1
|
Li H, Bu L, Sun X, Chu X, Xue Y, Zhang M, Shi J, Liu Y, Guan S, Han X, Wang H. Mechanistic investigation of the ameliorative effect of liquiritin on hypoxia/reoxygenation‑induced cardiomyocyte injury based on network pharmacology and in vitro validation. Exp Ther Med 2024; 27:117. [PMID: 38361515 PMCID: PMC10867724 DOI: 10.3892/etm.2024.12405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/18/2023] [Indexed: 02/17/2024] Open
Abstract
Liquiritin (LIQ) is a flavonoid known for its cardioprotective properties, extracted from Glycyrrhiza uralensis Fisch. The purpose of the present study was to investigate the protective mechanism of LIQ against hypoxia/reoxygenation (H/R) injury through in vitro experiments, with the goal of enhancing its pharmacological effects. Initially, network pharmacology was employed to explore the targets and mechanisms of LIQ. Subsequently, an in vitro H/R model was established using H9c2 cells. Potential targets for LIQ and myocardial ischemia-reperfusion injury (MIRI) were identified through online databases. The STRING, Cytoscape and DAVID databases were used to extract intersecting targets and mechanisms. In vitro experiments were conducted to validate these findings, assessing cardiac enzymes, oxidative stress indicators, mitochondrial fluorescence, apoptotic fluorescence, inflammation and related protein expression. The network pharmacological analysis revealed that the protective effects of LIQ on MIRI involve oxidative stress, inflammation and apoptosis. The results of in vitro experimental validation demonstrated that LIQ significantly reduced the activities of lactated dehydrogenase and creatine kinase isoenzyme-MB (P<0.05 or 0.01), as well as the level of malondialdehyde (P<0.01). It also inhibited the production of reactive oxygen species (P<0.01), the release of inflammatory factors (P<0.05 or 0.01) and apoptosis (P<0.01). By contrast, the LIQ pre-treatment group exhibited a significant increase in mitochondrial membrane potential level (P<0.05 or 0.01) and the activities of antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase (P<0.05 or 0.01). Furthermore, LIQ reduced the protein expressions of TNF-α receptor type 1 (TNFR1) and MMP9, along with the level of NF-κB phosphorylation (P<0.05 or 0.01). In conclusion, LIQ mitigated H/R-induced cardiomyocyte injury through mechanisms that may involve antioxidants, anti-apoptotic effects, protection against mitochondrial damage and suppression of inflammatory levels. These effects are achieved via inhibition of the TNFR1/NF-κB/MMP9 pathway.
Collapse
Affiliation(s)
- Haoying Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Linlin Bu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xiaoqi Sun
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yucong Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Muqing Zhang
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yanshuang Liu
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
- College of Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Hongfang Wang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
2
|
Lu W, Khatibi Shahidi F, Khorsandi K, Hosseinzadeh R, Gul A, Balick V. An update on molecular mechanisms of curcumin effect on diabetes. J Food Biochem 2022; 46:e14358. [PMID: 35945662 DOI: 10.1111/jfbc.14358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/16/2022] [Accepted: 07/19/2022] [Indexed: 12/01/2022]
Abstract
Owing to its prevalent nature, diabetes mellitus has become one of the most serious endocrine illnesses affecting a patient's quality of life due to the manifestation of side effects such as cardiovascular diseases, retinopathy, neuropathy, and nephropathy. Curcumin ((1E, 6E) 21, 7-bis (4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione), a major compound of turmeric, has been used in conventional medicine because of its safe nature and cost-effectiveness to meliorate diabetes and its comorbidities. These effects have also been observed in rodent models of diabetes resulting in a reduction of glycemia and blood lipids. Both the preventive and therapeutic activities of this compound are due to its antioxidant and anti-inflammatory characteristics. Furthermore, preclinical outcomes and clinical investigation demonstrate that the use of curcumin neutralizes insulin resistance, obesity, and hyperglycemia. Despite the many benefits of curcumin, its two limiting factors, solubility and bioavailability, remain a challenge for researchers; therefore, several methods such as drug formulation, nano-drug delivery, and the use of curcumin analogs have been developed to deliver curcumin and increase its bioavailability. PRACTICAL APPLICATIONS: The rise of people with type 2 diabetes has become a major concern at the global healthcare level. The best diabetes treatments today are anti-diabetic drug administration, lifestyle-related interventions (such as healthy eating and daily physical activity), arterial pressure detection, and fat control. The polyphenol curcumin, found in turmeric, can promote health by acting on a variety of cellular signaling pathways. This review article discusses curcumin and its role in the treatment of diabetes.
Collapse
Affiliation(s)
- Wensong Lu
- People's Hospital of Longhua, Shenzhen, China
| | - Fedora Khatibi Shahidi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.,Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Reza Hosseinzadeh
- Department of Chromatography Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Urmia, Iran
| | - Asma Gul
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University, Islamabad, Pakistan
| | - Veronica Balick
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
3
|
Sivakumar B, Kurian GA. PM 2.5 Exposure Lowers Mitochondrial Endurance During Cardiac Recovery in a Rat Model of Myocardial Infarction. Cardiovasc Toxicol 2022; 22:545-557. [PMID: 35404004 DOI: 10.1007/s12012-022-09737-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/25/2022] [Indexed: 11/28/2022]
Abstract
Many studies have reported the negative effect of PM2.5 exposure on heart function which is likely to impair postcardiac surgery rehabilitation that is involved in recovery and wound healing, yet the direct effects of PM2.5 from diesel exhaust (DPM) on cardiac recovery is unknown. To study the impact of DPM on cardiac recovery and repair, we utilized isoproterenol induced myocardial infarction (MI) model where female rats were exposed to DPM prior and after MI induction. The experimental groups comprise of normal, ISO control, DPM control (42 days of exposure), DPM exposed prior (21 days) and after (21 days) MI induction (D + I + D) and DPM exposed (21 days) after MI (I + D). Post-MI rat hearts from D + I + D group exhibited higher fibrosis, elevated cardiac injury and altered electrophysiology, where this pathology was also observed in I + D group animals which was mild. Loss of mitochondrial quality was evident in DPM exposed animals with and without MI, where severe mitochondrial damage persisted in D + I + D group. In addition, these animals showed striking decline in ETC enzyme activity, ATP levels, mitochondrial copy number and down regulation of PGC1-α, TFAM and POLG along with the genes involved in mitophagy and mitofusion. Besides, the MI associated inactivation of cardio protective signalling pathways like PI3K and Akt were persistent in D + I + D group. In fact, I + D group animals also showed a similar pattern of change, but in a mild form. Taken together, exposure to PM2.5 increases the risk, frequency or progression of MI by impairing the recovery potential of the myocardium.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Gino A Kurian
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India. .,School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur, Tamil Nadu, 613401, India.
| |
Collapse
|
4
|
Sivakumar B, Kurian GA. Inhalation of PM 2.5 from diesel exhaust promote impairment of mitochondrial bioenergetics and dysregulate mitochondrial quality in rat heart: implications in isoproterenol-induced myocardial infarction model. Inhal Toxicol 2022; 34:107-119. [PMID: 35290147 DOI: 10.1080/08958378.2022.2049931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Aim: Ambient exposure of PM2.5 from diesel exhaust (termed as diesel particulate matter [DPM]) can induce cardiotoxicity that can be manifested into myocardial ischemia/infarction, where the survival depends on mitochondrial function. The mechanism for DPM-induced mitochondrial dysfunction is yet to be elucidated and the consequential impact of impaired mitochondria on the severity of myocardial infarction (MI) has not been established.Materials and methods: Female Wistar rats were exposed to DPM (0.5 mg/ml) for 3 h daily (to achieve a PM2.5 concentration of 250 µg/m3) for 21 d trailed by an induction of MI using isoproterenol (ISO).Conclusion: DPM exposure altered the basal ECG pattern and increased heart weight (HW) to body weight (BW) ratio from control. Loss of mitochondrial quality in the cardiac tissue was observed in DPM exposed animals, measured via declined ETC enzyme activity, reduced ATP levels, high oxidative stress, low mitochondrial copy number, and low expression of the mitochondrial genes involved in mitophagy (PINK and PARKIN) and mitochondrial fusion (MFN-1). Subsequent induction of MI in DPM exposed animals (DPM + ISO) further deteriorated the normal sinus rhythm, accompanied by elevated plasma CK and LDH level, increased myocardial caspase activity, downregulation of Peroxisome proliferator-activated receptor-gamma coactivator (PGC1-α), transcription factor A (TFAM), DNA polymerase subunit gamma (POLG), and other mitochondrial quality control genes. Based on these results, we conclude that DPM alters the electrophysiology and ultrastructure of the heart that aggravates the MI-induced cardiotoxicity, where the diminished mitochondrial quality can be the potential contributor.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- School of Chemical and Biotechnology, Vascular Biology lab, SASTRA Deemed University, Thanjavur, India
| | - Gino A Kurian
- School of Chemical and Biotechnology, Vascular Biology lab, SASTRA Deemed University, Thanjavur, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, India
| |
Collapse
|
5
|
Zhao L, Han L, Wei X, Zhou Y, Zhang Y, Si N, Wang H, Yang J, Bian B, Zhao H. Toxicokinetics of Arenobufagin and its Cardiotoxicity Mechanism Exploration Based on Lipidomics and Proteomics Approaches in Rats. Front Pharmacol 2022; 12:780016. [PMID: 35002716 PMCID: PMC8727535 DOI: 10.3389/fphar.2021.780016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
Arenobufagin (ArBu), one of the main active bufadienolides of toad venom with cardiotonic effect, analgesic effect, and outstanding anti-tumor potentiality, is also a potential cardiotoxic component. In the present study, the cardiac effect of ArBu and its underlying mechanism were explored by integrating data such as heart rates, toxicokinetics, myocardial enzyme and brain natriuretic peptide (BNP) activity, pathological sections, lipidomics and proteomics. Under different doses, the cardiac effects turned out to be different. The oral dose of 60 mg/kg of ArBu sped up the heart rate. However, 120 mg/kg ArBu mainly reduced the heart rate. Over time, they all returned to normal, consisting of the trend of ArBu concentration-time curve. High concentrations of myocardial enzymes and BNP indicated that ArBu inhibited or impaired the cardiac function of rats. Pathological sections of hearts also showed that ArBu caused myocardial fiber disorder and rupture, in which the high-dose group was more serious. At the same time, serum and heart tissue lipidomics were used to explore the changes in body lipid metabolism under different doses. The data indicated a larger difference in the high-dose ArBu group. There were likewise many significant differences in the proteomics of the heart. Furthermore, a multi-layered network was used to integrate the above information to explore the potential mechanism. Finally, 4 proteins that were shown to be significantly and differentially expressed were validated by targeted proteomics using parallel reaction monitoring (PRM) analysis. Our findings indicated that ArBu behaved as a bidirectional regulation of the heart. The potential mechanism of cardiac action was revealed with the increased dose, which provided a useful reference for the safety of clinical application of ArBu.
Collapse
Affiliation(s)
- Lijuan Zhao
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Shaanxi Chinese Medicine Institute (Shaanxi Pharmaceutical Information Center), Xianyang, China
| | - Lingyu Han
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaolu Wei
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanyan Zhou
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqiong Zhang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Si
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjie Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian Yang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolin Bian
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Zhang X, Li G. Effects of a mitochondrial-acting drug on ischemia/reperfusion-induced ventricular arrhythmias, cardiac mitochondrial function and inflammatory cytokines in rat: Role of adenosine triphosphate-sensitive potassium channels. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221115041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Ischemia/reperfusion (IR)-induced myocardial arrhythmias are a common clinical manifestation in patients with myocardial infarction after reperfusion therapy. Mitochondria play a critical role in cardioprotection. Here, we investigated the effects of KH176 as a new mitochondrial-acting drug on IR-induced ventricular arrhythmias, mitochondrial function, pro-inflammatory cytokines production, and the role of mitochondrial ATP-dependent K (mK-ATP) channels in rats’ hearts. Methods The hearts of Sprague Dawley rats (250 ± 30 g; 36 rats) underwent 35 min of ischemia followed by 120 min of reperfusion. Myocardial in vivo ischemia was induced by ligation of the left anterior descending coronary artery. KH176 at concentrations of 10 and 50 μM was intraperitoneally injected to rats 10 min before reperfusion onset. Ventricular arrhythmias were quantified during reperfusion, and cardiac mitochondrial function, nitric oxide, and pro-inflammatory cytokines levels were measured by fluorometric, spectrophotometric, and ELISA techniques. Results Administration of KH176 significantly reduced lactate-dehydrogenase release and the number, duration, incidence, and severity of ventricular arrhythmias induced by reperfusion injury. IR-induced elevation of mitochondrial reactive oxygen species production, and cardiac pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, as well as reduction of mitochondrial membrane potential, ATP and nitric oxide levels were significantly restored by KH176 at 50 μM. However, the blockade of mK-ATP channels by 5-hydroxydecanoate considerably inhibited the effects of KH176 on all parameters except nitric oxide. Conclusion KH176 showed strong cardiac antiarrhythmic effects on IR-induced injury through improving mitochondrial function and reducing inflammatory and oxidative responses, and these protective effects are mediated by cardiac mK-ATP channels.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacy, Jiangsu Taizhou People’s Hospital, China
| | - Geng Li
- Department of Cardiology, Hubei Third People's Hospital Affiliated to Jianghan University, Wuhan, Hubei, China
| |
Collapse
|
7
|
The dynamin-related protein 1 is decreased and the mitochondrial network is altered in Friedreich's ataxia cardiomyopathy. Int J Biochem Cell Biol 2021; 143:106137. [PMID: 34923139 DOI: 10.1016/j.biocel.2021.106137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022]
Abstract
Friedreich ataxia is an autosomal recessive congenital neurodegenerative disease caused by a deficiency in the frataxin protein and is often diagnosed in young adulthood. An expansion of guanine-adenine-adenine repeats in the first intron of the FXN gene leads to decreased frataxin expression. Frataxin plays an essential role in mitochondrial metabolism. Most Friedreich ataxia patients are diagnosed with left ventricular hypertrophic cardiomyopathy, and 60% of patients die with hypertrophic cardiomyopathy. However, the mitochondrial anatomy in Friedreich ataxia hypertrophic cardiomyopathy is still poorly understood. We investigated mitochondrial fission, fusion, and function using biochemical, microscopy, and computational stochastic analysis in human induced pluripotent stem cell derived cardiomyocytes from a patient with Friedreich ataxia hypertrophic cardiomyopathy and a healthy individual. We found a significantly higher mitochondrial footprint, decreased mitochondrial fission protein dynamin-related protein, and mitochondrial fission rate over fusion with more giant mitochondrial clusters in human induced pluripotent stem cell derived cardiomyocytes from a patient with Friedreich ataxia hypertrophic cardiomyopathy, compared to an unaffected individual. We also found significantly depolarized mitochondrial membrane potential and higher reactive oxygen species levels in Friedreich ataxia human induced pluripotent stem cell cardiomyocytes. Our results show that frataxin's depletion may dampen the mitochondrial fission machinery by reducing dynamin-related protein1. The loss of mitochondrial fission might lead to elevated reactive oxygen species and depolarized mitochondrial membrane potential, which may cause oxidative damage in Friedreich ataxia hypertrophic cardiomyopathy. Further investigations are needed to identify the mechanism of downregulating dynamin-related protein1 due to the frataxin deficiency in Friedreich ataxia hypertrophic cardiomyopathy.
Collapse
|
8
|
Rajabi M, Vafaee MS, Hosseini L, Badalzadeh R. Pretreatment with Nicotinamide Mononucleotide Increases the Effect of Ischemic-Postconditioning on Cardioprotection and Mitochondrial Function Following ex vivo Myocardial Reperfusion Injury in Aged Rats. Clin Exp Pharmacol Physiol 2021; 49:474-482. [PMID: 34854121 DOI: 10.1111/1440-1681.13616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/11/2021] [Accepted: 11/20/2021] [Indexed: 11/29/2022]
Abstract
The present study aims to evaluate the combined effect of ischemic-postconditioning (IPostC) and nicotinamide mononucleotide (NMN) on cardioprotection and mitochondrial function in aged rats subjected to myocardial ischemia-reperfusion (IR) injury. Sixty aged Wistar rats were randomly divided into 5 groups (n=12), including sham, control, NMN, IPostC, and NMN+IPostC. Regional ischemia was induced by 30-min occlusion of the left anterior descending coronary artery (LAD) followed by 60-min reperfusion. IPostC was applied at the onset of reperfusion, by 6 cycles of 10-s reperfusion/ischemia. NMN (100 mg/kg) was intraperitoneally injected every other day for 28 days before IR. Myocardial hemodynamics and infarct size (IS) were measured, and the left ventricles samples were harvested to assess cardiac mitochondrial function. The results showed that all treatments reduced lactate dehydrogenase release compared to those of the control group. IPostC alone failed to reduce IS and myocardial function. However, NMN and combined therapy could significantly improve myocardial function and decrease the IS compared to the control animals. Moreover, the effects of combined therapy on the decrease of IS, mitochondrial reactive oxygen species (ROS), and improvement of mitochondrial membrane potential (MMP) were greater than those of alone treatments. These results demonstrated that cardioprotection by combined therapy with NMN+IPostC was superior to individual treatments, and pretreatment of aged rats with NMN was able to correct the failure of IPostC in protecting the hearts of aged rats against IR injury.
Collapse
Affiliation(s)
- Mojgan Rajabi
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr S Vafaee
- Psychiatry Research Unit, Southern Denmark Region, Odense, Denmark.,Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Leila Hosseini
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Shi X, Li Y, Wang Y, Ding T, Zhang X, Wu N. Pharmacological postconditioning with sappanone A ameliorates myocardial ischemia reperfusion injury and mitochondrial dysfunction via AMPK-mediated mitochondrial quality control. Toxicol Appl Pharmacol 2021; 427:115668. [PMID: 34358556 DOI: 10.1016/j.taap.2021.115668] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/26/2023]
Abstract
Pharmacological postconditioning (PPC), drug intervention before or during the early minutes of reperfusion, could stimulate cardioprotection as ischemic postconditioning. In this study, we examined whether PPC with sappanone A (SA), a homoisoflavanone with potent antioxidant and anti-inflammatory activity, has a protective effect on myocardial ischemia reperfusion injury (MIRI), and explored the underlying mechanism. A MIRI model was established using the Langendorff method. After 30 min of ischemia, isolated rat hearts were treated with SA at the onset of reperfusion to stimulate PPC. The changes in myocardial infarct size, mitochondrial function, mitochondrial biogenesis, mitophagy, and mitochondrial fission and fusion were detected. The results showed that SA postconditioning decreased the myocardial infarct size, inhibited the release of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and cardiac troponin (cTnI), as well as improved cardiac function, enhanced myocardial ATP content and mitochondrial complex activity, and prevented the loss of mitochondrial membrane potential and opening of mitochondrial permeability transition pore (mPTP). Mechanistically, we found that SA was an AMP-activated protein kinase (AMPK) activator, and SA postconditioning could facilitate mitochondrial biogenesis by increasing mitochondrial DNA (mtDNA) copy number and the expression of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α). In addition, it balanced mitochondrial dynamics by decreasing fission and increasing fusion, and enhanced mitophagy in an AMPK-dependent manner. Moreover, AMPK silencing abolished the cardioprotection of SA postconditioning. Collectively, our study demonstrated that SA postconditioning ameliorated MIRI and mitochondrial dysfunction by regulation of mitochondrial quality control via activating AMPK. This finding provides a new insight into pharmacological action and clinical use of SA.
Collapse
Affiliation(s)
- Xiaojing Shi
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Yang Li
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Yaguang Wang
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Tiejia Ding
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, PR China.
| | - Nan Wu
- The Central Laboratory of the First Affiliated Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
10
|
All-Trans Retinoic Acid Increases DRP1 Levels and Promotes Mitochondrial Fission. Cells 2021; 10:cells10051202. [PMID: 34068960 PMCID: PMC8156392 DOI: 10.3390/cells10051202] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
In the heart, mitochondrial homeostasis is critical for sustaining normal function and optimal responses to metabolic and environmental stressors. Mitochondrial fusion and fission are thought to be necessary for maintaining a robust population of mitochondria, and disruptions in mitochondrial fission and/or fusion can lead to cellular dysfunction. The dynamin-related protein (DRP1) is an important mediator of mitochondrial fission. In this study, we investigated the direct effects of the micronutrient retinoid all-trans retinoic acid (ATRA) on the mitochondrial structure in vivo and in vitro using Western blot, confocal, and transmission electron microscopy, as well as mitochondrial network quantification using stochastic modeling. Our results showed that ATRA increases DRP1 protein levels, increases the localization of DRP1 to mitochondria in isolated mitochondrial preparations. Our results also suggested that ATRA remodels the mitochondrial ultrastructure where the mitochondrial area and perimeter were decreased and the circularity was increased. Microscopically, mitochondrial network remodeling is driven by an increased rate of fission over fusion events in ATRA, as suggested by our numerical modeling. In conclusion, ATRA results in a pharmacologically mediated increase in the DRP1 protein. It also results in the modulation of cardiac mitochondria by promoting fission events, altering the mitochondrial network, and modifying the ultrastructure of mitochondria in the heart.
Collapse
|
11
|
Yehualashet AS, Belachew TF, Kifle ZD, Abebe AM. Targeting Cardiac Metabolic Pathways: A Role in Ischemic Management. Vasc Health Risk Manag 2020; 16:353-365. [PMID: 32982263 PMCID: PMC7501978 DOI: 10.2147/vhrm.s264130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Among the vast number of noncommunicable diseases encountered worldwide, cardiovascular diseases accounted for about 17.8 million deaths in 2017 and ischemic heart disease (IHD) remains the single-largest cause of death in countries across all income groups. Because conventional medications are not without shortcomings and patients still refractory to these medications, scientific investigation is ongoing to advance the management of IHD, and shows a great promise for better treatment modalities, but additional research can warrant improvement in terms of the quality of life of patients. Metabolic modulation is one promising strategy for the treatment of IHD, because alterations in energy metabolism are involved in progression of the disease. Therefore, the purpose of this review was to strengthen attention toward the use of metabolic modulators and to review the current level of knowledge on cardiac energy metabolic pathways.
Collapse
Affiliation(s)
- Awgichew Shewasinad Yehualashet
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
| | | | - Zemene Demelash Kifle
- School of Pharmacy, Department of Pharmacology, University of Gondar, Gondar, Ethiopia
| | - Ayele Mamo Abebe
- Department of Nursing, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
| |
Collapse
|
12
|
Purushottam Dharaskar S, Paithankar K, Kanugovi Vijayavittal A, Shabbir Kara H, Amere Subbarao S. Mitochondrial chaperone, TRAP1 modulates mitochondrial dynamics and promotes tumor metastasis. Mitochondrion 2020; 54:92-101. [PMID: 32784002 DOI: 10.1016/j.mito.2020.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria play a central role in regulating cellular energy metabolism. However, the present understanding of mitochondria has changed from its unipotent functions to pluripotent and insists on understanding the role of mitochondria not only in regulating the life and death of cells, but in pathological conditions such as cancer. Unlike other cellular organelles, subtle alterations in mitochondrial organization may significantly influence the balance between metabolic networks and cellular behavior. Therefore, the delicate balance between the fusion and fission dynamics of mitochondrion can indicate cell fate. Here, we present mitochondrial chaperone TRAP1 influence on mitochondrial architecture and its correlation with tumor growth and metastasis. We show that TRAP1 overexpression (TRAP1 OE) promotes mitochondrial fission, whereas, TRAP1 knockdown (TRAP1 KD) promotes mitochondrial fusion. Interestingly, TRAP1 OE or KD had a negligible effect on mitochondrial integrity. However, TRAP1 OE cells exhibited enhanced proliferative potential, while TRAP1 KD cells showing increased doubling time. Further, TRAP1 dependent mitochondrial dynamic alterations appeared to be unique since mitochondrial localization of TRAP1 is a mandate for dynamic changes. The expression patterns of fusion and fission genes have failed to correlate with TRAP1 expression, indicating a possibility that the dynamic changes can be independent of these genes. In agreement with enhanced proliferative potential, TRAP1 OE cells also exhibited enhanced migration in vitro and tumor metastasis in vivo. Further, TRAP1 OE cells showed altered homing properties, which may challenge site-specific anticancer treatments. Our findings unravel the TRAP1 role in tumor metastasis, which is in addition to altered energy metabolism.
Collapse
Affiliation(s)
- Shrikant Purushottam Dharaskar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, Telangana, India; AcSIR - Academy of Scientific & Innovative Research, Government of India, India
| | - Khanderao Paithankar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, Telangana, India
| | | | - Hatim Shabbir Kara
- Presently at Life Sciences & Chemistry, Jacobs University Bremen gGmbh, Bremen, Germany
| | - Sreedhar Amere Subbarao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, Telangana, India.
| |
Collapse
|
13
|
Olmedo I, Pino G, Riquelme JA, Aranguiz P, Díaz MC, López-Crisosto C, Lavandero S, Donoso P, Pedrozo Z, Sánchez G. Inhibition of the proteasome preserves Mitofusin-2 and mitochondrial integrity, protecting cardiomyocytes during ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165659. [PMID: 31891806 DOI: 10.1016/j.bbadis.2019.165659] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 12/13/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022]
Abstract
Cardiomyocyte loss is the main cause of myocardial dysfunction following an ischemia-reperfusion (IR) injury. Mitochondrial dysfunction and altered mitochondrial network dynamics play central roles in cardiomyocyte death. Proteasome inhibition is cardioprotective in the setting of IR; however, the mechanisms underlying this protection are not well-understood. Several proteins that regulate mitochondrial dynamics and energy metabolism, including Mitofusin-2 (Mfn2), are degraded by the proteasome. The aim of this study was to evaluate whether proteasome inhibition can protect cardiomyocytes from IR damage by maintaining Mfn2 levels and preserving mitochondrial network integrity. Using ex vivo Langendorff-perfused rat hearts and in vitro neonatal rat ventricular myocytes, we showed that the proteasome inhibitor MG132 reduced IR-induced cardiomyocyte death. Moreover, MG132 preserved mitochondrial mass, prevented mitochondrial network fragmentation, and abolished IR-induced reductions in Mfn2 levels in heart tissue and cultured cardiomyocytes. Interestingly, Mfn2 overexpression also prevented cardiomyocyte death. This effect was apparently specific to Mfn2, as overexpression of Miro1, another protein implicated in mitochondrial dynamics, did not confer the same protection. Our results suggest that proteasome inhibition protects cardiomyocytes from IR damage. This effect could be partly mediated by preservation of Mfn2 and therefore mitochondrial integrity.
Collapse
Affiliation(s)
- Ivonne Olmedo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Gonzalo Pino
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Jaime A Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380492, Chile; Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago de Chile 8380492, Chile
| | - Pablo Aranguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, Viña del Mar 2520000, Chile
| | - Magda C Díaz
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380492, Chile; Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Pontificia Universidad Javeriana de Cali, Colombia
| | - Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380492, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380492, Chile; Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago de Chile 7680201, Chile; Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Paulina Donoso
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Zully Pedrozo
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380492, Chile.
| | - Gina Sánchez
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile.
| |
Collapse
|
14
|
The Role of Mitochondria in the Mechanisms of Cardiac Ischemia-Reperfusion Injury. Antioxidants (Basel) 2019; 8:antiox8100454. [PMID: 31590423 PMCID: PMC6826663 DOI: 10.3390/antiox8100454] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria play a critical role in maintaining cellular function by ATP production. They are also a source of reactive oxygen species (ROS) and proapoptotic factors. The role of mitochondria has been established in many aspects of cell physiology/pathophysiology, including cell signaling. Mitochondria may deteriorate under various pathological conditions, including ischemia-reperfusion (IR) injury. Mitochondrial injury can be one of the main causes for cardiac and other tissue injuries by energy stress and overproduction of toxic reactive oxygen species, leading to oxidative stress, elevated calcium and apoptotic and necrotic cell death. However, the interplay among these processes in normal and pathological conditions is still poorly understood. Mitochondria play a critical role in cardiac IR injury, where they are directly involved in several pathophysiological mechanisms. We also discuss the role of mitochondria in the context of mitochondrial dynamics, specializations and heterogeneity. Also, we wanted to stress the existence of morphologically and functionally different mitochondrial subpopulations in the heart that may have different sensitivities to diseases and IR injury. Therefore, various cardioprotective interventions that modulate mitochondrial stability, dynamics and turnover, including various pharmacologic agents, specific mitochondrial antioxidants and uncouplers, and ischemic preconditioning can be considered as the main strategies to protect mitochondrial and cardiovascular function and thus enhance longevity.
Collapse
|
15
|
Protective effects of echinacoside against anoxia/reperfusion injury in H9c2 cells via up-regulating p-AKT and SLC8A3. Biomed Pharmacother 2018; 104:52-59. [PMID: 29763795 DOI: 10.1016/j.biopha.2018.04.188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/25/2018] [Accepted: 04/29/2018] [Indexed: 11/21/2022] Open
Abstract
Echinacoside is a natural ingredient with various pharmacological activities. In this study, we investigated the protective effects of echinacoside on cardiomyocytes (rat H9c2 cells) in an anoxia/reperfusion (A/R) model. Further, the regulatory function of sodium-calcium exchanger protein 3 (SLC8A3/NCX3) as well as the protein kinase B (AKT) signaling were studied. The present results indicated that echinacoside protected against A/R-induced apoptosis in a dose manner, which was characterized by a decrease in the apoptosis and caspase 3 protein levels in H9c2 cells. Further, Ca2+ uptake were dose-dependently reduced in H9c2 cells by echinacoside under A/R conditions. Whereas, relative mRNA expression of SLC8A3 and protein levels of SLC8A3 and p-AKT showed opposite tendency. On the one hand, the A/R-induced abnormalities in H9c2 cells were remarkably ameliorated by activated p-AKT and over-expression of SLC8A3 but aggravated by inhibited p-AKT, and the aggravated effection were ameliorated by echinacoside. Moreover, protein levels of SLC8A3 were positively regulated by p-AKT signaling. On the other hand, apoptosis and Ca2+ uptake as well as protein levels of caspase 3 were significantly increased by SLC8A3 silencing in H9c2 cells under normoxic conditions, and this symptom was remarkably reversed by echinacoside or Nimodipine (an antagonis of Ca2+) treatment. Collectively, echinacoside has showed a cardioprotective effect against A/R treatment in a dose dependent manner in vitro, and this cardioprotective effect was potentially achieved via up-regulating p-AKT and SLC8A3.
Collapse
|
16
|
Xia CY, Liu Y, Yang HR, Yang HY, Liu JX, Ma YN, Qi Y. Reference Intervals of Mitochondrial DNA Copy Number in Peripheral Blood for Chinese Minors and Adults. Chin Med J (Engl) 2017; 130:2435-2440. [PMID: 29052564 PMCID: PMC5684636 DOI: 10.4103/0366-6999.216395] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) content measured by different techniques cannot be compared between studies, and age- and tissue-related control values are hardly available. In the present study, we aimed to establish the normal reference range of mtDNA copy number in the Chinese population. METHODS Two healthy cohorts of 200 Chinese minors (0.1-18.0 years) and 200 adults (18.0-88.0 years) were recruited. Then, they were further categorized into eight age groups. The absolute mtDNA copy number per cell was measured by a quantitative real-time polymerase chain reaction. We subsequently used this range to evaluate mtDNA content in four patients (0.5-4.0 years) with molecularly proven mitochondrial depletion syndromes (MDSs) and 83 cases of mitochondrial disease patients harboring the m.3243A>G mutation. RESULTS The reference range of mtDNA copy number in peripheral blood was 175-602 copies/cell (mean: 325 copies/cell) in minors and 164-500 copies/cell (mean: 287 copies/cell) in adults. There was a decreasing trend in mtDNA copy number in blood with increasing age, especially in 0-2-year-old and >50-year-old donors. The mean mtDNA copy number level among the mitochondrial disease patients with m.3243A>G mutation was significantly higher than that of healthy controls. The mtDNA content of POLG, DGUOK, TK2, and SUCLA2 genes in blood samples from MDS patients was reduced to 25%, 38%, 32%, and 24%, respectively. CONCLUSIONS We primarily establish the reference intervals of mtDNA copy number, which might contribute to the clinical diagnosis and monitoring of mitochondrial disease.
Collapse
Affiliation(s)
- Chang-Yu Xia
- Department of Central Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Yu Liu
- Department of Central Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Hui-Rong Yang
- Department of Clinical Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Hong-Yun Yang
- Department of Clinical Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Jing-Xia Liu
- Department of Clinical Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Yi-Nan Ma
- Department of Central Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Yu Qi
- Department of Central Laboratory, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
17
|
Lesnefsky EJ, Chen Q, Hoppel CL. Mitochondrial Metabolism in Aging Heart. Circ Res 2017; 118:1593-611. [PMID: 27174952 DOI: 10.1161/circresaha.116.307505] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Altered mitochondrial metabolism is the underlying basis for the increased sensitivity in the aged heart to stress. The aged heart exhibits impaired metabolic flexibility, with a decreased capacity to oxidize fatty acids and enhanced dependence on glucose metabolism. Aging impairs mitochondrial oxidative phosphorylation, with a greater role played by the mitochondria located between the myofibrils, the interfibrillar mitochondria. With aging, there is a decrease in activity of complexes III and IV, which account for the decrease in respiration. Furthermore, aging decreases mitochondrial content among the myofibrils. The end result is that in the interfibrillar area, there is ≈50% decrease in mitochondrial function, affecting all substrates. The defective mitochondria persist in the aged heart, leading to enhanced oxidant production and oxidative injury and the activation of oxidant signaling for cell death. Aging defects in mitochondria represent new therapeutic targets, whether by manipulation of the mitochondrial proteome, modulation of electron transport, activation of biogenesis or mitophagy, or the regulation of mitochondrial fission and fusion. These mechanisms provide new ways to attenuate cardiac disease in elders by preemptive treatment of age-related defects, in contrast to the treatment of disease-induced dysfunction.
Collapse
Affiliation(s)
- Edward J Lesnefsky
- From the Division of Cardiology, Department of Medicine, Pauley Heart Center (E.J.L, Q.C.), Departments of Biochemistry and Molecular Biology and Physiology and Biophsyics (E.J.L.), Virginia Commonwealth University, Richmond, VA (E.J.L., Q.C.); Medical Service, McGuire Veterans Affairs Medical Center, Richmond, VA (E.J.L.); and Departments of Pharmacology (C.L.H.) and Medicine (E.J.L., C.L.H.), Center for Mitochondrial Disease (C.L.H.), Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Qun Chen
- From the Division of Cardiology, Department of Medicine, Pauley Heart Center (E.J.L, Q.C.), Departments of Biochemistry and Molecular Biology and Physiology and Biophsyics (E.J.L.), Virginia Commonwealth University, Richmond, VA (E.J.L., Q.C.); Medical Service, McGuire Veterans Affairs Medical Center, Richmond, VA (E.J.L.); and Departments of Pharmacology (C.L.H.) and Medicine (E.J.L., C.L.H.), Center for Mitochondrial Disease (C.L.H.), Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Charles L Hoppel
- From the Division of Cardiology, Department of Medicine, Pauley Heart Center (E.J.L, Q.C.), Departments of Biochemistry and Molecular Biology and Physiology and Biophsyics (E.J.L.), Virginia Commonwealth University, Richmond, VA (E.J.L., Q.C.); Medical Service, McGuire Veterans Affairs Medical Center, Richmond, VA (E.J.L.); and Departments of Pharmacology (C.L.H.) and Medicine (E.J.L., C.L.H.), Center for Mitochondrial Disease (C.L.H.), Case Western Reserve University, School of Medicine, Cleveland, OH.
| |
Collapse
|
18
|
Li Y, Li F, Chen S, Duan Y, Guo Q, Wang W, Wen C, Yin Y. Protein-Restricted Diet Regulates Lipid and Energy Metabolism in Skeletal Muscle of Growing Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:9412-9420. [PMID: 27960278 DOI: 10.1021/acs.jafc.6b03959] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The aim of this work was to study the lipid metabolism and energy status of skeletal muscle of pigs as affected by dietary protein restriction. Eighteen growing pigs were distributed into three treatments, and each group was fed one of three levels (20, 17, and 14%) of crude protein (CP) diets. Our results showed that pigs fed the 20% CP had greater (P < 0.05) gain:feed and muscle weight than those fed the 14% CP, but no differences between the 20 and 17% CP treatments. Additionally, protein restriction tended to increase (P = 0.07) the content of intramuscular fat (IMF) and up-regulated (P < 0.05) expression of lipogenic-related genes. Energy status was changed and, concomitantly, AMP-activated protein kinase α pathway was inhibited by reducing the dietary protein level. These results indicate protein restriction could be useful to improve IMF content of pigs through regulating lipid metabolism and associated energy utilization in muscle.
Collapse
Affiliation(s)
- Yinghui Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- University of Chinese Academy of Sciences , Beijing 10008, China
| | - Fengna Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients , Changsha, Hunan 410128, China
| | - Shuai Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- University of Chinese Academy of Sciences , Beijing 10008, China
| | - Yehui Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- University of Chinese Academy of Sciences , Beijing 10008, China
| | - Qiuping Guo
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- University of Chinese Academy of Sciences , Beijing 10008, China
| | - Wenlong Wang
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| | - Chaoyue Wen
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture , Changsha, Hunan 410125, China
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University , Changsha, Hunan 410018, China
| |
Collapse
|
19
|
Wende AR, Young ME, Chatham J, Zhang J, Rajasekaran NS, Darley-Usmar VM. Redox biology and the interface between bioenergetics, autophagy and circadian control of metabolism. Free Radic Biol Med 2016; 100:94-107. [PMID: 27242268 PMCID: PMC5124549 DOI: 10.1016/j.freeradbiomed.2016.05.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 12/01/2022]
Abstract
Understanding molecular mechanisms that underlie the recent emergence of metabolic diseases such as diabetes and heart failure has revealed the need for a multi-disciplinary research integrating the key metabolic pathways which change the susceptibility to environmental or pathologic stress. At the physiological level these include the circadian control of metabolism which aligns metabolism with temporal demand. The mitochondria play an important role in integrating the redox signals and metabolic flux in response to the changing activities associated with chronobiology, exercise and diet. At the molecular level this involves dynamic post-translational modifications regulating transcription, metabolism and autophagy. In this review we will discuss different examples of mechanisms which link these processes together. An important pathway capable of linking signaling to metabolism is the post-translational modification of proteins by O-linked N-acetylglucosamine (O-GlcNAc). This is a nutrient regulated protein modification that plays an important role in impaired cellular stress responses. Circadian clocks have also emerged as critical regulators of numerous cardiometabolic processes, including glucose/lipid homeostasis, hormone secretion, redox status and cardiovascular function. Central to these pathways are the response of autophagy, bioenergetics to oxidative stress, regulated by Keap1/Nrf2 and mechanisms of metabolic control. The extension of these ideas to the emerging concept of bioenergetic health will be discussed.
Collapse
Affiliation(s)
- Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John Chatham
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namakkal S Rajasekaran
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor M Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; UAB Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
20
|
Wang X, Dong W, Yuan B, Yang Y, Yang D, Lin X, Chen C, Zhang W. Vitamin E confers cytoprotective effects on cardiomyocytes under conditions of heat stress by increasing the expression of metallothionein. Int J Mol Med 2016; 37:1429-36. [PMID: 27035111 DOI: 10.3892/ijmm.2016.2543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 03/18/2016] [Indexed: 11/05/2022] Open
Abstract
Heat stress (HS) is commonly used to refer to the heat load that an individual is subjected to due to either metabolic heat, or environmental factors, including high temperatures and high humidity levels. HS has been reported to affect and even damage the functioning of various organs; overexposure to high temperatures and high humidity may lead to accidental deaths. It has been suggested that the cardiovascular system is primarily targeted by exposure to HS conditions; the HS-induced dysfunction of cardiomyocytes, which is characterized by mitochondrial dysfunction, may result in the development of cardiovascular diseases. The excessive production of reactive oxygen species (ROS) also participates in mitochondrial dysfunction. However, effective methods for the prevention and treatment of mitochondrial and cardiovascular dysfunction induced by exposure to HS are lacking. In the present study, we hypothesized that vitamin E (VE), an antioxidant, is capable of preventing oxidative stress and mitochondrial injury in cardiomyocytes induced by exposure to HS. The results revealed that pre‑treatment with VE increased the expression of metallothionein (MT), which has previously been reported to confer cytoprotective effects, particularly on the cardiovascular system. Pre-treatment with VE restored mitochondrial function in cardiomyocytes under conditions of HS by increasing the expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM), and by increasing adenosine triphosphate (ATP) levels. Furthermore, pre-treatment with VE decreased the production of ROS, which was induced by exposure to HS and thus exerted antioxidant effects. In addition, pre-treatment with VE attenuated oxidative stress induced by exposure to HS, as demonstrated by the increased levels of antioxidant enzymes [superoxide dismutase (SOD) and glutathione (GSH)], and by the decreased levels of markers of oxidative injury [malondialdehyde (MDA) and lactate dehydrogenase (LDH)]. Taken together, these findings suggest that pre-treatment with VE can prevent mitochondrial dysfunction and oxidative stress in cardiomyocytes induced by exposure to HS, by increasing the expression of MT.
Collapse
Affiliation(s)
- Xiaowu Wang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Wenpeng Dong
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Binbin Yuan
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Yongchao Yang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Dongpeng Yang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Xi Lin
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Changfu Chen
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Weida Zhang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
21
|
Chen K, Li J, Chen K, Hou X, Mai H, Xue X. The Mechanism of <i>Cornus officinalis</i> Total Glycosides and Cornus Polysaccharide on Myocardial Protection in Rats with Acute Myocardial Infarction. Chin Med 2016. [DOI: 10.4236/cm.2016.72007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
22
|
Nishida K, Otsu K. Autophagy during cardiac remodeling. J Mol Cell Cardiol 2015; 95:11-8. [PMID: 26678624 DOI: 10.1016/j.yjmcc.2015.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/23/2015] [Accepted: 12/04/2015] [Indexed: 11/18/2022]
Abstract
Despite progress in cardiovascular research and evidence-based therapies, heart failure is a leading cause of morbidity and mortality in industrialized countries. Cardiac remodeling is a chronic maladaptive process, characterized by progressive ventricular dilatation, cardiac hypertrophy, fibrosis, and deterioration of cardiac performance, and arises from interactions between adaptive modifications of cardiomyocytes and negative aspects of adaptation such as cardiomyocyte death and fibrosis. Autophagy has evolved as a conserved process for bulk degradation and recycling of cytoplasmic components, such as long-lived proteins and organelles. Accumulating evidence demonstrates that autophagy plays an essential role in cardiac remodeling to maintain cardiac function and cellular homeostasis in the heart. This review discusses some recent advances in understanding the role of autophagy during cardiac remodeling. This article is part of a Special Issue entitled: Autophagy in the Heart.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, SE5 9NU, UK.
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, SE5 9NU, UK
| |
Collapse
|
23
|
Protective Effect of Sevoflurane Postconditioning against Cardiac Ischemia/Reperfusion Injury via Ameliorating Mitochondrial Impairment, Oxidative Stress and Rescuing Autophagic Clearance. PLoS One 2015; 10:e0134666. [PMID: 26263161 PMCID: PMC4532466 DOI: 10.1371/journal.pone.0134666] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/13/2015] [Indexed: 01/29/2023] Open
Abstract
Background and Purpose Myocardial infarction leads to heart failure. Autophagy is excessively activated in myocardial ischemia/reperfusion (I/R) in rats. The aim of this study is to investigate whether the protection of sevoflurane postconditioning (SPC) in myocardial I/R is through restored impaired autophagic flux. Methods Except for the sham control (SHAM) group, each rat underwent 30 min occlusion of the left anterior descending coronary (LAD) followed by 2 h reperfusion. Cardiac infarction was determined by 2,3,5-triphenyltetrazolium chloride triazole (TTC) staining. Cardiac function was examined by hemodynamics and echocardiography. The activation of autophagy was evaluated by autophagosome accumulation, LC3 conversion and p62 degradation. Potential molecular mechanisms were investigated by immunoblotting, real-time PCR and immunofluorescence staining. Results SPC improved the hemodynamic parameters, cardiac dysfunction, histopathological and ultrastructural damages, and decreased myocardial infarction size after myocardial I/R injury (P < 0.05 vs. I/R group). Compared with the cases in I/R group, myocardial ATP and NAD+ content, mitochondrial function related genes and proteins, and the expressions of SOD2 and HO-1 were increased, while the expressions of ROS and Vimentin were decreased in the SPC group (P < 0.05 vs. I/R group). SPC significantly activated Akt/mTOR signaling, and inhibited the formation of Vps34/Beclin1 complex via increasing expression of Bcl2 protein (P < 0.05 vs. I/R group). SPC suppressed elevated expressions of LC3 II/I ratio, Beclin1, Atg5 and Atg7 in I/R rat, which indicated that SPC inhibited over-activation of autophagy, and promoted autophagosome clearance. Meanwhile, SPC significantly suppressed the decline of Opa1 and increases of Drp1 and Parkin induced by I/R injury (P < 0.05 vs. I/R group). Moreover, SPC maintained the contents of ATP by reducing impaired mitochondria. Conclusion SPC protects rat hearts against I/R injury via ameliorating mitochondrial impairment, oxidative stress and rescuing autophagic clearance.
Collapse
|
24
|
Dănilă MD, Privistirescu AI, Mirica SN, Sturza A, Ordodi V, Noveanu L, Duicu OM, Muntean DM. Acute inhibition of monoamine oxidase and ischemic preconditioning in isolated rat hearts: interference with postischemic functional recovery but no effect on infarct size reduction. Can J Physiol Pharmacol 2015; 93:819-25. [PMID: 26322912 DOI: 10.1139/cjpp-2015-0103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Monoamine oxidases (MAOs) have recently emerged as important mitochondrial sources of oxidative stress in the cardiovascular system. Generation of reactive oxygen species during the brief episodes of ischemic preconditioning (IPC) is responsible for the cardioprotection at reperfusion. The aim of this study was to assess the effects of two MAO inhibitors (clorgyline and pargyline) on the IPC-related protection in isolated rat hearts. Animals subjected to 30 min global ischemia and 120 min reperfusion were assigned to the following groups: (i) Control, no additional intervention; (ii) IPC, 3 cycles of 5 min ischemia and 5 min reperfusion before the index ischemia; (iii) IPC-clorgyline, IPC protocol bracketed for 5 min with clorgyline (50 μmol/L); (iv) IPC-pargyline, IPC protocol bracketed for 5 min with pargyline (0.5 mmol/L). The postischemic functional recovery was assessed by the left ventricular developed pressure (LVDP) and the indices of contractility (+dLVP/dt max) and relaxation (-dLVP/dt max). Infarct size (IS) was quantified by TTC staining. In both genders, IPC significantly improved functional recovery that was further enhanced in the presence of either clorgyline or pargyline. IS reduction was comparable among all the preconditioned groups, regardless of the presence of MAO inhibitors. In isolated rat hearts, acute inhibition of MAOs potentiates the IPC-induced postischemic functional recovery without interfering with the anti-necrotic protection.
Collapse
Affiliation(s)
- Maria D Dănilă
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Andreea I Privistirescu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Silvia N Mirica
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Adrian Sturza
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Valentin Ordodi
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania
| | - Lavinia Noveanu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Oana M Duicu
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| | - Danina M Muntean
- a Department of Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timișoara, Romania.,b Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy of Timişoara, Timişoara, Romania
| |
Collapse
|
25
|
Xie Q, Deng Y, Huang C, Liu P, Yang Y, Shen W, Gao P. Chemerin-induced mitochondrial dysfunction in skeletal muscle. J Cell Mol Med 2015; 19:986-95. [PMID: 25754411 PMCID: PMC4420601 DOI: 10.1111/jcmm.12487] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/10/2014] [Indexed: 01/18/2023] Open
Abstract
Chemerin is a novel adipocyte-derived factor that induces insulin resistance in skeletal muscle. However, the effect of chemerin on skeletal muscle mitochondrial function has received little attention. In the present study, we investigated whether mitochondrial dysfunction is involved in the pathogenesis of chemerin-mediated insulin resistance. In this study, we used recombinant adenovirus to express murine chemerin in C57BL/6 mice. The mitochondrial function and structure were evaluated in isolated soleus muscles from mice. The oxidative mechanism of mitochondrial dysfunction in cultured C2C12 myotubes exposed to recombinant chemerin was analysed by western blotting, immunofluorescence and quantitative real-time polymerase chain reaction. The overexpression of chemerin in mice reduced the muscle mitochondrial content and increased mitochondrial autophagy, as determined by the increased conversion of LC3-I to LC3-II and higher expression levels of Beclin1 and autophagy-related protein-5 and 7. The chemerin treatment of C2C12 myotubes increased the generation of mitochondrial reactive oxygen species, concomitant with a reduced mitochondrial membrane potential and increased the occurrence of mitochondrial protein carbonyls and mitochondrial DNA deletions. Knockdown of the expression of chemokine-like receptor 1 or the use of mitochondria-targeting antioxidant Mito-TEMPO restored the mitochondrial dysfunction induced by chemerin. Furthermore, chemerin exposure in C2C12 myotubes not only reduced the insulin-stimulated phosphorylation of protein kinase B (AKT) but also dephosphorylated forkhead box O3α (FoxO3α). Chemerin-induced mitochondrial autophagy likely through an AKT-FoxO3α-dependent signalling pathway. These findings provide direct evidence that chemerin may play an important role in regulating mitochondrial remodelling and function in skeletal muscle.
Collapse
Affiliation(s)
- Qihai Xie
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Zhang J, Wang C, Yu S, Luo Z, Chen Y, Liu Q, Hua F, Xu G, Yu P. Sevoflurane postconditioning protects rat hearts against ischemia-reperfusion injury via the activation of PI3K/AKT/mTOR signaling. Sci Rep 2014; 4:7317. [PMID: 25471136 PMCID: PMC4255182 DOI: 10.1038/srep07317] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/17/2014] [Indexed: 12/28/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway plays a key role in myocardial ischemia-reperfusion (I/R) injury. Mammalian target of rapamycin (mTOR), a downstream target of PI3K/AKT signaling, is necessary and sufficient to protect the heart from I/R injury. Inhaled anesthetic sevoflurane is widely used in cardiac surgeries because its induction and recovery are faster and smoother than other inhaled anesthetics. Sevoflurane proved capable of inducing postconditioning effects in the myocardium. However, the underlying molecular mechanisms for sevoflurane-induced postconditioning (SPC) were largely unclear. In the present study, we demonstrated that SPC protects myocardium from I/R injury with narrowed cardiac infarct focus, increased ATP content, and decreased cardiomyocyte apoptosis, which are mainly due to the activation of PI3K/AKT/mTOR signaling and the protection of mitochondrial energy metabolism. Application of dactolisib (BEZ235), a PI3K/mTOR dual inhibitor, abolishes the up-regulation of pho-AKT, pho-GSK, pho-mTOR, and pho-p70s6k induced by SPC, hence abrogating the anti-apoptotic effect of sevoflurane and reducing SPC-mediated protection of heart from I/R injury. As such, this study proved that PI3K/AKT/mTOR pathway plays an important role in SPC induced cardiac protection against I/R injury.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Chen Wang
- Department of Anesthesiology, the Second Affiliate Hospital of Soochow University, Suzhou, 215000, China
| | - Shuchun Yu
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Zhenzhong Luo
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Yong Chen
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Qin Liu
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Guohai Xu
- Department of Anesthesiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| | - Peng Yu
- Department of Cardiology, the Second Affiliate Hospital of Nanchang University, Nanchang, 330000, China
| |
Collapse
|
27
|
Cardio-protective signalling by glyceryl trinitrate and cariporide in a model of donor heart preservation. Heart Lung Circ 2014; 24:306-18. [PMID: 25459486 DOI: 10.1016/j.hlc.2014.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 09/30/2014] [Accepted: 10/05/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND Storage of donor hearts in cardioplegic solutions supplemented with agents that mimic the ischaemic preconditioning response enhanced their post-reperfusion function. The present study examines the minimisation of cell death and activation of pro-survival signalling directed towards maintenance of mitochondrial homeostasis in hearts arrested and stored in two such agents, glyceryl-trinitrate, a nitric oxide donor and cariporide, (a sodium-hydrogen exchange inhibitor). METHODS After baseline functional measurement, isolated working rat hearts were arrested and stored for 6h at 4°C in either Celsior(®), Celsior(®) containing 0.1mg/ml glyceryl-trinitrate, 10μM cariporide or both agents. After reperfusion, function was remeasured. Hearts were then processed for immunoblotting or histology. RESULTS Necrotic and apoptotic markers present in the Celsior(®) group post-reperfusion were abolished by glyceryl-trinitrate, cariporide or both. Increased phosphorylation of ERK and Bcl2, after reperfusion in groups stored in glyceryl-trinitrate, cariporide or both along with increased phospho-STAT3 levels in the glyceryl-trinitrate/cariporide group correlated with functional recovery. Inhibition of STAT3 phosphorylation blocked recovery. No phospho-Akt increase was seen in any treatment. CONCLUSIONS Activation of signalling pathways that favour mitophagy activation (ERK and Bcl2 phosphorylation) and maintenance of mitochondrial transition pore closure after reperfusion (STAT3 and ERK phosphorylation) were crucial for functional recovery of the donor heart.
Collapse
|
28
|
Morimoto RI, Cuervo AM. Proteostasis and the aging proteome in health and disease. J Gerontol A Biol Sci Med Sci 2014; 69 Suppl 1:S33-8. [PMID: 24833584 DOI: 10.1093/gerona/glu049] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The maintenance of the proteome is essential to preserve cell functionality and the ability to respond and adapt to the changing environment. This is regulated by the proteostasis network, a dedicated set of molecular components comprised of molecular chaperones and protein clearance mechanisms, regulated by cell stress signaling pathways, that prevents the toxicity associated with protein misfolding and accumulation of toxic aggregates in different subcellular compartments and tissues. The efficiency of the proteostasis network declines with age and this failure in protein homeostasis has been proposed to underlie the basis of common age-related human disorders. The current advances in the understanding of the mechanisms and regulation of proteostasis and of the different types of digressions in this process in aging have turned the attention toward the therapeutic opportunities offered by the restoration of proteostasis in age-associated degenerative diseases. Here, we discuss some of the unresolved questions on proteostasis that need to be addressed to enhance healthspan and to diminish the pathology associated with persistent protein damage.
Collapse
Affiliation(s)
- Richard I Morimoto
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois.
| | - Ana Maria Cuervo
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
29
|
Wang X, Yuan B, Dong W, Yang B, Yang Y, Lin X, Gong G. Induction of heat-shock protein 70 expression by geranylgeranylacetone shows cytoprotective effects in cardiomyocytes of mice under humid heat stress. PLoS One 2014; 9:e93536. [PMID: 24695789 PMCID: PMC3973581 DOI: 10.1371/journal.pone.0093536] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/04/2014] [Indexed: 11/23/2022] Open
Abstract
Background Increasing evidence has revealed that humid heat stress (HHS) causes considerable damage to human health. The cardiovascular system has been suggested to be the primary target of heat stress, which results in serious cardiovascular diseases. However, there is still a lack of effective approaches for the prevention and treatment of cardiovascular diseases induced by HHS. Objective Heat-shock proteins (Hsps), especially Hsp70, are reported to provide effective cytoprotection under various stress stimuli. In the present study, we evaluated the cytoprotective effect of geranylgeranylacetone (GGA), which was previously been reported to induce Hsp70 expression in cardiomyocytes under HHS. Methods and Principal Findings Using a mouse model of HHS, we showed that the pretreatment of GGA enhanced Hsp70 expression under HHS, as examined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. We then examined the effect of GGA pretreatment on the cardiomyocyte apoptosis induced by HHS using terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) staining, and found that GGA pretreatment inhibited mitochondria-mediated apoptosis. GGA pretreatment could reverse the effect of HHS on cell apoptosis by increasing expression of Bcl-2, decreasing cytochrome c in cytosol, and increasing cytochrome c in mitochondria. However, GGA pretreatment had no effect on the oxidative stress induced by HHS as determined by levels of superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH). Conclusion We have demonstrated that GGA pretreatment suppressed HHS-induced apoptosis of cardiomyocytes through the induction of Hsp70 overexpression.
Collapse
Affiliation(s)
- Xiaowu Wang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Binbin Yuan
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Wenpeng Dong
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Bo Yang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yongchao Yang
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Xi Lin
- Center of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Gu Gong
- Department of Anesthesiology, General Hospital of Chengdu Military Command, Chengdu, China
- * E-mail:
| |
Collapse
|
30
|
Zuo L, Li Q, Sun B, Xu Z, Ge Z. Cilostazol promotes mitochondrial biogenesis in human umbilical vein endothelial cells through activating the expression of PGC-1α. Biochem Biophys Res Commun 2013; 433:52-7. [PMID: 23485471 DOI: 10.1016/j.bbrc.2013.02.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 11/30/2022]
Abstract
Mitochondrial dysfunction is frequently observed in vascular diseases. Cilostazol is a drug approved by the US Food and Drug Administration for the treatment of intermittent claudication. Cilostazol increases intracellular cyclic adenosine monophosphate (cAMP) levels through inhibition of type III phosphodiesterase. The effects of cilostazol in mitochondrial biogenesis in human umbilical vein endothelial cells (HUVECs) were investigated in this study. Cilostazol treated HUVECs displayed increased levels of ATP, mitochondrial DNA/nuclear DNA ratio, expressions of cytochrome B, and mitochondrial mass, suggesting an enhanced mitochondrial biogenesis induced by cilostazol. The promoted mitochondrial biogenesis could be abolished by Protein kinase A (PKA) specific inhibitor H-89, implying that PKA pathway played a critical role in increased mitochondrial biogenesis after cilostazol treatment. Indeed, expression levels of peroxisome proliferator activator receptor gamma-coactivator 1α (PGC-1α), NRF 1 and mitochondrial transcription factor A (TFAM) were significantly increased in HUVECs after incubation with cilostazol at both mRNA levels and protein levels. Importantly, knockdown of PGC-1α could abolish cilostazol-induced mitochondrial biogenesis. Enhanced expression of p-CREB and PGC-1α induced by cilostazol could be inhibited by H-89. Moreover, the increased expression of PGC-1α induced by cilostazol could be inhibited by downregulation of CREB using CREB siRNA at both mRNA and protein levels. All the results indicated that cilostazol promoted mitochondrial biogenesis through activating the expression of PGC-1α in HUVECs, which was mediated by PKA/CREB pathway.
Collapse
Affiliation(s)
- Luning Zuo
- Shandong University, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | | | | | | | | |
Collapse
|
31
|
Hill BG, Benavides GA, Lancaster JR, Ballinger S, Dell’Italia L, Zhang J, Darley-Usmar VM. Integration of cellular bioenergetics with mitochondrial quality control and autophagy. Biol Chem 2012; 393:1485-1512. [PMID: 23092819 PMCID: PMC3594552 DOI: 10.1515/hsz-2012-0198] [Citation(s) in RCA: 360] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Accepted: 06/22/2012] [Indexed: 02/06/2023]
Abstract
Bioenergetic dysfunction is emerging as a cornerstone for establishing a framework for understanding the pathophysiology of cardiovascular disease, diabetes,cancer and neurodegeneration. Recent advances in cellular bioenergetics have shown that many cells maintain a substantial bioenergetic reserve capacity, which is a prospective index of ‘ healthy ’ mitochondrial populations.The bioenergetics of the cell are likely regulated by energy requirements and substrate availability. Additionally,the overall quality of the mitochondrial population and the relative abundance of mitochondria in cells and tissues also impinge on overall bioenergetic capacity and resistance to stress. Because mitochondria are susceptible to damage mediated by reactive oxygen/nitrogen and lipid species, maintaining a ‘ healthy ’ population of mitochondria through quality control mechanisms appears to be essential for cell survival under conditions of pathological stress. Accumulating evidence suggest that mitophagy is particularly important for preventing amplification of initial oxidative insults, which otherwise would further impair the respiratory chain or promote mutations in mitochondrial DNA (mtDNA). The processes underlying the regulation of mitophagy depend on several factors, including the integrity of mtDNA, electron transport chain activity, and the interaction and regulation of the autophagic machinery. The integration and interpretation of cellular bioenergetics in the context of mitochondrial quality control and genetics is the theme of this review.
Collapse
Affiliation(s)
- Bradford G. Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, and Department of Medicine, University of Louisville, Louisville, KY
- Departments of Biochemistry and Molecular Biology and Physiology and Biophysics, University of Louisville, Louisville, KY
| | - Gloria A. Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jack R. Lancaster
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lou Dell’Italia
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Victor M. Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
32
|
Kim JH, Lee DC. Mitochondrial DNA copy number in peripheral blood is associated with femoral neck bone mineral density in postmenopausal women. J Rheumatol 2012; 39:1465-72. [PMID: 22589267 DOI: 10.3899/jrheum.111444] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE It has been suggested that mitochondrial dysfunction is related to aging and metabolic disorders. Yet there are few studies of the relationship between bone mineral density (BMD) and mitochondrial content in humans. We investigated the relationship between BMD and mitochondrial DNA (mtDNA) copy number in peripheral blood of postmenopausal women. METHODS The study included 146 postmenopausal women. Enrolled subjects were taking no medications and had no disorders that altered bone metabolism. We measured BMD using dual-energy x-ray absorptiometry and leukocyte mtDNA copy number using real-time polymerase chain reaction. Anthropometric evaluations and biochemical tests were performed. RESULTS Patients with osteopenia or osteoporosis had lower mtDNA copy numbers than normal subjects (p < 0.0001). Femoral neck BMD was negatively correlated with age (r = -0.01, p = 0.04) and with serum levels of adiponectin (r = -0.22, p = 0.01) and osteocalcin (r = -0.31, p = 0.0001). Serum levels of 25-OH vitamin D (r = 0.32, p < 0.0001) and mtDNA copy number (r = 0.36, p < 0.0001) were positively correlated with femoral neck BMD. Multiple regression analysis showed that mtDNA copy number (ß = 0.156, p < 0.001) was an independent factor associated with femoral neck BMD after adjustment for age, body mass index, waist circumference, waist-hip ratio, blood pressure, homeostatic model assessment of insulin resistance, high-sensitivity C-reactive protein, adiponectin, osteocalcin, homocysteine, lipid profiles, 25-OH vitamin D, and regular exercise. mtDNA copy number was not related to lumbar BMD. CONCLUSION Low mtDNA content in peripheral blood is related to decreased femoral neck BMD in postmenopausal women. Our findings suggest that mitochondrial dysfunction may be a potential pathophysiologic mechanism of osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Jung-Ha Kim
- Department of Family Medicine, Chung-Ang University Healthcare Center, Korea
| | | |
Collapse
|
33
|
Vagnozzi RJ, Hoffman NE, Elrod JW, Madesh M, Force T. Protein Kinase Signaling at the Crossroads of Myocyte Life and Death in Ischemic Heart Disease. ACTA ACUST UNITED AC 2012; 9:e173-e182. [PMID: 24839450 DOI: 10.1016/j.ddstr.2013.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Myocardial ischemia results in death of cardiac myocytes via tightly-regulated and interconnected signaling pathways. Protein kinases play crucial roles in this regulation and are highly amenable to therapeutic intervention, making targeted inhibition an attractive strategy for ischemic heart disease. Recent studies have uncovered numerous kinases that participate in the cardiomyocyte response to ischemic injury, thus potentiating the development of new therapeutics. Moreover, many kinase signaling pathways converge at the mitochondria, a key participant in both cardiomyocyte physiology and the pathogenesis of ischemic heart disease. Herein we highlight kinase pathways regulating three major drivers of cell death: mitochondrial permeability transition pore opening (mPTP), programmed necrosis and Ca2+ overload-induced mitochondrial dysfunction. Inhibition of each of these kinase pathways has been proposed as a means to limit cardiomyocyte death from ischemia/reperfusion (I/R) injury.
Collapse
Affiliation(s)
- Ronald J Vagnozzi
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - Nicholas E Hoffman
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - John W Elrod
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - Muniswamy Madesh
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| | - Thomas Force
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3026, USA
| |
Collapse
|
34
|
Cuadrado I, Fernández-Velasco M, Boscá L, de las Heras B. Labdane diterpenes protect against anoxia/reperfusion injury in cardiomyocytes: involvement of AKT activation. Cell Death Dis 2011; 2:e229. [PMID: 22071634 PMCID: PMC3223697 DOI: 10.1038/cddis.2011.113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several labdane diterpenes exert anti-inflammatory and cytoprotective actions; therefore, we have investigated whether these molecules protect cardiomyocytes in an anoxia/reperfusion (A/R) model, establishing the molecular mechanisms involved in the process. The cardioprotective activity of three diterpenes (T1, T2 and T3) was studied in the H9c2 cell line and in isolated rat cardiomyocyte subjected to A/R injury. In both cases, treatment with diterpenes T1 and T2 protected from A/R-induced apoptosis, as deduced by a decrease in the percentage of apoptotic and caspase-3 active positive cells, a decrease in the Bcl-2/Bax ratio and an increase in the expression of antiapoptotic proteins. Analysis of cell survival signaling pathways showed that diterpenes T1 and T2 added after A/R increased phospho-AKT and phospho-ERK 1/2 levels. These cardioprotective effects were lost when AKT activity was pharmacologically inhibited. Moreover, the labdane-induced cardioprotection involves activation of AMPK, suggesting a role for energy homeostasis in their mechanism of action. Labdane diterpenes (T1 and T2) also exerted cardioprotective effects against A/R-induced injury in isolated cardiomyocytes and the mechanisms involved activation of specific survival signals (PI3K/AKT pathways, ERK1/2 and AMPK) and inhibition of apoptosis.
Collapse
Affiliation(s)
- I Cuadrado
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - M Fernández-Velasco
- Instituto de Investigaciones Biomédicas ‘Alberto Sols' (CSIC-UAM), Madrid, Spain
| | - L Boscá
- Instituto de Investigaciones Biomédicas ‘Alberto Sols' (CSIC-UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas ‘Alberto Sols' (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain. Tel: +34 9149 72747; Fax: +34 9158 54401; E-mail:
| | - B de las Heras
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain. Tel: +34 9139 42276; Fax: +34 9139 41726; E-mail:
| |
Collapse
|