1
|
He X, Sun H, Zhao Y, Fu X, Wang M, Liu M, Su Y, Hu F, Qin P, Zhang M, Hu D. Association of environmental metallic and metalloid contaminants with cardiovascular and all-cause mortality: An umbrella review of systematic reviews and meta-analyses. Eur J Prev Cardiol 2025:zwaf118. [PMID: 40037379 DOI: 10.1093/eurjpc/zwaf118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/28/2024] [Accepted: 01/11/2025] [Indexed: 03/06/2025]
Abstract
AIM The aim was to examine the relationship between exposure to environmental metallic and metalloid pollutants and cardiovascular disease (CVD) and all-cause mortality by integrating the information currently available from systematic reviews and meta-analyses. METHOD PubMed, Embase, and Web of Science for systematic reviews and meta-analyses were thoroughly searched up to October 9, 2024. Systematic reviews and meta-analyses of various kinds that evaluated the relationship between exposure to ambient metallic and metalloid pollutants and CVD and all-cause mortality were included. The methodological quality and the evidence quality were assessed using AMSTAR2 and GRADE, respectively. RESULTS We identified 25 meta-analyses and 81 health outcomes-76 unique outcomes from observational studies and 5 unique outcomes from RCTs-from 8,841 independent publications. Exposure to non-essential metallic and metalloid pollutants, including arsenic, lead, and cadmium as well as essential metallic and metalloid contaminants like copper, has been associated with an elevated risk of CVD events and CVD mortality, according to moderate-quality evidence. According to low-quality evidence, exposure to arsenic, lead, and cadmium increases the risk of CHD, while exposure to lead, cadmium, and copper is strongly associated with an increased risk of stroke and all-cause mortality. Further, zinc and selenium may be protective factors for CVD and all-cause mortality. CONCLUSION Despite variations in evidence gradients, environmental metallic and metalloid contaminants like arsenic, lead, cadmium, mercury, and copper are linked to CVD events and mortality, whereas zinc and selenium may offer protection.
Collapse
Affiliation(s)
- Xinxin He
- Department of General Practice, The Affiliated Luohu Hospital Shenzhen University Medical School, Shenzhen, Guangdong, 518001
| | - Haohang Sun
- Cardiovascular Department, Zhengzhou Yihe Hospital Affiliated to Henan University, Zhengzhou, Henan, 450041, People's Republic of China
| | - Yang Zhao
- Department of General Practice, The Affiliated Luohu Hospital Shenzhen University Medical School, Shenzhen, Guangdong, 518001
| | - Xueru Fu
- Department of General Practice, The Affiliated Luohu Hospital Shenzhen University Medical School, Shenzhen, Guangdong, 518001
| | - Mengdi Wang
- Center for Clinical Epidemiology and Evidence-based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
| | - Mengna Liu
- Center for Clinical Epidemiology and Evidence-based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
| | - Yijia Su
- Center for Clinical Epidemiology and Evidence-based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, People's Republic of China
| | - Pei Qin
- Center for Clinical Epidemiology and Evidence-based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, People's Republic of China
| | - Dongsheng Hu
- Department of General Practice, The Affiliated Luohu Hospital Shenzhen University Medical School, Shenzhen, Guangdong, 518001
| |
Collapse
|
2
|
Matsumura S, Yasuda J, Notomi T, Suzuki Y, Chen IS, Murakami D, Hotomi M, Nakamura TY. Direct toxicity of cigarette smoke extract on cardiac function mediated by mitochondrial dysfunction in Sprague-Dawley rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes. PLoS One 2024; 19:e0295737. [PMID: 38165883 PMCID: PMC10760691 DOI: 10.1371/journal.pone.0295737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2023] [Indexed: 01/04/2024] Open
Abstract
Cigarette smoke has been recognized as a major risk factor for cardiovascular disease. However, its direct effects on rodent and human cardiomyocytes and its cellular mechanisms are not fully understood. In this study, we examined the direct effects of cigarette smoke extract (CSE) on contractile functions, intracellular Ca2+ dynamics, and mitochondrial function using cultured or freshly isolated rat ventricular myocytes and human induced pluripotent stem cell (iPS)-derived cardiomyocytes. In rat cardiomyocytes, CSE (≥0.1%) resulted in a time- and concentration-dependent cessation of spontaneous beating of cultured cardiomyocytes, eventually leading to cell death, which indicates direct toxicity. In addition, 1% CSE reduced contractile function of freshly isolated ventricular myocytes. Similar contractile dysfunction (declined spontaneous beating rate and contractility) was also observed in human iPS-derived cardiomyocytes. Regarding intracellular Ca2+ dynamics, 1% CSE increased the Ca2+ transient amplitude by greatly increasing systolic Ca2+ levels and slightly increasing diastolic Ca2+ levels. CSE also accelerated the decay of Ca2+ transients, and triggered spike-shaped Ca2+ transients in some cells. These results indicate that CSE causes abnormal Ca2+ dynamics in cardiomyocytes. Furthermore, CSE induced a cascade of mitochondrial dysfunctions, including increased mitochondrial reactive oxygen species, opening of mitochondrial permeability transition pore, reduction of mitochondrial membrane potential, and release of cytochrome c from mitochondria. These results suggest that CSE-induced contractile dysfunction and myocardial cell death is caused by abnormal Ca2+ dynamics and subsequent mitochondrial dysregulation, which would result in reduced bioenergetics and activation of cell death pathways.
Collapse
Affiliation(s)
- Sakiko Matsumura
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Jumpei Yasuda
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Takuya Notomi
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Yoshihiro Suzuki
- R&D Headquarters Development Department, SIBATA Scientific Technology Ltd, Saitama, Japan
| | - I-Shan Chen
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Daichi Murakami
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| | - Tomoe Y. Nakamura
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama city, Wakayama, Japan
| |
Collapse
|
3
|
Anyaegbunam UA, More P, Fontaine JF, Cate VT, Bauer K, Distler U, Araldi E, Bindila L, Wild P, Andrade-Navarro MA. A Systematic Review of Lipid-Focused Cardiovascular Disease Research: Trends and Opportunities. Curr Issues Mol Biol 2023; 45:9904-9916. [PMID: 38132464 PMCID: PMC10742042 DOI: 10.3390/cimb45120618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/28/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
Lipids are important modifiers of protein function, particularly as parts of lipoproteins, which transport lipophilic substances and mediate cellular uptake of circulating lipids. As such, lipids are of particular interest as blood biological markers for cardiovascular disease (CVD) as well as for conditions linked to CVD such as atherosclerosis, diabetes mellitus, obesity and dietary states. Notably, lipid research is particularly well developed in the context of CVD because of the relevance and multiple causes and risk factors of CVD. The advent of methods for high-throughput screening of biological molecules has recently resulted in the generation of lipidomic profiles that allow monitoring of lipid compositions in biological samples in an untargeted manner. These and other earlier advances in biomedical research have shaped the knowledge we have about lipids in CVD. To evaluate the knowledge acquired on the multiple biological functions of lipids in CVD and the trends in their research, we collected a dataset of references from the PubMed database of biomedical literature focused on plasma lipids and CVD in human and mouse. Using annotations from these records, we were able to categorize significant associations between lipids and particular types of research approaches, distinguish non-biological lipids used as markers, identify differential research between human and mouse models, and detect the increasingly mechanistic nature of the results in this field. Using known associations between lipids and proteins that metabolize or transport them, we constructed a comprehensive lipid-protein network, which we used to highlight proteins strongly connected to lipids found in the CVD-lipid literature. Our approach points to a series of proteins for which lipid-focused research would bring insights into CVD, including Prostaglandin G/H synthase 2 (PTGS2, a.k.a. COX2) and Acylglycerol kinase (AGK). In this review, we summarize our findings, putting them in a historical perspective of the evolution of lipid research in CVD.
Collapse
Affiliation(s)
- Uchenna Alex Anyaegbunam
- Computational Biology and Data Mining Group (CBDM), Institute of Organismic and Molecular Evolution (iOME), Johannes Gutenberg University, 55122 Mainz, Germany
| | - Piyush More
- Computational Biology and Data Mining Group (CBDM), Institute of Organismic and Molecular Evolution (iOME), Johannes Gutenberg University, 55122 Mainz, Germany
- Department of Pharmacology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Jean-Fred Fontaine
- Computational Biology and Data Mining Group (CBDM), Institute of Organismic and Molecular Evolution (iOME), Johannes Gutenberg University, 55122 Mainz, Germany
- Central Institute for Decision Support Systems in Crop Protection (ZEPP), 55545 Bad Kreuznach, Germany
| | - Vincent ten Cate
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH), University Medical Center, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Katrin Bauer
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Computational Systems Medicine, Center for Thrombosis and Hemostasis (CTH), 55131 Mainz, Germany
| | - Ute Distler
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Research Centre for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Elisa Araldi
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Computational Systems Medicine, Center for Thrombosis and Hemostasis (CTH), 55131 Mainz, Germany
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center, 55131 Mainz, Germany
| | - Philipp Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH), University Medical Center, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Miguel A. Andrade-Navarro
- Computational Biology and Data Mining Group (CBDM), Institute of Organismic and Molecular Evolution (iOME), Johannes Gutenberg University, 55122 Mainz, Germany
| |
Collapse
|
4
|
Dorward AM, Stewart AJ, Pitt SJ. The role of Zn2+ in shaping intracellular Ca2+ dynamics in the heart. J Gen Physiol 2023; 155:e202213206. [PMID: 37326614 PMCID: PMC10276528 DOI: 10.1085/jgp.202213206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Increasing evidence suggests that Zn2+ acts as a second messenger capable of transducing extracellular stimuli into intracellular signaling events. The importance of Zn2+ as a signaling molecule in cardiovascular functioning is gaining traction. In the heart, Zn2+ plays important roles in excitation-contraction (EC) coupling, excitation-transcription coupling, and cardiac ventricular morphogenesis. Zn2+ homeostasis in cardiac tissue is tightly regulated through the action of a combination of transporters, buffers, and sensors. Zn2+ mishandling is a common feature of various cardiovascular diseases. However, the precise mechanisms controlling the intracellular distribution of Zn2+ and its variations during normal cardiac function and during pathological conditions are not fully understood. In this review, we consider the major pathways by which the concentration of intracellular Zn2+ is regulated in the heart, the role of Zn2+ in EC coupling, and discuss how Zn2+ dyshomeostasis resulting from altered expression levels and efficacy of Zn2+ regulatory proteins are key drivers in the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Amy M. Dorward
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, UK
| | | |
Collapse
|
5
|
Hara T, Yoshigai E, Ohashi T, Fukada T. Zinc in Cardiovascular Functions and Diseases: Epidemiology and Molecular Mechanisms for Therapeutic Development. Int J Mol Sci 2023; 24:ijms24087152. [PMID: 37108314 PMCID: PMC10139119 DOI: 10.3390/ijms24087152] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/04/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Zinc is an essential trace element that plays an important physiological role in numerous cellular processes. Zinc deficiency can result in diverse symptoms, such as impairment of the immune response, skin disorders, and impairments in cardiovascular functions. Recent reports have demonstrated that zinc acts as a signaling molecule, and its signaling pathways, referred to as zinc signals, are related to the molecular mechanisms of cardiovascular functions. Therefore, comprehensive understanding of the significance of zinc-mediated signaling pathways is vital as a function of zinc as a nutritional component and of its molecular mechanisms and targets. Several basic and clinical studies have reported the relationship between zinc level and the onset and pathology of cardiovascular diseases, which has attracted much attention in recent years. In this review, we summarize the recent findings regarding the effects of zinc on cardiovascular function. We also discuss the importance of maintaining zinc homeostasis in the cardiovascular system and its therapeutic potential as a novel drug target.
Collapse
Affiliation(s)
- Takafumi Hara
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Emi Yoshigai
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Takuto Ohashi
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| |
Collapse
|
6
|
Ohlrogge AH, Frost L, Schnabel RB. Harmful Impact of Tobacco Smoking and Alcohol Consumption on the Atrial Myocardium. Cells 2022; 11:2576. [PMID: 36010652 PMCID: PMC9406618 DOI: 10.3390/cells11162576] [Citation(s) in RCA: 7] [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: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Tobacco smoking and alcohol consumption are widespread exposures that are legal and socially accepted in many societies. Both have been widely recognized as important risk factors for diseases in all vital organ systems including cardiovascular diseases, and with clinical manifestations that are associated with atrial dysfunction, so-called atrial cardiomyopathy, especially atrial fibrillation and stroke. The pathogenesis of atrial cardiomyopathy, atrial fibrillation, and stroke in context with smoking and alcohol consumption is complex and multifactorial, involving pathophysiological mechanisms, environmental, and societal aspects. This narrative review summarizes the current literature regarding alterations in the atrial myocardium that is associated with smoking and alcohol.
Collapse
Affiliation(s)
- Amelie H. Ohlrogge
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Lars Frost
- Diagnostic Centre, University Clinic for Development of Innovative Patient Pathways, Silkeborg Regional Hospital, 8600 Silkeborg, Denmark
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Renate B. Schnabel
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
7
|
Imraish A, Abu Thiab T, Al-Awaida W, Al-Ameer HJ, Bustanji Y, Hammad H, Alsharif M, Al-Hunaiti A. In vitro anti-inflammatory and antioxidant activities of ZnFe 2 O 4 and CrFe 2 O 4 nanoparticles synthesized using Boswellia carteri resin. J Food Biochem 2021; 45:e13730. [PMID: 33880765 DOI: 10.1111/jfbc.13730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/25/2021] [Accepted: 03/29/2021] [Indexed: 11/29/2022]
Abstract
The development of plant-based nano-materials is considered an eco-friendly technology because it does not involve hazardous chemicals. In this study, bimetallic ZnFe2 O4 and CrFe2 O4 nanoparticles were synthesized using an aqueous extract of Boswellia carteri resin. Synthesized ZnFe2 O4 and CrFe2 O4 nanoparticles were characterized by UV-Vis spectroscopy, FTIR, XRD, and HR-TEM. The anti-inflammatory activity was investigated in LPS-stimulated RAW 264.7 macrophages, whereas antioxidant activity was examined using a Hydrogen Peroxide Scavenging Activity Assay, Nitric Oxide Scavenging Activity Assay, and ABTS Radical Scavenging Assay. ZnFe2 O4 and CrFe2 O4 nanoparticles demonstrated a moderate scavenger of H2 O2 with IC50 values; 87.528 ± 8 μg/ml and 146.4468 ± 12 μg/ml, respectively. While they exhibited a strong scavenger of NO with IC50 values; 4.01 ± 0.7 μg/ml and 4.01 ± 0.7μg/ml, respectively. Interestingly, ZnFe2 O4 and CrFe2 O4 nanoparticles revealed an excellent anti-inflammatory activity by dose-dependently suppressing mRNA expressions of IL-1b, IL-6, and TNF-α. Also, ZnFe2 O4 and CrFe2 O4 nanoparticles suppress the protein expression of TNF-α. Together, our results proved that phyto-mediated ZnFe2 O4 and CrFe2 O4 nanoparticles using Boswellia carteri resin have great potential in biomedical applications such as anti-inflammatory and antioxidant. PRACTICAL APPLICATIONS: Our phyto-synthesized chromium iron oxide bimetallic nanoparticles (NPs) have shown a novel and potent anti-inflammatory activity, with remarkable biosafety toward tested macrophages. Zinc iron oxide bimetallic NPs exhibited anti-inflammatory effect with a lesser extent compared to the former, with moderate cytotoxicity against tested macrophages. Both zinc and chromium iron oxide NPs exhibited an equivalent antioxidant activity. Our resin-capped chromium iron oxide NPs are suggested to be a competing nonsteroidal anti-inflammatory agent; it is further recommended to establish advanced animal studies to confirm their biosafety, stability, and anti-inflammatory activity accompanied with the antioxidant activity.
Collapse
Affiliation(s)
- Amer Imraish
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Tuqa Abu Thiab
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Wajdy Al-Awaida
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Hamzeh J Al-Ameer
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Yasser Bustanji
- School of Pharmacy, The University of Jordan, Amman, Jordan.,Hamdi Mango Centre for Scientific Research, The University of Jordan, Amman, Jordan
| | - Hana Hammad
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Mays Alsharif
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Afnan Al-Hunaiti
- Department of Chemistry, School of Science, The University of Jordan, Amman, Jordan
| |
Collapse
|
8
|
Beclin1 Haploinsufficiency accentuates second-hand smoke exposure -induced myocardial Remodeling and contractile dysfunction through a STING-mediated mechanism. J Mol Cell Cardiol 2020; 148:78-88. [PMID: 32891637 DOI: 10.1016/j.yjmcc.2020.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
Second-hand smoking evokes inflammation and cardiovascular diseases. Recent evidence has revealed a pivotal role for deranged autophagy in smoke exposure-induced cardiac anomalies. This study evaluated the impact of haploinsufficiency of the mTOR-independent autophagy protein Beclin1 on side-stream smoke exposure-induced cardiac anomalies and mechanism(s) involved. Adult WT and Beclin1 haploinsufficiency (Becn+/-) mice were exposed to cigarette smoke for 1 h daily for 90 days. Echocardiographic, cardiomyocyte function, intracellular Ca2+, autophagy, mitophagy, apoptosis and inflammation were examined. DHE staining was employed to evaluate O2- level. Our data revealed that Beclin1 deficiency exacerbated smoke exposure-induced myocardial anomalies in geometry, fractional shortening, cardiomyocyte function, intracellular Ca2+ handling, TEM ultrastructure, and inflammation along with pronounced apoptosis and O2- production. Side-stream smoke provoked excessive autophagy/mitophagy, mtDNA release, and activation of innate immune response signals cyclic GMP-AMP synthase (cGAS) and its effector - stimulator of interferon genes (STING), the effect was abolished or unaffected by Becn haploinsufficiency. STING phosphorylation was overtly promoted by smoke exposure in Becn+/- mice. Smoke exposure also suppressed phosphorylation of mTOR although it facilitated that of ULK1 in both groups. In vitro data revealed that inhibition of cGAS or STING failed to affect smoke extract-induced mitophagy although they abrogated smoke extract-induced cardiomyocyte dysfunction except cGAS inhibition in Becn+/- mice. These data suggest that Beclin1 is integral in the maintenance of cardiac homeostasis under side-stream smoke exposure via a STING-mediated mechanism.
Collapse
|
9
|
Wang S, Chen X, Zeng B, Xu X, Chen H, Zhao P, Hilaire ML, Bucala R, Zheng Q, Ren J. Knockout of macrophage migration inhibitory factor accentuates side-stream smoke exposure-induced myocardial contractile dysfunction through dysregulated mitophagy. Pharmacol Res 2020; 157:104828. [PMID: 32339783 DOI: 10.1016/j.phrs.2020.104828] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/23/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
Second hand smoke exposure increases the prevalence of chronic diseases partly attributed to inflammatory responses. Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine, is involved in the pathogenesis of multiple diseases although its role in second hand smoke exposure-induced cardiac anomalies remains elusive. This study evaluated the impact of MIF knockout on side-stream smoke exposure-induced cardiac pathology and underlying mechanisms. Adult WT and MIF knockout (MIFKO) mice were placed in a chamber exposed to cigarette smoke for 1 h daily for 60 consecutive days. Echocardiographic, cardiomyocyte function and intracellular Ca2+ handling were evaluated. Autophagy, mitophagy and apoptosis were examined using western blot. DHE staining was used to evaluate superoxide anion (O2-) generation. Masson trichrome staining was employed to assess interstitial fibrosis. Our data revealed that MIF knockout accentuated side-stream smoke-induced cardiac anomalies in fractional shortening, cardiomyocyte function, intracellular Ca2+ homeostasis, myocardial ultrastructure and mitochondrial content along with overt apoptosis and O2- generation. In addition, unfavorable effects of side-stream smoke were accompanied by excessive formation of autophagolysosome and elevated TFEB, the effect of which was exacerbated by MIF knockout. Recombinant MIF rescued smoke extract-induced myopathic anomalies through promoting AMPK activation, mitophagy and lysosomal function. Taken together, our data suggest that MIF serves as a protective factor against side-stream smoke exposure-induced myopathic changes through facilitating mitophagy and autophagolysosome formation.
Collapse
Affiliation(s)
- Shuyi Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China; School of Pharmacy, University of Wyoming College of Health Sciences, Laramie 82071, WY USA.
| | - Xu Chen
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Biru Zeng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Xihui Xu
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie 82071, WY USA
| | - Huaguo Chen
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Ping Zhao
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Michelle L Hilaire
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie 82071, WY USA
| | - Richard Bucala
- Department of Medicine, Yale School of Medicine, New Haven 06520, CT USA
| | - Qijun Zheng
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie 82071, WY USA; Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen 518020, China.
| | - Jun Ren
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie 82071, WY USA; Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
Making a case for metallothioneins conferring cardioprotection in pulmonary hypertension. Med Hypotheses 2020; 137:109572. [DOI: 10.1016/j.mehy.2020.109572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/30/2019] [Accepted: 01/15/2020] [Indexed: 11/23/2022]
|
11
|
DNA-PKcs promotes cardiac ischemia reperfusion injury through mitigating BI-1-governed mitochondrial homeostasis. Basic Res Cardiol 2020; 115:11. [PMID: 31919590 DOI: 10.1007/s00395-019-0773-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/27/2019] [Indexed: 01/24/2023]
Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a novel inducer to promote mitochondrial apoptosis and suppress tumor growth in a variety of cells although its role in cardiovascular diseases remains obscure. This study was designed to examine the role of DNA-PKcs in cardiac ischemia reperfusion (IR) injury and mitochondrial damage. Cardiomyocyte-specific DNA-PKcs knockout (DNA-PKcsCKO) mice were subjected to IR prior to assessment of myocardial function and mitochondrial apoptosis. Our data revealed that IR challenge, hypoxia-reoxygenation (HR) or H2O2-activated DNA-PKcs through post-transcriptional phosphorylation in murine hearts or cardiomyocytes. Mice deficient in DNA-PKcs in cardiomyocytes were protected against cardiomyocyte death, infarct area expansion and cardiac dysfunction. DNA-PKcs ablation countered IR- or HR-induced oxidative stress, mPTP opening, mitochondrial fission, mitophagy failure and Bax-mediated mitochondrial apoptosis, possibly through suppression of Bax inhibitor-1 (BI-1) activity. A direct association between DNA-PKcs and BI-1 was noted where DNA-PKcs had little effect on BI-1 transcription but interacted with BI-1 to promote its degradation. Loss of DNA-PKcs stabilized BI-1, thus offering resistance of mitochondria and cardiomyocytes against IR insult. Moreover, DNA-PKcs ablation-induced beneficial cardioprotection against IR injury was mitigated by concurrent knockout of BI-1. Double deletion of DNA-PKcs and BI-1 failed to exert protection against global IR injury and mitochondrial damage, confirming a permissive role of BI-1 in DNA-PKcs deletion-elicited cardioprotection against IR injury. DNA-PKcs serves as a novel causative factor for mitochondrial damage via suppression of BI-1, en route to the onset and development of cardiac IR injury.
Collapse
|
12
|
Szostak J, Titz B, Schlage WK, Guedj E, Sewer A, Phillips B, Leroy P, Buettner A, Neau L, Trivedi K, Martin F, Ivanov NV, Vanscheeuwijck P, Peitsch MC, Hoeng J. Structural, functional, and molecular impact on the cardiovascular system in ApoE -/- mice exposed to aerosol from candidate modified risk tobacco products, Carbon Heated Tobacco Product 1.2 and Tobacco Heating System 2.2, compared with cigarette smoke. Chem Biol Interact 2020; 315:108887. [PMID: 31705857 DOI: 10.1016/j.cbi.2019.108887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/26/2022]
Abstract
AIM To investigate the molecular, structural, and functional impact of aerosols from candidate modified risk tobacco products (cMRTP), the Carbon Heated Tobacco Product (CHTP) 1.2 and Tobacco Heating System (THS) 2.2, compared with that of mainstream cigarette smoke (CS) on the cardiovascular system of ApoE-/- mice. METHODS Female ApoE-/- mice were exposed to aerosols from THS 2.2 and CHTP 1.2 or to CS from the 3R4F reference cigarette for up to 6 months at matching nicotine concentrations. A Cessation and a Switching group (3 months exposure to 3R4F CS followed by filtered air or CHTP 1.2 for 3 months) were included. Cardiovascular effects were investigated by echocardiographic, histopathological, immunohistochemical, and transcriptomics analyses. RESULTS Continuous exposure to cMRTP aerosols did not affect atherosclerosis progression, heart function, left ventricular (LV) structure, or the cardiovascular transcriptome. Exposure to 3R4F CS triggered atherosclerosis progression, reduced systolic ejection fraction and fractional shortening, caused heart LV hypertrophy, and initiated significant dysregulation in the transcriptomes of the heart ventricle and thoracic aorta. Importantly, the structural, functional, and molecular changes caused by 3R4F CS were improved in the smoking cessation and switching groups. CONCLUSION Exposure to cMRTP aerosols lacked most of the CS exposure-related functional, structural, and molecular effects. Smoking cessation or switching to CHTP 1.2 aerosol caused similar recovery from the 3R4F CS effects in the ApoE-/- model, with no further acceleration of plaque progression beyond the aging-related rate.
Collapse
Affiliation(s)
- Justyna Szostak
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany.
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Laurent Neau
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| |
Collapse
|
13
|
Testicular Toxicity of Water Pipe Smoke Exposure in Mice and the Effect of Treatment with Nootkatone Thereon. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2416935. [PMID: 31341528 PMCID: PMC6614988 DOI: 10.1155/2019/2416935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/02/2019] [Indexed: 12/17/2022]
Abstract
There is a worldwide increase in the popularity of water pipe (shisha) tobacco smoking including in Europe and North America. However, little is known about the effects of water pipe smoke (WPS) exposure on male reproductivity. We have recently demonstrated that WPS exposure in mice induces testicular toxicity including inflammation and oxidative stress. Nootkatone, a sesquiterpenoid found in grapefruit, has antioxidant and anti-inflammatory effects. However, the possible protective effect of nootkatone on WPS-induced testicular toxicity has not been reported before. Here, we tested the effects of treatment of mice with nootkatone on WPS-induced testicular toxicity. Mice were exposed to normal air or WPS (30 minutes/day, for 30 days). Nootkatone (90 mg/kg) was given orally to mice by gavage, 1 h before WPS or air exposure. Nootkatone treatment significantly ameliorated the WPS-induced increase in plasma levels of inhibin, uric acid, and lactate dehydrogenase activity. Nootkatone also significantly mitigated the decrease in testosterone, androgen-binding protein, and estradiol concentrations in the plasma induced by WPS. In testicular homogenates, WPS exposure caused a decrease in the total nitric oxide level and an increase in the proinflammatory cytokine interleukin-1β level and oxidative stress markers including malondialdehyde, cytochrome C, and 8-Oxo-2'-deoxyguanosine. All the latter effects were significantly alleviated by nootkatone treatment. Moreover, in testicular homogenate, nootkatone inhibited the expression of nuclear factor-kappaB induced by WPS. Likewise, histological examination of mouse testes showed that nootkatone treatment ameliorated the deterioration of spermatogenesis induced by WPS exposure. We conclude that nootkatone ameliorated the WPS-induced testicular inflammation and oxidative stress and hormonal and spermatogenesis alterations.
Collapse
|
14
|
Yang L, Ma J, Tan Y, Zheng Q, Dong M, Guo W, Xiong L, Yang J, Ren J. Cardiac-specific overexpression of metallothionein attenuates L-NAME-induced myocardial contractile anomalies and apoptosis. J Cell Mol Med 2019; 23:4640-4652. [PMID: 31104354 PMCID: PMC6584723 DOI: 10.1111/jcmm.14375] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/14/2019] [Accepted: 04/09/2019] [Indexed: 01/21/2023] Open
Abstract
Hypertension contributes to the high cardiac morbidity and mortality. Although oxidative stress plays an essential role in hypertensive heart diseases, the mechanism remains elusive. Transgenic mice with cardiac overexpression of metallothionein, a heavy metal‐binding scavenger, were challenged with NG‐nitro‐L‐arginine methyl ester (L‐NAME) for 14 days prior to measurement of myocardial contractile and intracellular Ca2+ anomalies as well as cell signalling mechanisms using Western blot and immunofluorescence analysis. L‐NAME challenge elicited hypertension, macrophage infiltration, oxidative stress, inflammation and cardiac dysfunction manifested as increased proinflammatory macrophage marker F4/80, interleukin‐1β (IL‐1β), intracellular O2- production, LV end systolic and diastolic diameters as well as depressed fractional shortening. L‐NAME treatment reduced mitochondrial membrane potential (MMP), impaired cardiomyocyte contractile and intracellular Ca2+ properties as evidenced by suppressed peak shortening, maximal velocity of shortening/relengthening, rise in intracellular Ca2+, along with elevated baseline and peak intracellular Ca2+. These unfavourable mechanical changes and decreased MMP (except blood pressure and macrophage infiltration) were alleviated by overexpression of metallothionein. Furthermore, the apoptosis markers including BAD, Bax, Caspase 9, Caspase 12 and cleaved Caspase 3 were up‐regulated while the anti‐apoptotic marker Bcl‐2 was decreased by L‐NAME treatment. Metallothionein transgene reversed L‐NAME‐induced changes in Bax, Bcl‐2, BAD phosphorylation, Caspase 9, Caspase 12 and cleaved Caspase 3. Our results suggest that metallothionein protects against L‐NAME‐induced myocardial contractile anomalies in part through inhibition of apoptosis.
Collapse
Affiliation(s)
- Lifang Yang
- Department of Anesthesiology, Xi'an Children Hospital, Xi'an, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming
| | - Jipeng Ma
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming.,Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Tan
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming.,Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Maolong Dong
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming.,Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Guo
- Department of Animal Sciences, University of Wyoming, Laramie, Wyoming
| | - Lize Xiong
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming
| |
Collapse
|
15
|
Al-Awaida WJ, Zihlif MA, Al-Ameer HJ, Sharab A, Akash M, Aburubaiha ZA, Fattash IA, Imraish A, Ali KH. The effect of green tea consumption on the expression of antioxidant- and inflammation-related genes induced by nicotine. J Food Biochem 2019; 43:e12874. [PMID: 31353688 DOI: 10.1111/jfbc.12874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/22/2022]
Abstract
The aim of this study is to investigate the protective effect of green tea (GT) against the toxicity of nicotine. BALB/c mice were divided into four groups. Group I received food and water intake ad libidium, Group II received GT solution at a dose of 1 ml/kg body weight orally twice a day via gastric gavage, Group III was injected intraperitoneally with nicotine (2.5 mg/kg) once per day for 4 weeks, and Group IV received both nicotine and GT; GT was introduced using gastric gavage 1 hr before and 1 hr after the nicotine injection. The administration of nicotine altered the cellular antioxidant defense system by inducing inflammation and damage in the tissues of liver, lungs, and kidneys. In addition, nicotine treatment significantly enhanced the expression antioxidant- and inflammation-related genes. There were significant improvements when the nicotine-exposed mice treated with GT. PRACTICAL APPLICATIONS: In this study, it is revealed that the administration of nicotine altered the cellular antioxidant defense system by inducing inflammation manifested by the infiltration of inflammatory cells and damage seen in liver, lungs, and kidneys. GT contributed to the reduction of toxicity of nicotine, probably mediated by free radicals, through downregulation of nicotine-induced upregulated antioxidant- and inflammation-related genes. Never the less, further in depth investigation on characterization of the active constituents of GT responsible for their effect seen here and the mechanism that contributes to the effects seen in this reports is highly demanded. Furthermore, GT extract could be considered as a dietary supplement for the reduction of nicotine toxicity among cigarette smoker.
Collapse
Affiliation(s)
- Wajdy J Al-Awaida
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Malek A Zihlif
- Faculty of Medicine, Department of Pharmacology, The University of Jordan, Amman, Jordan
| | - Hamzeh J Al-Ameer
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Ahmad Sharab
- Faculty of Medicine, Department of Pharmacology, The University of Jordan, Amman, Jordan
| | - Muhanad Akash
- Faculty of Agriculture, Department of Horticulture and Crop Science, The University of Jordan, Amman, Jordan
| | - Zaid A Aburubaiha
- Faculty of Health Sciences, Department of Medical Laboratories, American University of Madaba, Madaba, Jordan
| | - Isam A Fattash
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Amer Imraish
- Faculty of Science, Department of Biology, The University of Jordan, Amman, Jordan
| | - Khedhir H Ali
- Faculty of Health Sciences, Department of Medical Laboratories, American University of Madaba, Madaba, Jordan
| |
Collapse
|
16
|
Guo H, Tian L, Zhang JZ, Kitani T, Paik DT, Lee WH, Wu JC. Single-Cell RNA Sequencing of Human Embryonic Stem Cell Differentiation Delineates Adverse Effects of Nicotine on Embryonic Development. Stem Cell Reports 2019; 12:772-786. [PMID: 30827876 PMCID: PMC6449785 DOI: 10.1016/j.stemcr.2019.01.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 12/22/2022] Open
Abstract
Nicotine, the main chemical constituent of tobacco, is highly detrimental to the developing fetus by increasing the risk of gestational complications and organ disorders. The effects of nicotine on human embryonic development and related mechanisms, however, remain poorly understood. Here, we performed single-cell RNA sequencing (scRNA-seq) of human embryonic stem cell (hESC)-derived embryoid body (EB) in the presence or absence of nicotine. Nicotine-induced lineage-specific responses and dysregulated cell-to-cell communication in EBs, shedding light on the adverse effects of nicotine on human embryonic development. In addition, nicotine reduced cell viability, increased reactive oxygen species (ROS), and altered cell cycling in EBs. Abnormal Ca2+ signaling was found in muscle cells upon nicotine exposure, as verified in hESC-derived cardiomyocytes. Consequently, our scRNA-seq data suggest direct adverse effects of nicotine on hESC differentiation at the single-cell level and offer a new method for evaluating drug and environmental toxicity on human embryonic development in utero.
Collapse
Affiliation(s)
- Hongchao Guo
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lei Tian
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joe Z Zhang
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David T Paik
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
17
|
Zhang Y, Wang Y, Xu J, Tian F, Hu S, Chen Y, Fu Z. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res 2019; 66:e12542. [PMID: 30516280 DOI: 10.1111/jpi.12542] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/24/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022]
Abstract
Optic atrophy 1 (OPA1)-related mitochondrial fusion and mitophagy are vital to sustain mitochondrial homeostasis under stress conditions. However, no study has confirmed whether OPA1-related mitochondrial fusion/mitophagy is activated by melatonin and, consequently, attenuates cardiomyocyte death and mitochondrial stress in the setting of cardiac ischemia-reperfusion (I/R) injury. Our results indicated that OPA1, mitochondrial fusion, and mitophagy were significantly repressed by I/R injury, accompanied by infarction area expansion, heart dysfunction, myocardial inflammation, and cardiomyocyte oxidative stress. However, melatonin treatment maintained myocardial function and cardiomyocyte viability, and these effects were highly dependent on OPA1-related mitochondrial fusion/mitophagy. At the molecular level, OPA1-related mitochondrial fusion/mitophagy, which was normalized by melatonin, substantially rectified the excessive mitochondrial fission, promoted mitochondria energy metabolism, sustained mitochondrial function, and blocked cardiomyocyte caspase-9-involved mitochondrial apoptosis. However, genetic approaches with a cardiac-specific knockout of OPA1 abolished the beneficial effects of melatonin on cardiomyocyte survival and mitochondrial homeostasis in vivo and in vitro. Furthermore, we demonstrated that melatonin affected OPA1 stabilization via the AMPK signaling pathway and that blockade of AMPK repressed OPA1 expression and compromised the cardioprotective action of melatonin. Overall, our results confirm that OPA1-related mitochondrial fusion/mitophagy is actually modulated by melatonin in the setting of cardiac I/R injury. Moreover, manipulation of the AMPK-OPA1-mitochondrial fusion/mitophagy axis via melatonin may be a novel therapeutic approach to reduce cardiac I/R injury.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junnan Xu
- Beijing Key Laboratory of Immunology Regulatory and Organ Transplantation, Organ Transplant Institute of People's Liberation Army, The 309th Hospital of People's Liberation Army, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Zhenhong Fu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
18
|
Zhou H, Zhu P, Wang J, Zhu H, Ren J, Chen Y. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2α-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ 2018. [PMID: 29540794 PMCID: PMC5988750 DOI: 10.1038/s41418-018-0086-7] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Disturbed mitochondrial homeostasis contributes to the pathogenesis of cardiac ischemia reperfusion (IR) injury, although the underlying mechanism remains elusive. Here, we demonstrated that casein kinase 2α (CK2α) was upregulated following acute cardiac IR injury. Increased CK2α was shown to be instrumental to mitochondrial damage, cardiomyocyte death, infarction area expansion and cardiac dysfunction, whereas cardiac-specific CK2α knockout (CK2αCKO) mice were protected against IR injury and mitochondrial damage. Functional assay indicated that CK2α enhanced the phosphorylation (inactivation) of FUN14 domain containing 1 (FUNDC1) via post-transcriptional modification at Ser13, thus effectively inhibiting mitophagy. Defective mitophagy failed to remove damaged mitochondria induced by IR injury, resulting in mitochondrial genome collapse, electron transport chain complex (ETC) inhibition, mitochondrial biogenesis arrest, cardiolipin oxidation, oxidative stress, mPTP opening, mitochondrial debris accumulation and eventually mitochondrial apoptosis. In contrast, loss of CK2α reversed the FUNDC1-mediated mitophagy, providing a survival advantage to myocardial tissue following IR stress. Interestingly, mice deficient in both CK2α and FUNDC1 failed to show protection against IR injury and mitochondrial damage through a mechanism possible attributed to lack of mitophagy. Taken together, our results confirmed that CK2α serves as a negative regulator of mitochondrial homeostasis via suppression of FUNDC1-required mitophagy, favoring the development of cardiac IR injury.
Collapse
Affiliation(s)
- Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China. .,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
| | - Pingjun Zhu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Hong Zhu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
| | - Yundai Chen
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
19
|
Hu N, Ren J, Zhang Y. Mitochondrial aldehyde dehydrogenase obliterates insulin resistance-induced cardiac dysfunction through deacetylation of PGC-1α. Oncotarget 2018; 7:76398-76414. [PMID: 27634872 PMCID: PMC5363518 DOI: 10.18632/oncotarget.11977] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022] Open
Abstract
Insulin resistance contributes to the high prevalence of type 2 diabetes mellitus, leading to cardiac anomalies. Emerging evidence depicts a pivotal role for mitochondrial injury in oxidative metabolism and insulin resistance. Mitochondrial aldehyde dehydrogenase (ALDH2) is one of metabolic enzymes detoxifying aldehydes although its role in insulin resistance remains elusive. This study was designed to evaluate the impact of ALDH2 overexpression on insulin resistance-induced myocardial damage and mechanisms involved with a focus on autophagy. Wild-type (WT) and transgenic mice overexpressing ALDH2 were fed sucrose or starch diet for 8 weeks and cardiac function and intracellular Ca2+ handling were assessed using echocardiographic and IonOptix systems. Western blot analysis was used to evaluate Akt, heme oxygenase-1 (HO-1), PGC-1α and Sirt-3. Our data revealed that sucrose intake provoked insulin resistance and compromised fractional shortening, cardiomyocyte function and intracellular Ca2+ handling (p < 0.05) along with unaltered cardiomyocyte size (p > 0.05), mitochondrial injury (elevated ROS generation, suppressed NAD+ and aconitase activity, p < 0.05 for all), the effect of which was ablated by ALDH2. In vitro incubation of the ALDH2 activator Alda-1, the Sirt3 activator oroxylin A and the histone acetyltransferase inhibitor CPTH2 rescued insulin resistance-induced changes in aconitase activity and cardiomyocyte function (p < 0.05). Inhibiting Sirt3 deacetylase using 5-amino-2-(4-aminophenyl) benzoxazole negated Alda-1-induced cardioprotective effects. Taken together, our data suggest that ALDH2 serves as an indispensable cardioprotective factor against insulin resistance-induced cardiomyopathy with a mechanism possibly associated with facilitation of the Sirt3-dependent PGC-1α deacetylation.
Collapse
Affiliation(s)
- Nan Hu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| |
Collapse
|
20
|
Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J, Zhou H. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol 2017; 14:576-587. [PMID: 29149759 PMCID: PMC5691221 DOI: 10.1016/j.redox.2017.11.004] [Citation(s) in RCA: 372] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/31/2017] [Accepted: 11/04/2017] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial fission and selective mitochondrial autophagy (mitophagy) form an essential axis of mitochondrial quality control that plays a critical role in the development of cardiac ischemia-reperfusion (IR) injury. However, the precise upstream molecular mechanism of fission/mitophagy remains unclear. Dual-specificity protein phosphatase1 (DUSP1) regulates cardiac metabolism, but its physiological contribution in the reperfused heart, particularly its influence on mitochondrial homeostasis, is unknown. Here, we demonstrated that cardiac DUSP1 was downregulated following acute cardiac IR injury. In vivo, compared to wild-type mice, DUSP1 transgenic mice (DUSP1TG mice) demonstrated a smaller infarcted area and the improved myocardial function. In vitro, the IR-induced DUSP1 deficiency promoted the activation of JNK which upregulated the expression of the mitochondrial fission factor (Mff). A higher expression level of Mff was associated with elevated mitochondrial fission and mitochondrial apoptosis. Additionally, the loss of DUSP1 also amplified the Bnip3 phosphorylated activation via JNK, leading to the activation of mitophagy. Increased mitophagy overtly consumed mitochondrial mass resulting into the mitochondrial metabolism disorder. However, the reintroduction of DUSP1 blunted Mff/Bnip3 activation and therefore alleviated the fatal mitochondrial fission/mitophagy by inactivating the JNK pathway, providing a survival advantage to myocardial tissue following IR stress. The results of our study suggest that DUSP1 and its downstream JNK pathway are therapeutic targets for conferring protection against IR injury by repressing Mff-mediated mitochondrial fission and Bnip3-required mitophagy.
Collapse
Affiliation(s)
- Qinhua Jin
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Ruibing Li
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China; Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Nan Hu
- Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA
| | - Ting Xin
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China; Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Pingjun Zhu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Shunying Hu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Sai Ma
- Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA
| | - Hong Zhu
- Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA
| | - Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China; Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA.
| |
Collapse
|
21
|
Szostak J, Boué S, Talikka M, Guedj E, Martin F, Phillips B, Ivanov NV, Peitsch MC, Hoeng J. Aerosol from Tobacco Heating System 2.2 has reduced impact on mouse heart gene expression compared with cigarette smoke. Food Chem Toxicol 2017; 101:157-167. [PMID: 28111298 DOI: 10.1016/j.fct.2017.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 02/05/2023]
Abstract
Experimental studies clearly demonstrate a causal effect of cigarette smoking on cardiovascular disease. To reduce the individual risk and population harm caused by smoking, alternative products to cigarettes are being developed. We recently reported on an apolipoprotein E-deficient (Apoe-/-) mouse inhalation study that compared the effects of exposure to aerosol from a candidate modified risk tobacco product, Tobacco Heating System 2.2 (THS2.2), and smoke from the reference cigarette (3R4F) on pulmonary and vascular biology. Here, we applied a transcriptomics approach to evaluate the impact of the exposure to 3R4F smoke and THS2.2 aerosol on heart tissues from the same cohort of mice. The systems response profiles demonstrated that 3R4F smoke exposure led to time-dependent transcriptomics changes (False Discovery Rate (FDR) < 0.05; 44 differentially expressed genes at 3-months; 491 at 8-months). Analysis of differentially expressed genes in the heart tissue indicated that 3R4F exposure induced the downregulation of genes involved in cytoskeleton organization and the contractile function of the heart, notably genes that encode beta actin (Actb), actinin alpha 4 (Actn4), and filamin C (Flnc). This was accompanied by the downregulation of genes related to the inflammatory response. None of these effects were observed in the group exposed to THS2.2 aerosol.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Stéphanie Boué
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Emmanuel Guedj
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Florian Martin
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Blaine Phillips
- Philip Morris International Research Laboratories Pte Ltd, Science Park II, Singapore.
| | - Nikolai V Ivanov
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland.
| |
Collapse
|
22
|
Nemmar A, Al-Salam S, Yuvaraju P, Beegam S, Yasin J, Ali BH. Chronic exposure to water-pipe smoke induces cardiovascular dysfunction in mice. Am J Physiol Heart Circ Physiol 2017; 312:H329-H339. [PMID: 27940964 DOI: 10.1152/ajpheart.00450.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/29/2016] [Accepted: 12/04/2016] [Indexed: 12/21/2022]
Abstract
Water-pipe tobacco smoking is becoming prevalent in all over the world including Western countries. There are limited data on the cardiovascular effects of water-pipe smoke (WPS), in particular following chronic exposure. Here, we assessed the chronic cardiovascular effects of nose-only WPS exposure in C57BL/6 mice. The duration of the session was 30 minutes/day, 5 days/week for 6 consecutive months. Control mice were exposed to air. WPS significantly increased systolic blood pressure. The relative heart weight and plasma concentrations of troponin-I and B-type natriuretic peptide were increased in mice exposed to WPS. Arterial blood gas analysis showed that WPS caused a significant decrease in [Formula: see text] and an increase in [Formula: see text] WPS significantly shortened the thrombotic occlusion time in pial arterioles and venules and increased the number of circulating platelet. Cardiac lipid peroxidation, measured as thiobarbituric acid-reactive substances, was significantly increased, while superoxide dismutase activity, total nitric oxide activity, and glutathione concentration were reduced by WPS exposure. Likewise, immunohistochemical analysis of the heart revealed an increase in the expression of inducible nitric oxide synthase and cytochrome c by cardiomyocytes of WPS-exposed mice. Moreover, hearts of WPS-exposed mice showed the presence of focal interstitial fibrosis. WPS exposure significantly increased heart DNA damage assessed by Comet assay. We conclude that chronic nose-only exposure to WPS impairs cardiovascular homeostasis. Our findings provide evidence that long-term exposure to WPS is harmful to the cardiovascular system and supports interventions to control the spread of WPS, particularly amid youths.NEW & NOTEWORTHY No data are available on the chronic cardiovascular effects of water-pipe smoke (WPS). Our findings provide experimental evidence that chronic exposure to WPS increased blood pressure, relative heart weight, troponin I, and B-type natriuretic peptide in plasma and induced hypoxemia, hypercapnia, and thrombosis. Moreover, WPS caused cardiac oxidative stress, DNA damage, and fibrosis.
Collapse
Affiliation(s)
- Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates;
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Priya Yuvaraju
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Javed Yasin
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates; and
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khod, Sultanate of Oman
| |
Collapse
|
23
|
Yang L, Wang J, Yang J, Schamber R, Hu N, Nair S, Xiong L, Ren J. Antioxidant metallothionein alleviates endoplasmic reticulum stress-induced myocardial apoptosis and contractile dysfunction. Free Radic Res 2016; 49:1187-98. [PMID: 25968954 DOI: 10.3109/10715762.2015.1013952] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AIMS Endoplasmic reticulum (ER) stress exerts myocardial oxidative stress, apoptosis, and contractile anomalies, although the precise interplay between ER stress and apoptosis remains elusive. This study was designed to examine the impact of the cysteine-rich free radical scavenger metallothionein on ER stress-induced myocardial contractile defect and underlying mechanisms. METHODS AND RESULTS Wild-type friendly virus B and transgenic mice with cardiac-specific overexpression of metallothionein were challenged with the ER stress inducer tunicamycin (1 mg/kg, intraperitoneal, 48 h) prior to the assessment of myocardial function, oxidative stress, and apoptosis. Our results revealed that tunicamycin promoted cardiac remodeling (enlarged left ventricular end systolic/diastolic diameters with little changes in left ventricular wall thickness), suppressed fractional shortening and cardiomyocyte contractile function, elevated resting Ca(2+), decreased stimulated Ca(2+) release, prolonged intracellular Ca(2+) clearance, and downregulated sarco(endo)plasmic reticulum Ca(2+)-ATPase levels, the effects of which were negated by metallothionein. Treatment with tunicamycin caused cardiomyocyte mitochondrial injury, as evidenced by decreased mitochondrial membrane potential (∆Ѱm, assessed by JC-1 staining), the effect of which was negated by the antioxidant. Moreover, tunicamycin challenge dramatically facilitated myocardial apoptosis as manifested by increased Bax, caspase 9, and caspase 12 protein levels, as well as elevated caspase 3 activity. Interestingly, metallothionein transgene significantly alleviated tunicamycin-induced myocardial apoptosis. CONCLUSION Taken together, our data favor a beneficial effect of metallothionein against ER stress-induced cardiac dysfunction possibly associated with attenuation of myocardial apoptosis.
Collapse
Affiliation(s)
- L Yang
- a Department of Anesthesiology , Xijing Hospital, the Fourth Military Medical University , Xi'an , P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Dziegiel P, Pula B, Kobierzycki C, Stasiolek M, Podhorska-Okolow M. Metallothioneins in Normal and Cancer Cells. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2016; 218:1-117. [PMID: 26847563 DOI: 10.1007/978-3-319-27472-0_1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metallothioneins (MTs) are low molecular weight proteins, which are present in almost all types of organisms. In mammals, four main MT isoforms designated from MT-1 to MT-4 were identified. Their biological role, according to their characteristic structure, was shown to be mostly associated with cellular metabolism of metal ions, especially zinc. Moreover, the available evidence suggests broad regulatory properties of MTs in the control of cell senescence and various pathological processes including neurodegeneration, cardiovascular pathology, metabolic disorders, and various malignancies. This extensive review provides general in formation on the structure of MT family members and the cellular functions of MT-1, MT-2, and MT-4 isoforms as well as insights into divergent biological roles of MT-3. Due to the involvement of MT molecules in various processes related to carcinogenesis, an organ-specific presentation of current data concerning their potential impact on the progression of various tumors is given. The regulatory role of MT family members in the function of the immune system is also discussed in depth.
Collapse
Affiliation(s)
- Piotr Dziegiel
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland.,Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Bartosz Pula
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Christopher Kobierzycki
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland.,Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | | |
Collapse
|
25
|
Ryan TE, Schmidt CA, Green TD, Brown DA, Neufer PD, McClung JM. Mitochondrial Regulation of the Muscle Microenvironment in Critical Limb Ischemia. Front Physiol 2015; 6:336. [PMID: 26635622 PMCID: PMC4649016 DOI: 10.3389/fphys.2015.00336] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe clinical presentation of peripheral arterial disease and manifests as chronic limb pain at rest and/or tissue necrosis. Current clinical interventions are largely ineffective and therapeutic angiogenesis based trials have shown little efficacy, highlighting the dire need for new ideas and novel therapeutic approaches. Despite a decade of research related to skeletal muscle as a determinant of morbidity and mortality outcomes in CLI, very little progress has been made toward an effective therapy aimed directly at the muscle myopathies of this disease. Within the muscle cell, mitochondria are well positioned to modulate the ischemic cellular response, as they are the principal sites of cellular energy production and the major regulators of cellular redox charge and cell death. In this mini review, we update the crucial importance of skeletal muscle to CLI pathology and examine the evolving influence of muscle and endothelial cell mitochondria in the complex ischemic microenvironment. Finally, we discuss the novelty of muscle mitochondria as a therapeutic target for ischemic pathology in the context of the complex co-morbidities often associated with CLI.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
26
|
Emeny RT, Kasten-Jolly J, Mondal T, Lynes MA, Lawrence DA. Metallothionein differentially affects the host response to Listeria infection both with and without an additional stress from cold-restraint. Cell Stress Chaperones 2015; 20:1013-22. [PMID: 26267326 PMCID: PMC4595426 DOI: 10.1007/s12192-015-0630-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 01/04/2023] Open
Abstract
Acute stress alters anti-bacterial defenses, but the neuroimmunological mechanisms underlying this association are not yet well understood. Metallothionein (MT), a cysteine-rich protein, is a stress response protein that is induced by a variety of chemical, biological, and psychological stressors, and MT has been shown to influence immune activities. We investigated MT's role in the management of anti-bacterial responses that occur during stress, using a C57BL/6 (B6) strain that has targeted disruptions of the Mt1 and Mt2 genes (B6-MTKO), and a B6 strain that has additional copies of Mt (B6-MTTGN). The well-characterized listeriosis model was used to examine immune mechanisms that are altered by a 1-h stress treatment (cold-restraint, CR) administered just prior to bacterial infection. Intriguingly, MT gene doses both greater and lower than that of wild-type (WT) B6 mice were associated with improved host defenses against Listeria monocytogenes (LM). This augmented protection was diminished by CR stress in the MTKO mice, but transgenic mice with additional MT copies had no CR stress-induced increase in their listerial burden. During the transition from innate to adaptive immunity, on day 3 after infection, oxidative burst and apoptosis were assessed by flow cytometric methods, and cytokine transcription was measured by real-time quantitative PCR. MT gene expression and CR-stress affected the expression of IL-6 and TNFα. Additionally, these genetic and environmental modulations altered the generation of ROS responses as well as the number of apoptotic cells in livers and spleens. Although the level of MT altered the listerial response, MT expression was equally elevated by listerial infection with or without CR stress. These results indicate the ability of MT to regulate immune response mechanisms and demonstrate that increased amounts of MT can eliminate the immunosuppression induced by CR.
Collapse
Affiliation(s)
- Rebecca T Emeny
- Laboratory of Immunology, Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY, 12201, USA
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, GmbH, Ingolstädter Landstraße 1, D-85764, Neuherberg, Germany
| | - Jane Kasten-Jolly
- Laboratory of Immunology, Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY, 12201, USA
| | - Tapan Mondal
- Laboratory of Immunology, Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY, 12201, USA
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - David A Lawrence
- Laboratory of Immunology, Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY, 12201, USA.
| |
Collapse
|
27
|
Hu N, Yang L, Dong M, Ren J, Zhang Y. Deficiency in adiponectin exaggerates cigarette smoking exposure-induced cardiac contractile dysfunction: Role of autophagy. Pharmacol Res 2015; 100:175-89. [PMID: 26276084 DOI: 10.1016/j.phrs.2015.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/17/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
Abstract
Second hand smoke is an independent risk factor for cardiovascular disease. Adiponectin (APN), an adipose-derived adipokine, has been shown to offer cardioprotective effect through an AMPK-dependent manner. This study was designed to evaluate the impact of adiponectin deficiency on second hand smoke-induced cardiac pathology and underlying mechanisms using a mouse model of side-stream smoke exposure. Adult wild-type (WT) and adiponectin knockout (APNKO) mice were placed in a chamber exposed to cigarette smoke for 1 hour daily for 40 days. Echocardiographic, cardiomyocyte function, and intracellular Ca2+ handling were evaluated. Autophagy and apoptosis were examined using western blot. 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) staining was used to evaluate reactive oxygen species (ROS) generation. Masson trichrome staining was employed to measure interstitial fibrosis. Our data revealed that adiponectin deficiency provoked smoke exposure-induced cardiomyopathy (compromised fractional shortening, disrupted cardiomyocyte function and intracellular Ca2+ homeostasis, apoptosis and ROS generation). In addition, these detrimental effects of side-stream smoke were accompanied by defective autophagolysosome formation, the effect of which was exacerbated by adiponectin deficiency. Blocking autophagolysosome formation using bafilomycin A1 (BafA1) negated the cardioprotective effect of rapamycin against smoke extract. Induction of autophagy using rapamycin and AMPKα activation using AICAR rescued against smoke extract-induced myopathic anomalies in APNKO mice. Our data suggest that adiponectin serves as an indispensable cardioprotective factor against side-stream smoke exposure-induced myopathic changes possibly through facilitating autophagolysosome formation.
Collapse
Affiliation(s)
- Nan Hu
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Lifang Yang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China; Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Maolong Dong
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China; Department of Burn and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Jun Ren
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - Yingmei Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
28
|
Metallothionein-I/II Knockout Mice Aggravate Mitochondrial Superoxide Production and Peroxiredoxin 3 Expression in Thyroid after Excessive Iodide Exposure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:267027. [PMID: 26101557 PMCID: PMC4458558 DOI: 10.1155/2015/267027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/13/2015] [Accepted: 02/11/2015] [Indexed: 12/30/2022]
Abstract
Purpose. We aim to figure out the effect of metallothioneins on iodide excess induced oxidative stress in the thyroid. Methods. Eight-week-old MT-I/II knockout (MT-I/II KO) mice and background-matched wild-type (WT) mice were used. Mitochondrial superoxide production and peroxiredoxin (Prx) 3 expression were measured. Results. In in vitro study, more significant increases in mitochondrial superoxide production and Prx 3 expression were detected in the MT-I/II KO groups. In in vivo study, significantly higher concentrations of urinary iodine level were detected in MT-I/II KO mice in 100 HI group. Compared to the NI group, there was no significant difference existing in serum thyroid hormones level in either groups (P > 0.05), while the mitochondrial superoxide production was significantly increased in 100 HI groups with significantly increased LDH activity and decreased relative cell viability. Compared to WT mice, more significant changes were detected in MT-I/II KO mice in 100 HI groups. No significant differences were detected between the NI group and 10 HI group in both the MT-I/II KO and WT mice groups (P > 0.05). Conclusions. Iodide excess in a thyroid without MT I/II protection may result in strong mitochondrial oxidative stress, which further leads to the damage of thyrocytes.
Collapse
|
29
|
Wu Z, He EY, Scott GI, Ren J. α,β-Unsaturated aldehyde pollutant acrolein suppresses cardiomyocyte contractile function: Role of TRPV1 and oxidative stress. ENVIRONMENTAL TOXICOLOGY 2015; 30:638-647. [PMID: 24376112 DOI: 10.1002/tox.21941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/12/2013] [Accepted: 12/12/2013] [Indexed: 06/03/2023]
Abstract
Air pollution is associated with an increased prevalence of heart disease and is known to trigger a proinflammatory response via stimulation of transient receptor potential vanilloid cation channels (TRPV1, also known as the capsaicin receptor). This study was designed to examine the effect of acrolein, an essential α,β-unsaturated aldehyde pollutant, on myocardial contractile function and the underlying mechanism involved with a focus on TRPV1 and oxidative stress. Cardiomyocyte mechanical and intracellular Ca(2+) properties were evaluated using an IonOptix MyoCam® system including peak shortening (PS), maximal velocity of shortening/relengthening (± dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90 ), fura-2 fluorescence intensity (FFI) and intracellular Ca(2+) decay. Changes in apoptosis and TRPV1 were evaluated using Western blot analysis. The degree of oxidative stress was assessed using the ratio between reduced and oxidized glutathione. Results obtained revealed that exposure of cardiomyocytes to acrolein acutely compromised contractile and intracellular Ca(2+) properties including depressed PS, ± dL/dt and ΔFFI, as well as prolonged TR90 and intracellular Ca(2+) decay. In addition, acrolein exposure upregulated TRPV1 associated with an increase in both apoptosis and oxidative stress. However, the acrolein-induced cardiomyocyte contractile and intracellular Ca(2+) anomalies, as well as apoptosis (as evidenced by Bcl-2, Bax, FasL, Caspase-3 and -8), were negated by the reactive oxygen species (ROS) scavenger glutathione or the TRPV1 antagonist capsazepine. Collectively these data suggest that the α,β-unsaturated aldehyde pollutant acrolein may play a role in the pathogenesis and sequelae of air pollution-induced heart disease via a TRPV1- and oxidative stress-dependent mechanism.
Collapse
Affiliation(s)
- Zhenbiao Wu
- Department of Clinical Immunology, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, China
| | - Emily Y He
- Department of Clinical Immunology, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, China
- Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, Wyoming, 82071
| | - Glenda I Scott
- Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, Wyoming, 82071
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, Wyoming, 82071
| |
Collapse
|
30
|
Vlahos R, Bozinovski S. Preclinical murine models of Chronic Obstructive Pulmonary Disease. Eur J Pharmacol 2015; 759:265-71. [PMID: 25818750 DOI: 10.1016/j.ejphar.2015.03.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/03/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a major incurable global health burden and is the 4th leading cause of death worldwide. It is believed that an exaggerated inflammatory response to cigarette smoke causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T lymphocytes are prominent, leads to oxidative stress, emphysema, small airway fibrosis and mucus hypersecretion. Much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and infectious (viral and bacterial) exacerbations (AECOPD). Comorbidities, defined as other chronic medical conditions, in particular skeletal muscle wasting and cardiovascular disease markedly impact on disease morbidity, progression and mortality. The mechanisms and mediators underlying COPD and its comorbidities are poorly understood and current COPD therapy is relatively ineffective. Thus, there is an obvious need for new therapies that can prevent the induction and progression of COPD and effectively treat AECOPD and comorbidities of COPD. Given that access to COPD patients can be difficult and that clinical samples often represent a "snapshot" at a particular time in the disease process, many researchers have used animal modelling systems to explore the mechanisms underlying COPD, AECOPD and comorbidities of COPD with the goal of identifying novel therapeutic targets. This review highlights the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and the recent advances in modelling infectious exacerbations and comorbidities of COPD.
Collapse
Affiliation(s)
- Ross Vlahos
- School of Health Sciences, Health Innovations Research Institute, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia; Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Steven Bozinovski
- School of Health Sciences, Health Innovations Research Institute, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia; Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
31
|
Molecular signatures in the prevention of radiation damage by the synergistic effect of N-acetyl cysteine and qingre liyan decoction, a traditional chinese medicine, using a 3-dimensional cell culture model of oral mucositis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:425760. [PMID: 25705238 PMCID: PMC4326035 DOI: 10.1155/2015/425760] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/24/2014] [Indexed: 02/03/2023]
Abstract
Qingre Liyan decoction (QYD), a Traditional Chinese medicine, and N-acetyl cysteine (NAC) have been used to prevent radiation induced mucositis. This work evaluates the protective mechanisms of QYD, NAC, and their combination (NAC-QYD) at the cellular and transcriptional level. A validated organotypic model of oral mucosal consisting of a three-dimensional (3D) cell tissue-culture of primary human keratinocytes exposed to X-ray irradiation was used. Six hours after the irradiation, the tissues were evaluated by hematoxylin and eosin (H and E) and a TUNEL assay to assess histopathology and apoptosis, respectively. Total RNA was extracted and used for microarray gene expression profiling. The tissue-cultures treated with NAC-QYD preserved their integrity and showed no apoptosis. Microarray results revealed that the NAC-QYD caused the upregulation of genes encoding metallothioneins, HMOX1, and other components of the Nrf2 pathway, which protects against oxidative stress. DNA repair genes (XCP, GADD45G, RAD9, and XRCC1), protective genes (EGFR and PPARD), and genes of the NFκB pathway were upregulated. Finally, tissue-cultures treated prophylactically with NAC-QYD showed significant downregulation of apoptosis, cytokines and chemokines genes, and constrained damage-associated molecular patterns (DAMPs). NAC-QYD treatment involves the protective effect of Nrf2, NFκB, and DNA repair factors.
Collapse
|
32
|
Combined effects of current-smoking and the aldehyde dehydrogenase 2*2 allele on the risk of myocardial infarction in Japanese patients. Toxicol Lett 2015; 232:221-5. [DOI: 10.1016/j.toxlet.2014.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 01/27/2023]
|
33
|
|
34
|
Pei Z, Zhuang Z, Sang H, Wu Z, Meng R, He EY, Scott GI, Maris JR, Li R, Ren J. α,β-Unsaturated aldehyde crotonaldehyde triggers cardiomyocyte contractile dysfunction: role of TRPV1 and mitochondrial function. Pharmacol Res 2014; 82:40-50. [PMID: 24705155 DOI: 10.1016/j.phrs.2014.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/11/2014] [Accepted: 03/26/2014] [Indexed: 11/25/2022]
Abstract
Recent evidence has suggested that cigarette smoking is associated with an increased prevalence of heart diseases. Given that cigarette smoking triggers proinflammatory response via stimulation of the capsaicin-sensitive transient receptor potential cation channel TRPV1, this study was designed to evaluate the effect of an essential α,β-unsaturated aldehyde from cigarette smoke crotonaldehyde on myocardial function and the underlying mechanism with a focus on TRPV1 and mitochondria. Cardiomyocyte mechanical and intracellular Ca2+ properties were evaluated including peak shortening (PS), maximal velocity of shortening/relengthening (±dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90), fura-2 fluorescence intensity (FFI), intracellular Ca2+ decay and SERCA activity. Apoptosis and TRPV1 were evaluated using Western blot analysis. Production of reactive oxygen species (ROS) and DNA damage were measured using the intracellular fluoroprobe 5-(6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate and 8-hydroxy-2'-deoxyguanosine (8-OHdG), respectively. Our data revealed that crotonaldehyde interrupted cardiomyocyte contractile and intracellular Ca2+ property including depressed PS, ±dL/dt, ΔFFI and SERCA activity, as well as prolonged TR90 and intracellular Ca2+ decay. Crotonaldehyde exposure increased TRPV1 and NADPH oxidase levels, promoted apoptosis, mitochondrial injury (decreased aconitase activity, PGC-1α and UCP-2) as well as production of ROS and 8-OHdG. Interestingly, crotonaldehyde-induced cardiac defect was obliterated by the ROS scavenger glutathione and the TRPV1 inhibitor capsazepine. Capsazepine (not glutathione) ablated crotonaldehyde-induced mitochondrial damage. Capsazepine, glutathione and the NADPH inhibitor apocynin negated crotonaldehyde-induced ROS accumulation. Our data suggest a role of crotonaldehyde compromises cardiomyocyte mechanical function possibly through a TRPV1- and mitochondria-dependent oxidative stress mechanism.
Collapse
Affiliation(s)
- Zhaohui Pei
- Department of Cardiology, The Third Hospital of Nanchang, Nanchang, Jiangxi 330009, China
| | - Zhiqiang Zhuang
- Department of Rehabilitation Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Hanfei Sang
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Clinical Pharmacology, Bethune International Peace Hospital of People's Liberation Army, Shijiazhuang, Hebei 050082, China
| | - Zhenbiao Wu
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Rongsen Meng
- Department of Cardiology, The Second People's Hospital of Guangdong Province, Guangzhou, Guangdong 511442, China
| | - Emily Y He
- Department of Clinical Immunology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China; University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Glenda I Scott
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Jackie R Maris
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Ruiman Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 511442, China.
| | - Jun Ren
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
35
|
Therapeutic targets in heart failure: refocusing on the myocardial interstitium. J Am Coll Cardiol 2014; 63:2188-98. [PMID: 24657693 DOI: 10.1016/j.jacc.2014.01.068] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 12/28/2013] [Accepted: 01/14/2014] [Indexed: 01/13/2023]
Abstract
New therapeutic targets, agents, and strategies are needed to prevent and treat heart failure (HF) after a decade of failed research efforts to improve long-term patient outcomes, especially in patients after hospitalization for HF. Conceptually, an accurate assessment of left ventricular structure is an essential step in the development of novel therapies because heterogeneous pathophysiologies underlie chronic HF and hospitalization for HF. Improved left ventricular characterization permits the identification and targeting of the intrinsic fundamental disease-modifying pathways that culminate in HF. Interstitial heart disease is one such pathway, characterized by extracellular matrix (ECM) expansion that is associated with mechanical, electrical, and vasomotor dysfunction and adverse outcomes. Previous landmark trials that appear to treat interstitial heart disease were effective in improving outcomes. Advances in cardiovascular magnetic resonance now enable clinicians and researchers to assess the interstitium and quantify ECM expansion using extracellular volume fraction measures and other derangements in cardiovascular structure. These capabilities may provide a mechanistic platform to advance understanding of the role of the ECM, foster the development of novel therapeutics, and target specific disease-modifying pathways intrinsic to the ventricle. Refocusing on the interstitium may potentially improve care through the identification and targeted treatment of key patient subgroups.
Collapse
|
36
|
Hu N, Dong M, Ren J. Hydrogen sulfide alleviates cardiac contractile dysfunction in an Akt2-knockout murine model of insulin resistance: role of mitochondrial injury and apoptosis. Am J Physiol Regul Integr Comp Physiol 2014; 306:R761-71. [PMID: 24622975 DOI: 10.1152/ajpregu.00327.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hydrogen sulfide (H2S) is a toxic gas now being recognized as an endogenous signaling molecule in multiple organ systems, in particular, the cardiovascular system. H2S is known to regulate cardiac function and protect against ischemic injury. However, little information is available regarding the effect of H2S on cardiac function in insulin resistance. This study was designed to examine the impact of H2S supplementation on cardiac function using an Akt2 knockout model of insulin resistance. Wild-type and Akt2 knockout mice were treated with NaHS (50 μM·kg(-1)·day(-1) ip for 10 days) prior to evaluation of echocardiographic, cardiomyocyte contractile, and intracellular Ca(2+) properties, apoptosis, and mitochondrial damage. Our results revealed that Akt2 ablation led to overtly enlarged ventricular end-systolic diameter, reduced myocardial and cardiomyocyte contractile function, and disrupted intracellular Ca(2+) homeostasis and apoptosis, the effects of which were ameliorated by H2S. Furthermore, Akt2 knockout displayed upregulated apoptotic protein markers (Bax, caspase-3, caspase-9, and caspace-12) and mitochondrial damage (reduced aconitase activity and NAD(+), elevated cytochrome-c release from mitochondria) along with reduced phosphorylation of PTEN, Akt, and GSK3β in the absence of changes in pan protein expression, the effects of which were abolished or significantly ameliorated by H2S treatment. In vitro data revealed that H2S-induced beneficial effect against Akt2 ablation was obliterated by mitochondrial uncoupling. Taken together, our findings suggest the H2S may reconcile Akt2 knockout-induced myocardial contractile defect and intracellular Ca(2+) mishandling, possibly via attenuation of mitochondrial injury and apoptosis.
Collapse
Affiliation(s)
- Nan Hu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming; and
| | - Maolong Dong
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming; and Department of Burn and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming; and
| |
Collapse
|
37
|
Vlahos R, Bozinovski S. Recent advances in pre-clinical mouse models of COPD. Clin Sci (Lond) 2014; 126:253-65. [PMID: 24144354 PMCID: PMC3878607 DOI: 10.1042/cs20130182] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/17/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023]
Abstract
COPD (chronic obstructive pulmonary disease) is a major incurable global health burden and will become the third largest cause of death in the world by 2020. It is currently believed that an exaggerated inflammatory response to inhaled irritants, in particular cigarette smoke, causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T-cells are prominent, leads to oxidative stress, emphysema, small airways fibrosis and mucus hypersecretion. The mechanisms and mediators that drive the induction and progression of chronic inflammation, emphysema and altered lung function are poorly understood. Current treatments have limited efficacy in inhibiting chronic inflammation, do not reverse the pathology of disease and fail to modify the factors that initiate and drive the long-term progression of disease. Therefore there is a clear need for new therapies that can prevent the induction and progression of COPD. Animal modelling systems that accurately reflect disease pathophysiology continue to be essential to the development of new therapies. The present review highlights some of the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and whether they can be used to predict the efficacy of new therapeutics for COPD.
Collapse
Key Words
- acute exacerbations of chronic obstructive pulmonary disease (aecopd)
- chronic obstructive pulmonary disease (copd)
- emphysema
- inflammation
- skeletal muscle wasting
- smoking
- aecopd, acute exacerbations of copd
- bal, bronchoalveolar lavage
- balf, bal fluid
- copd, chronic obstructive pulmonary disease
- gm-csf, granulocyte/macrophage colony-stimulating factor
- gold, global initiative on chronic obstructive lung disease
- gpx, glutathione peroxidase
- hdac, histone deacetylation
- il, interleukin
- ltb4, leukotriene b4
- mapk, mitogen-activated protein kinase
- mcp-1, monocyte chemotactic protein-1
- mmp, matrix metalloproteinase
- ne, neutrophil elastase
- nf-κb, nuclear factor κb
- nrf2, nuclear erythroid-related factor 2
- o2•−, superoxide radical
- onoo−, peroxynitrite
- pde, phosphodiesterase
- pi3k, phosphoinositide 3-kinase
- ros, reactive oxygen species
- rv, rhinovirus
- slpi, secretory leucocyte protease inhibitor
- sod, superoxide dismutase
- tgf-β, transforming growth factor-β
- timp, tissue inhibitor of metalloproteinases
- tnf-α, tumour necrosis factor-α
- v/q, ventilation/perfusion
Collapse
Affiliation(s)
- Ross Vlahos
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Steven Bozinovski
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
38
|
Hu N, Ren J. Nicotine, cigarette smoking and cardiac function: an update. Toxicol Res (Camb) 2014. [DOI: 10.1039/c3tx50044f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
39
|
Zhang L, Huang D, Shen D, Zhang C, Ma Y, Babcock SA, Chen B, Ren J. Inhibition of protein kinase C βII isoform ameliorates methylglyoxal advanced glycation endproduct-induced cardiomyocyte contractile dysfunction. Life Sci 2013; 94:83-91. [PMID: 24269213 DOI: 10.1016/j.lfs.2013.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 01/02/2023]
Abstract
AIMS Accumulation of advanced glycation endproduct (AGE) contributes to diabetic complication including diabetic cardiomyopathy although the precise underlying mechanism still remains elusive. Recent evidence depicted a pivotal role of protein kinase C (PKC) in diabetic complications. To this end, this study was designed to examine if PKCβII contributes to AGE-induced cardiomyocyte contractile and intracellular Ca(2+) aberrations. MAIN METHODS Adult rat cardiomyocytes were incubated with methylglyoxal-AGE (MG-AGE) in the absence or presence of the PKCβII inhibitor LY333531 for 12h. Contractile and intracellular Ca(2+) properties were assessed using an IonOptix system including peak shortening (PS), maximal velocity of shortening/relengthening (±dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90), rise in intracellular Ca(2+) Fura-2 fluorescence intensity and intracellular Ca(2+) decay. Oxidative stress, O2(-) production and mitochondrial integrity were examined using TBARS, fluorescence imaging, aconitase activity and Western blotting. KEY FINDINGS MG-AGE compromised contractile and intracellular Ca(2+) properties including reduced PS, ±dL/dt, prolonged TPS and TR90, decreased electrically stimulated rise in intracellular Ca(2+) and delayed intracellular Ca(2+) clearance, the effects of which were ablated by the PKCβII inhibitor LY333531. Inhibition of PKCβII rescued MG-AGE-induced oxidative stress, O2(-) generation, cell death, apoptosis and mitochondrial injury (reduced aconitase activity, UCP-2 and PGC-1α). In vitro studies revealed that PKCβII inhibition-induced beneficial effects were replicated by the NADPH oxidase inhibitor apocynin and were mitigated by the mitochondrial uncoupler FCCP. SIGNIFICANCE These findings implicated the therapeutic potential of specific inhibition of PKCβII isoform in the management of AGE accumulation-induced myopathic anomalies.
Collapse
Affiliation(s)
- Liwei Zhang
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China.
| | - Dangsheng Huang
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China
| | - Dong Shen
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China
| | - Chunhong Zhang
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China
| | - Yongjiang Ma
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China
| | - Sara A Babcock
- University of Wyoming College of Health Sciences, Laramie, WY 82071, United States
| | - Bingyang Chen
- Cardiology Department, The First Affiliated Hospital of PLA General Hospital, Beijing, 100037, China
| | - Jun Ren
- University of Wyoming College of Health Sciences, Laramie, WY 82071, United States; Department of Cardiology, Xijing Hospital, Fourth Military Medical University Xi'an, 710032, China.
| |
Collapse
|
40
|
Wang Z, Zhang Y, Guo J, Jin K, Li J, Guo X, Scott GI, Zheng Q, Ren J. Inhibition of protein kinase C βII isoform rescues glucose toxicity-induced cardiomyocyte contractile dysfunction: role of mitochondria. Life Sci 2013; 93:116-24. [PMID: 23770211 DOI: 10.1016/j.lfs.2013.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/16/2013] [Accepted: 06/02/2013] [Indexed: 12/28/2022]
Abstract
AIMS Hyperglycemia leads to cytotoxicity in the heart. Although theories were postulated for glucose toxicity-induced cardiomyocyte dysfunction including oxidative stress, the mechanism involved still remains unclear. Recent evidence has depicted a role of protein kinase C (PKC) in diabetic complications while high concentrations of glucose stimulate PKC. This study examined the role of PKCβII in glucose toxicity-induced cardiomyocyte contractile and intracellular Ca(2+) aberrations. MAIN METHODS Adult rat cardiomyocytes were maintained in normal (NG, 5.5 mM) or high glucose (HG, 25.5 mM) medium for 12 h. Contractile and intracellular Ca(2+) properties were measured using a video edge-detection system including peak shortening (PS), maximal velocity of shortening/relengthening (±dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90), rise in intracellular Ca(2+) Fura-2 fluorescence intensity and intracellular Ca(2+) decay. Production of ROS/O2(-) and mitochondrial integrity were examined using fluorescence imaging, aconitase activity and Western blotting. KEY FINDINGS High glucose triggered abnormal contractile and intracellular Ca(2+) properties including reduced PS, ±dL/dt, prolonged TR90, decreased electrically-stimulated rise in intracellular Ca(2+) and delayed intracellular Ca(2+) clearance, the effects of which were ablated by the PKCβII inhibitor LY333531. Inhibition of PKCβII rescued glucose toxicity-induced generation of ROS and O2(-), apoptosis, cell death and mitochondrial injury (reduced aconitase activity, UCP-2 and PGC-1α). In vitro studies revealed that PKCβII inhibition-induced beneficial effects were mimicked by the NADPH oxidase inhibitor apocynin and were canceled off by mitochondrial uncoupling using FCCP. SIGNIFICANCE These findings suggest the therapeutic potential of specific inhibition of PKCβII isoform in the management of hyperglycemia-induced cardiac complications.
Collapse
Affiliation(s)
- Zikuan Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | | | | | | | | | |
Collapse
|