15. [DOI: 10.1016/j.bbadis.2015.01.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 12/21/2022]
|
39
|
Gorin Y, Wauquier F. Upstream regulators and downstream effectors of NADPH oxidases as novel therapeutic targets for diabetic kidney disease. Mol Cells 2015; 38:285-96. [PMID: 25824546 PMCID: PMC4400302 DOI: 10.14348/molcells.2015.0010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress has been linked to the pathogenesis of diabetic nephropathy, the complication of diabetes in the kidney. NADPH oxidases of the Nox family, and in particular the homologue Nox4, are a major source of reactive oxygen species in the diabetic kidney and are critical mediators of redox signaling in glomerular and tubulointerstitial cells exposed to the diabetic milieu. Here, we present an overview of the current knowledge related to the understanding of the role of Nox enzymes in the processes that control mesangial cell, podocyte and tubulointerstitial cell injury induced by hyperglycemia and other predominant factors enhanced in the diabetic milieu, including the renin-angiotensin system and transforming growth factor-β. The nature of the upstream modulators of Nox enzymes as well as the downstream targets of the Nox NADPH oxidases implicated in the propagation of the redox processes that alter renal biology in diabetes will be highlighted.
Collapse
Affiliation(s)
- Yves Gorin
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas,
USA
| | - Fabien Wauquier
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas,
USA
| |
Collapse
|
40
|
Asrar Ul Haq M, Goh CY, Levinger I, Wong C, Hare DL. Clinical utility of exercise training in heart failure with reduced and preserved ejection fraction. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2015; 9:1-9. [PMID: 25698883 PMCID: PMC4324467 DOI: 10.4137/cmc.s21372] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/29/2014] [Accepted: 01/04/2015] [Indexed: 12/13/2022]
Abstract
Reduced exercise tolerance is an independent predictor of hospital readmission and mortality in patients with heart failure (HF). Exercise training for HF patients is well established as an adjunct therapy, and there is sufficient evidence to support the favorable role of exercise training programs for HF patients over and above the optimal medical therapy. Some of the documented benefits include improved functional capacity, quality of life (QoL), fatigue, and dyspnea. Major trials to assess exercise training in HF have, however, focused on heart failure with reduced ejection fraction (HFREF). At least half of the patients presenting with HF have heart failure with preserved ejection fraction (HFPEF) and experience similar symptoms of exercise intolerance, dyspnea, and early fatigue, and similar mortality risk and rehospitalization rates. The role of exercise training in the management of HFPEF remains less clear. This article provides a brief overview of pathophysiology of reduced exercise tolerance in HFREF and heart failure with preserved ejection fraction (HFPEF), and summarizes the evidence and mechanisms by which exercise training can improve symptoms and HF. Clinical and practical aspects of exercise training prescription are also discussed.
Collapse
Affiliation(s)
- Muhammad Asrar Ul Haq
- Northern Heart, The Northern Hospital, Melbourne, Vic, Australia
- Austin Health, Melbourne, Vic, Australia
- University of Melbourne, Melbourne, Vic, Australia
| | - Cheng Yee Goh
- Northern Heart, The Northern Hospital, Melbourne, Vic, Australia
| | - Itamar Levinger
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, Australia
| | - Chiew Wong
- Northern Heart, The Northern Hospital, Melbourne, Vic, Australia
- University of Melbourne, Melbourne, Vic, Australia
| | - David L Hare
- Austin Health, Melbourne, Vic, Australia
- University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
41
|
Kazama K, Okada M, Yamawaki H. Adipocytokine, omentin inhibits doxorubicin-induced H9c2 cardiomyoblasts apoptosis through the inhibition of mitochondrial reactive oxygen species. Biochem Biophys Res Commun 2015; 457:602-7. [DOI: 10.1016/j.bbrc.2015.01.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/11/2015] [Indexed: 01/21/2023]
|
42
|
Edwardson DW, Narendrula R, Chewchuk S, Mispel-Beyer K, Mapletoft JPJ, Parissenti AM. Role of Drug Metabolism in the Cytotoxicity and Clinical Efficacy of Anthracyclines. Curr Drug Metab 2015; 16:412-26. [PMID: 26321196 PMCID: PMC5398089 DOI: 10.2174/1389200216888150915112039] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 01/19/2023]
Abstract
Many clinical studies involving anti-tumor agents neglect to consider how these agents are metabolized within the host and whether the creation of specific metabolites alters drug therapeutic properties or toxic side effects. However, this is not the case for the anthracycline class of chemotherapy drugs. This review describes the various enzymes involved in the one electron (semi-quinone) or two electron (hydroxylation) reduction of anthracyclines, or in their reductive deglycosidation into deoxyaglycones. The effects of these reductions on drug antitumor efficacy and toxic side effects are also discussed. Current evidence suggests that the one electron reduction of anthracyclines augments both their tumor toxicity and their toxicity towards the host, in particular their cardiotoxicity. In contrast, the two electron reduction (hydroxylation) of anthracyclines strongly reduces their ability to kill tumor cells, while augmenting cardiotoxicity through their accumulation within cardiomyocytes and their direct effects on excitation/contraction coupling within the myocytes. The reductive deglycosidation of anthracyclines appears to inactivate the drug and only occurs under rare, anaerobic conditions. This knowledge has resulted in the identification of important new approaches to improve the therapeutic index of anthracyclines, in particular by inhibiting their cardiotoxicity. The true utility of these approaches in the management of cancer patients undergoing anthracycline-based chemotherapy remains unclear, although one such agent (the iron chelator dexrazoxane) has recently been approved for clinical use.
Collapse
Affiliation(s)
| | | | | | | | | | - Amadeo M Parissenti
- Dept. of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|
43
|
Activation of volume-sensitive outwardly rectifying chloride channel by ROS contributes to ER stress and cardiac contractile dysfunction: involvement of CHOP through Wnt. Cell Death Dis 2014; 5:e1528. [PMID: 25412307 PMCID: PMC4260737 DOI: 10.1038/cddis.2014.479] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum (ER) stress occurring in stringent conditions is critically involved in cardiomyocytes apoptosis and cardiac contractile dysfunction (CCD). However, the molecular machinery that mediates cardiac ER stress and subsequent cell death remains to be fully deciphered, which will hopefully provide novel therapeutic targets for these disorders. Here, we establish tunicamycin-induced model of cardiomyocyte ER stress, which effectively mimicks pathological stimuli to trigger CCD. Tunicamycin activates volume-sensitive outward rectifying Cl− currents. Blockade of the volume-sensitive outwardly rectifying (VSOR) Cl− channel by 4,4'-diisothiocya-natostilbene-2,2'-disulfonic acid (DIDS), a non-selective Cl− channel blocker, and 4-(2-butyl-6,7-dichlor-2-cyclopentyl-indan-1-on-5-yl) oxybutyric acid (DCPIB), a selective VSOR Cl− channel blocker, improves cardiac contractility, which correlates with suppressed ER stress through inhibiting the canonical GRP78/eIF2α/ATF4 and XBP1 pathways, and promotes survival of cardiomyocytes by inverting tunicamycin-induced decrease of Wnt through the CHOP pathway. VSOR activation of tunicamycin-treated cardiomyocytes is attributed to increased intracellular levels of reactive oxygen species (ROS). Our study demonstrates a pivotal role of ROS/VSOR in mediating ER stress and functional impairment of cardiomyocytes via the CHOP-Wnt pathway, and suggests the therapeutic values of VSOR Cl− channel blockers against ER stress-associated cardiac anomalies.
Collapse
|
44
|
Kurdi M, Booz GW. Carvedilol protects the infarcted heart by upregulating miR-133: first evidence that disease state affects β-adrenergic arrestin-biased signaling? J Mol Cell Cardiol 2014; 76:12-4. [PMID: 25128784 DOI: 10.1016/j.yjmcc.2014.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 08/05/2014] [Indexed: 01/14/2023]
Affiliation(s)
- Mazen Kurdi
- Department of Chemistry and Biochemistry, Faculty of Sciences, Lebanese University, Rafic Hariri Educational Campus, Hadath, Lebanon; Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, USA; The Mississippi Center for Heart Research, The University of Mississippi Medical Center, Jackson, MS, USA; The Cardiovascular-Renal Research Center, The University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, USA; The Mississippi Center for Heart Research, The University of Mississippi Medical Center, Jackson, MS, USA; The Cardiovascular-Renal Research Center, The University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
45
|
Propofol inhibits lipopolysaccharide-induced tumor necrosis factor-alpha expression and myocardial depression through decreasing the generation of superoxide anion in cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:157376. [PMID: 25180066 PMCID: PMC4144395 DOI: 10.1155/2014/157376] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/16/2014] [Indexed: 11/17/2022]
Abstract
TNF-α has been shown to be a major factor responsible for myocardial depression in sepsis. The aim of this study was to investigate the effect of an anesthetic, propofol, on TNF-α expression in cardiomyocytes treated with LPS both in vivo and in vitro. In cultured cardiomyocytes, compared with control group, propofol significantly reduced protein expression of gp91phox and phosphorylation of extracellular regulated protein kinases 1/2 (ERK1/2) and p38 MAPK, which associates with reduced TNF-α production. In in vivo mice studies, propofol significantly improved myocardial depression and increased survival rate of mice after LPS treatment or during endotoxemia, which associates with reduced myocardial TNF-α production, gp91phox, ERK1/2, and p38 MAPK. It is concluded that propofol abrogates LPS-induced TNF-α production and alleviates cardiac depression through gp91phox/ERK1/2 or p38 MAPK signal pathway. These findings have great clinical importance in the application of propofol for patients enduring sepsis.
Collapse
|
46
|
Infantino V, Iacobazzi V, Menga A, Avantaggiati ML, Palmieri F. A key role of the mitochondrial citrate carrier (SLC25A1) in TNFα- and IFNγ-triggered inflammation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1217-1225. [PMID: 25072865 DOI: 10.1016/j.bbagrm.2014.07.013] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/02/2014] [Accepted: 07/18/2014] [Indexed: 12/20/2022]
Abstract
The chronic induction of inflammation underlies multiple pathological conditions, including metabolic, autoimmune disorders and cancer. The mitochondrial citrate carrier (CIC), encoded by the SLC25A1 gene, promotes the export of citrate from the mitochondria to the cytoplasm, a process that profoundly influences energy balance in the cells. We have previously shown that SLC25A1 is a target gene for lipopolysaccharide signaling and promotes the production of inflammatory mediators. We now demonstrate that SLC25A1 is induced at the transcriptional level by two key pro-inflammatory cytokines, tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ), and such induction involves the activity of the nuclear factor kappa B and STAT1 transcription factors. By studying the down-stream events following SLC25A1 activation during signals that mimic inflammation, we demonstrate that CIC is required for regulating the levels of nitric oxide and of prostaglandins by TNFα or IFNγ. Importantly, we show that the citrate exported from mitochondria via CIC and its downstream metabolic intermediate, acetyl-coenzyme A, are necessary for TNFα or IFNγ to induce nitric oxide and prostaglandin production. These findings provide the first line of evidence that the citrate export pathway, via CIC, is central for cytokine-induced inflammatory signals and shed new light on the relationship between energy metabolism and inflammation.
Collapse
Affiliation(s)
- Vittoria Infantino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, via Orabona 4, 70125 Bari, Italy.,Department of Science, University of Basilicata, via N. Sauro 85, 85100 Potenza, Italy
| | - Vito Iacobazzi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
47
|
Desrois M, Kober F, Lan C, Dalmasso C, Cole M, Clarke K, Cozzone PJ, Bernard M. Effect of isoproterenol on myocardial perfusion, function, energy metabolism and nitric oxide pathway in the rat heart - a longitudinal MR study. NMR IN BIOMEDICINE 2014; 27:529-538. [PMID: 24677605 DOI: 10.1002/nbm.3088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/06/2014] [Accepted: 01/09/2014] [Indexed: 06/03/2023]
Abstract
The chronic administration of the β-adrenoreceptor agonist isoproterenol (IsoP) is used in animals to study the mechanisms of cardiac hypertrophy and failure associated with a sustained increase in circulating catecholamines. Time-dependent changes in myocardial blood flow (MBF), morphological and functional parameters were assessed in rats in vivo using multimodal cardiac MRI. Energy metabolism, oxidative stress and the nitric oxide (NO) pathway were evaluated in isolated perfused rat hearts following 7 days of treatment. Male Wistar rats were infused for 7 days with IsoP or vehicle using osmotic pumps. Cine-MRI and arterial spin labeling were used to determine left ventricular morphology, function and MBF at days 1, 2 and 7 after pump implantation. Isolated hearts were then perfused, and high-energy phosphate compounds and intracellular pH were followed using ³¹P MRS with simultaneous measurement of contractile function. Total creatine and malondialdehyde (MDA) contents were measured by high-performance liquid chromatography. The NO pathway was evaluated by NO synthase isoform expression and total nitrate concentration (NO(x)). In IsoP-treated rats, left ventricular mass was increased at day 1 and maintained. Wall thickness was increased with a peak at day 2 and a tendency to return to baseline values at day 7. MBF was markedly increased at day 1 and returned to normal values between days 1 and 2. The rate-pressure product and phosphocreatine/adenosine triphosphate ratio in perfused hearts were reduced. MDA, endothelial NO synthase expression and NO(x) were increased. Sustained high cardiac function and normal MBF after 24 h of IsoP infusion indicate imbalance between functional demand and blood flow, leading to morphological changes. After 1 week, cardiac hypertrophy and decreased function were associated with impaired phosphocreatine, increased oxidative stress and up-regulation of the NO pathway. These results provide supplemental information on the evolution of the different contributing factors leading to morphological and functional changes in this model of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Martine Desrois
- Aix-Marseille Université UMR CNRS n°7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Faculté de Médecine, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Corradi F, Paolini L, De Caterina R. Ranolazine in the prevention of anthracycline cardiotoxicity. Pharmacol Res 2014; 79:88-102. [DOI: 10.1016/j.phrs.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 11/06/2013] [Accepted: 11/06/2013] [Indexed: 12/19/2022]
|
49
|
Zehendner CM, Librizzi L, Hedrich J, Bauer NM, Angamo EA, de Curtis M, Luhmann HJ. Moderate hypoxia followed by reoxygenation results in blood-brain barrier breakdown via oxidative stress-dependent tight-junction protein disruption. PLoS One 2013; 8:e82823. [PMID: 24324834 PMCID: PMC3855783 DOI: 10.1371/journal.pone.0082823] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 10/28/2013] [Indexed: 11/19/2022] Open
Abstract
Re-canalization of cerebral vessels in ischemic stroke is pivotal to rescue dysfunctional brain areas that are exposed to moderate hypoxia within the penumbra from irreversible cell death. Goal of the present study was to evaluate the effect of moderate hypoxia followed by reoxygenation (MHR) on the evolution of reactive oxygen species (ROS) and blood-brain barrier (BBB) integrity in brain endothelial cells (BEC). BBB integrity was assessed in BEC in vitro and in microvessels of the guinea pig whole brain in situ preparation. Probes were exposed to MHR (2 hours 67-70 mmHg O2, 3 hours reoxygenation, BEC) or towards occlusion of the arteria cerebri media (MCAO) with or without subsequent reperfusion in the whole brain preparation. In vitro BBB integrity was evaluated using trans-endothelial electrical resistance (TEER) and transwell permeability assays. ROS in BEC were evaluated using 2',7'-dichlorodihydrofluorescein diacetate (DCF), MitoSox and immunostaining for nitrotyrosine. Tight-junction protein (TJ) integrity in BEC, stainings for nitrotyrosine and FITC-albumin extravasation in the guinea pig brain preparation were assessed by confocal microscopy. Diphenyleneiodonium (DPI) was used to investigate NADPH oxidase dependent ROS evolution and its effect on BBB parameters in BEC. MHR impaired TJ proteins zonula occludens 1 (ZO-1) and claudin 5 (Cl5), decreased TEER, and significantly increased cytosolic ROS in BEC. These events were blocked by the NADPH oxidase inhibitor DPI. MCAO with or without subsequent reoxygenation resulted in extravasation of FITC-albumin and ROS generation in the penumbra region of the guinea pig brain preparation and confirmed BBB damage. BEC integrity may be impaired through ROS in MHR on the level of TJ and the BBB is also functionally impaired in moderate hypoxic conditions followed by reperfusion in a complex guinea pig brain preparation. These findings suggest that the BBB is susceptible towards MHR and that ROS play a key role in this process.
Collapse
Affiliation(s)
- Christoph M. Zehendner
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- * E-mail:
| | - Laura Librizzi
- Unit of Experimental Neurophysiology and Epileptology, Fondazione Istituto Neurologico Carlo Besta, Milano, Italy
| | - Jana Hedrich
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Nina M. Bauer
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Eskedar A. Angamo
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Marco de Curtis
- Unit of Experimental Neurophysiology and Epileptology, Fondazione Istituto Neurologico Carlo Besta, Milano, Italy
| | - Heiko J. Luhmann
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
50
|
Joshi M, Kotha SR, Malireddy S, Selvaraju V, Satoskar AR, Palesty A, McFadden DW, Parinandi NL, Maulik N. Conundrum of pathogenesis of diabetic cardiomyopathy: role of vascular endothelial dysfunction, reactive oxygen species, and mitochondria. Mol Cell Biochem 2013; 386:233-49. [PMID: 24307101 DOI: 10.1007/s11010-013-1861-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/09/2013] [Indexed: 12/11/2022]
Abstract
Diabetic cardiomyopathy and heart failure have been recognized as the leading causes of mortality among diabetics. Diabetic cardiomyopathy has been characterized primarily by the manifestation of left ventricular dysfunction that is independent of coronary artery disease and hypertension among the patients affected by diabetes mellitus. A complex array of contributing factors including the hypertrophy of left ventricle, alterations of metabolism, microvascular pathology, insulin resistance, fibrosis, apoptotic cell death, and oxidative stress have been implicated in the pathogenesis of diabetic cardiomyopathy. Nevertheless, the exact mechanisms underlying the pathogenesis of diabetic cardiomyopathy are yet to be established. The critical involvement of multifarious factors including the vascular endothelial dysfunction, microangiopathy, reactive oxygen species (ROS), oxidative stress, mitochondrial dysfunction has been identified in the mechanism of pathogenesis of diabetic cardiomyopathy. Although it is difficult to establish how each factor contributes to disease, the involvement of ROS and mitochondrial dysfunction are emerging as front-runners in the mechanism of pathogenesis of diabetic cardiomyopathy. This review highlights the role of vascular endothelial dysfunction, ROS, oxidative stress, and mitochondriopathy in the pathogenesis of diabetic cardiomyopathy. Furthermore, the review emphasizes that the puzzle has to be solved to firmly establish the mitochondrial and/or ROS mechanism(s) by identifying their most critical molecular players involved at both spatial and temporal levels in diabetic cardiomyopathy as targets for specific and effective pharmacological/therapeutic interventions.
Collapse
Affiliation(s)
- Mandip Joshi
- Department of Surgery, University of Connecticut Health Center, Farmington Avenue, Farmington, CT, 06032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|