1
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
2
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Germanova E, Khmil N, Pavlik L, Mikheeva I, Mironova G, Lukyanova L. The Role of Mitochondrial Enzymes, Succinate-Coupled Signaling Pathways and Mitochondrial Ultrastructure in the Formation of Urgent Adaptation to Acute Hypoxia in the Myocardium. Int J Mol Sci 2022; 23:ijms232214248. [PMID: 36430733 PMCID: PMC9696391 DOI: 10.3390/ijms232214248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
The effect of a single one-hour exposure to three modes of hypobaric hypoxia (HBH) differed in the content of O2 in inhaled air (FiO2-14%, 10%, 8%) in the development of mitochondrial-dependent adaptive processes in the myocardium was studied in vivo. The following parameters have been examined: (a) an urgent reaction of catalytic subunits of mitochondrial enzymes (NDUFV2, SDHA, Cyt b, COX2, ATP5A) in the myocardium as an indicator of the state of the respiratory chain electron transport function; (b) an urgent activation of signaling pathways dependent on GPR91, HIF-1α and VEGF, allowing us to assess their role in the formation of urgent mechanisms of adaptation to hypoxia in the myocardium; (c) changes in the ultrastructure of three subpopulations of myocardial mitochondria under these conditions. The studies were conducted on two rat phenotypes: rats with low resistance (LR) and high resistance (HR) to hypoxia. The adaptive and compensatory role of the mitochondrial complex II (MC II) in maintaining the electron transport and energy function of the myocardium in a wide range of reduced O2 concentrations in the initial period of hypoxic exposure has been established. The features of urgent reciprocal regulatory interaction of NAD- and FAD-dependent oxidation pathways in myocardial mitochondria under these conditions have been revealed. The data indicating the participation of GPR91, HIF-1a and VEGF in this process have been obtained. The ultrastructure of the mitochondrial subpopulations in the myocardium of LR and HR rats differed in normoxic conditions and reacted differently to hypoxia of varying severity. The parameters studied together are highly informative indicators of the quality of cardiac activity and metabolic biomarkers of urgent adaptation in various hypoxic conditions.
Collapse
Affiliation(s)
- Elita Germanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
| | - Natalya Khmil
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Lyubov Pavlik
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Galina Mironova
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
- Correspondence: (G.M.); (L.L.)
| | - Ludmila Lukyanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
- Correspondence: (G.M.); (L.L.)
| |
Collapse
|
4
|
Mikheeva I, Mikhailova G, Zhujkova N, Shtanchaev R, Arkhipov V, Pavlik L. Studying the structure of the nucleus of the trochlear nerve in mice through 7 days of readaptation to earth gravity after spaceflight. Brain Res 2022; 1795:148077. [PMID: 36096199 DOI: 10.1016/j.brainres.2022.148077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022]
Abstract
The negative effect of hypogravity on the human organism is manifested to a greater extent after the astronauts return to the conditions of habitual gravity. In this work, to elucidate the causes underlying atypical nystagmus, arising after the flight, we studied structural changes in the motoneurons of the trochlear nerve after a 7-day readaptation of mice to the conditions of Earth's gravity. It is known, that motoneurons of the trochlear nerve innervate the muscle that controls the movement of the eyes in the vertical direction. We showed that the number of axodendritic synapses and some other morphological parameters of motoneurons changed by microgravity can return to their original state in 7 days. However, according to some parameters, motoneurons retain a "memory" of the action of microgravity and do not completely restore the structure. The volume of the soma, the shape of the nuclei, the number and orientation of dendrites do not return to pre-flight parameters. The number of dendrites after 7 days of adaptation remained increased, and the proportion of dendrites in the ventrolateral direction became 2.5 times greater than in motoneurons after space flight. The increased number of mitochondria after space flight became even more significant after readaptation. Microgravity-induced plastic changes retain to some extent "memory traces" after readaptation to Earth's gravity. It can be assumed that the restoration of the function of the trochlear nuclei (overcoming nystagmus) is carried out not only by reversible restoration of the structure of neurons, but partially using those mechanisms that are formed in weightlessness.
Collapse
Affiliation(s)
- Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia.
| | - Gulnara Mikhailova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Natalya Zhujkova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Rashid Shtanchaev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Vladimir Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Lyubov Pavlik
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| |
Collapse
|
5
|
Li D, Pi W, Sun Z, Liu X, Jiang J. Ferroptosis and its role in cardiomyopathy. Biomed Pharmacother 2022; 153:113279. [PMID: 35738177 DOI: 10.1016/j.biopha.2022.113279] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/09/2022] Open
Abstract
Heart disease is the leading cause of death worldwide. Cardiomyopathy is a disease characterized by the heart muscle damage, resulting heart in a structurally and functionally change, as well as heart failure and sudden cardiac death. The key pathogenic factor of cardiomyopathy is the loss of cardiomyocytes, but the related molecular mechanisms remain unclear. Ferroptosis is a newly discovered regulated form of cell death, characterized by iron accumulation and lipid peroxidation during cell death. Recent studies have shown that ferroptosis plays an important regulatory roles in the occurrence and development of many heart diseases such as myocardial ischemia/reperfusion injury, cardiomyopathy and heart failure. However, the systemic association of ferroptosis and cardiomyopathy remains largely unknown and needs to be elucidated. In this review, we provide an overview of the molecular mechanisms of ferroptosis and its role in individual cardiomyopathies, highlight that targeting ferroptosis maybe a potential therapeutic strategy for cardiomyopathy therapy in the future.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| |
Collapse
|
6
|
Li XL, Wang WG, Li MX, Liu TL, Tian XY, Wu L. Effects of Altitude and Duration of Differing Levels of Hypoxic Exposure on Hypoxia-Inducible Factor-1α in Rat Tissues. High Alt Med Biol 2022; 23:173-184. [PMID: 35708531 DOI: 10.1089/ham.2021.0100] [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: 11/12/2022] Open
Abstract
Li, Xiao-lin, Wei-gang Wang, Mao-xing Li, Tian-long Liu, Xiu-yu Tian, and Lan Wu. Effects of altitude and duration of differing levels of hypoxic exposure on hypoxia-inducible factor-1α in rat tissues. High Alt Med Biol. 23:173-184, 2022. Objective: This research aimed to evaluate the effects of hypoxia at different altitudes and durations on the expression of hypoxia-inducible factor-1α (HIF-1α) in rat tissues. Methods: A total of 72 Wistar rats were used to investigate the effect of hypoxia at different durations on rat tissues and 72 Wistar rats were used to investigate the effect of hypoxia at different altitudes. Hematoxylin and Eosin (HE) staining was performed to observe the pathological changes of hippocampus tissues, and the expression of HIF-1α of rats under conditions of hypoxia was detected by quantitative real-time polymerase chain reaction and western blotting. Results: According to the pathological results, we found that the degree of the brain, lung, and heart damage and HIF-1α expression, showed an increasing trend as the altitude (1,500, 3,000, 4,500, 6,000, 7,500, and 8,000 m for 12 hours) and duration (0, 6, 12, 24, 36, and 72 hours at 7,500 m) of hypoxia increased. Although there is a significant difference at 8,000 m, considering model stability, animal ethics and cost, we chose 7,500 m as a fixed altitude during hypoxia at different durations. Compared with the normoxic group, the expression of HIF-1α mRNA in the 7,500 m significantly increased by 30.48%, 21.00%, and 12.62%, in brain, lung, and heart tissue (p < 0.01), and HIF-1α mRNA in the 72-hour hypoxic exposure group significantly increased by 52.58%, 20.39%, 27.88% in tissues (p < 0.05). Compared with the normoxic group, HIF-1α protein expressions in the 7,500 m significantly increased by 10.26%, 31.71%, and 13.33% in brain, lung, and heart tissue (p < 0.01, p < 0.01, p < 0.05), and HIF-1α protein expressions in the 72-hour hypoxic exposure group significantly increased by 18.89%, 22.89%, and 29.75% in tissues (p < 0.05). Conclusion: HIF-1α expression in the rat was correlated with altitude and duration of hypoxic exposure.
Collapse
Affiliation(s)
- Xiao-Lin Li
- Department of Clinical Pharmacy, The 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China.,Gansu Plateau Pharmaceutical Technology Center, Lanzhou, China
| | - Wei-Gang Wang
- Department of Clinical Pharmacy, The 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China.,Gansu Plateau Pharmaceutical Technology Center, Lanzhou, China
| | - Mao-Xing Li
- Department of Clinical Pharmacy, The 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China.,Gansu Plateau Pharmaceutical Technology Center, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China.,Institute of Chemical Technology, Northwest Minzu University, Lanzhou, China
| | - Tian-Long Liu
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China.,Gansu Plateau Pharmaceutical Technology Center, Lanzhou, China
| | - Xiu-Yu Tian
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China.,Gansu Plateau Pharmaceutical Technology Center, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Lan Wu
- Institute of Chemical Technology, Northwest Minzu University, Lanzhou, China
| |
Collapse
|
7
|
Xin K, Hu B, Han L, Yu Q. Study on the HIF-1α regulated by glycolytic pathways and mitochondrial function in yaks of different altitudes during postmortem aging. J Food Biochem 2022; 46:e14205. [PMID: 35502450 DOI: 10.1111/jfbc.14205] [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: 12/09/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/24/2022]
Abstract
The study investigated the glycolysis pathway mediated by hypoxia-inducible factor-1α (HIF-1α) and the mechanism of its regulation. The results indicated that HIF-1α expression initially increased before subsequently decreasing with aging time during postmortem (p < .01). Glucose transporter-1 (GLUT-1), lactate dehydrogenase (LDH), and hexokinase (HK) displayed a similar trend with aging time (p < .01) while pyruvate dehydrogenase kinase 1 (PDK-1) increased gradually within the first 12 hr before decreasing at 24-120 hr. However, after treatment with a HIF-1α inhibitor, no significant differences were observed in the mitochondrial morphology. Furthermore, lactate content decreased, along with LDH, HK, and F0F1-ATP activities as well as GLUT-1 and PDK-1 expression (p < .01). The shear force for all groups also increased during postmortem aging (p < .01), with that of the controls being significantly higher compared with the treatment groups (p < .01). These findings confirmed that, after slaughter, the hypoxic environment within the muscles provided essential conditions for HIF-1α expression, which, in turn, activated the glycolysis pathway by mediating changes in the activities of glycolytic enzymes and mitochondrial function. Moreover, in accelerating glycolysis rate, the expression of HIF-1α further played a negative role in meat tenderization during postmortem aging. This, it was concluded that HIF-1α expression plays a significant role in postmortem yak meat tenderization by regulating the glycolysis pathway. PRATICAL APPLICATIONS: While converting muscle into meat through hypoxic glycolysis during postmortem aging is undeniable, the biochemical mechanism of this process mediated remains quite obscure. However, the meat quality difference which impact muscle regulation mechanism during postmortem aging has not been reported. The study investigated the HIF-1α played a major role in both the glycolytic pathway and as well as meat tenderness during the postmortem aging of yak meat. The glycolysis pathway is mediated by hypoxia-inducible factor-1α (HIF-1α), the mechanism of its regulation, and meat tenderness during the postmortem aging of yak meat.
Collapse
Affiliation(s)
- Keqi Xin
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Bo Hu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Ling Han
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
8
|
Deng X, Yang P, Gao T, Liu M, Li X. Allicin attenuates myocardial apoptosis, inflammation and mitochondrial injury during hypoxia-reoxygenation: an in vitro study. BMC Cardiovasc Disord 2021; 21:200. [PMID: 33882833 PMCID: PMC8059159 DOI: 10.1186/s12872-021-01918-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/11/2021] [Indexed: 01/19/2023] Open
Abstract
Background Myocardial ischemia–reperfusion (IR) injury is a damage due to an initial reduction in blood flow to the heart, preventing it from receiving enough oxygen, and subsequent restoration of blood flow through the opening of an occluded coronary artery producing paradoxical harmful effects. The finding of new therapies to prevent IR is of utmost importance. Allicin is a compound isolated from garlic having the ability to prevent and cure different diseases, and a protective effect on the myocardium was also demonstrated. Therefore, the aim of this study was to evaluate the in vitro protective effect of Allicin against myocardial IR injury on cardiomyocytes. Methods We established an in vitro hypoxia-reoxygenation (HR) model of primary porcine cardiomyocytes to simulate myocardial IR injury. Primary porcine cardiomyocytes were extracted from Mini-musk swines (1 day old). After a period of adaptation of at least 2–3 days, cardiomyocytes in good condition were selected and randomly divided into control group (normal oxygen for 5 h), HR group (2 h of hypoxia/3 h of reoxygenation), and HR + Allicin group (hypoxia/reoxygenation + Allicin treatment). Results After the induction of hypoxia/reoxygenation, Allicin treatment enhanced the cell viability. Moreover, Allicin treatment resulted in a reduction of apoptosis from 13.5 ± 1.2% to 6.11 ± 0.15% compared with the HR group (p < 0.05), and the apoptosis related proteins were regulated as well, with a decreased expression of Bax, cleaved caspase-3 and cytosolic cytochrome C and an increase in Bcl-2 expression in the HR + Allicin group (all p < 0.01). Pro-inflammatory cytokines, such as interleukin-6 and tumor necrosis factor alpha were down-regulated by the treatment with Allicin (both p < 0.01). In addition, it significantly decreased intracellular reactive oxygen species generation (p < 0.01) and reduced the loss of mitochondrial membrane potential (p < 0.01). Furthermore, the expression of PPARγ coactivator-1α and endothelial nitric oxide synthase was up-regulated (both p < 0.01), while the expression of Endothelin-1, hypoxia inducing factor-1α and transforming growth factor beta was down-regulated (all p < 0.01) by Allicin treatment. Conclusions These results suggested that Allicin protected the cardiomyocytes against HR damage by reducing apoptosis, inflammation and mitochondrial injury, thus providing a basis for its potential use in the treatment of myocardial IR. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-01918-6.
Collapse
Affiliation(s)
- Xinyi Deng
- Peking University China-Japan Friendship School of Clinical Medicine, 2 East Yinghuayuan Street, Hepingli, Beijing, 100029, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Peng Yang
- Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Tong Gao
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Mengru Liu
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Xianlun Li
- Peking University China-Japan Friendship School of Clinical Medicine, 2 East Yinghuayuan Street, Hepingli, Beijing, 100029, China. .,Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China. .,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
9
|
Zhang Q, Guo D, Wang Y, Wang X, Wang Q, Wu Y, Li C, Wang W, Wang Y. Danqi Pill Protects Against Heart Failure Post-Acute Myocardial Infarction via HIF-1α/PGC-1α Mediated Glucose Metabolism Pathway. Front Pharmacol 2020; 11:458. [PMID: 32372956 PMCID: PMC7187888 DOI: 10.3389/fphar.2020.00458] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/24/2020] [Indexed: 12/28/2022] Open
Abstract
AIM Heart failure (HF) post-acute myocardial infarction (AMI) leads to a large number of hospitalizations and deaths worldwide. Danqi pill (DQP) is included in the 2015 national pharmacopoeia and widely applied in the treatment of HF in clinics in China. We examined whether DQP acted on glucose metabolism to protect against HF post-AMI via hypoxia inducible factor-1 alpha (HIF-1α)/peroxisome proliferator-activated receptor α co-activator (PGC-1α) pathway. METHODS AND RESULTS In this study, left anterior descending (LAD) artery ligation induced HF post-AMI rats and oxygen-glucose deprivation-reperfusion (OGD/R)-induced H9C2 cell model were structured to explore the efficacy and mechanism of DQP. Here we showed that DQP protected the heart against ischemic damage as evidenced by improved cardiac functions and attenuated inflammatory infiltration. The expressions of critical proteins involved in glucose intake and transportation such as GLUT4 and PKM2 were up-regulated, while negative regulatory proteins involved in oxidative phosphorylation were attenuated in the treatment of DQP. Moreover, DQP up-regulated NRF1 and TFAM, promoted mitochondrial biogenesis and increased myocardial adenosine triphosphate (ATP) level. The protection effects of DQP were significantly compromised by HIF-1α siRNA, suggesting that HIF-1α signaling pathway was the potential target of DQP on HF post-AMI. CONCLUSIONS DQP exhibits the efficacy to improve myocardial glucose metabolism, mitochondrial oxidative phosphorylation and biogenesis by regulating HIF-1α/PGC-1α signaling pathway in HF post-AMI rats.
Collapse
Affiliation(s)
- Qian Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongqing Guo
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Wang
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qiyan Wang
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Wu
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Zhao D, Zheng H, Greasley A, Ling F, Zhou Q, Wang B, Ni T, Topiwala I, Zhu C, Mele T, Liu K, Zheng X. The role of miR-711 in cardiac cells in response to oxidative stress and its biogenesis: a study on H9C2 cells. Cell Mol Biol Lett 2020; 25:26. [PMID: 32308692 PMCID: PMC7146913 DOI: 10.1186/s11658-020-00206-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Oxidative stress results in cell apoptosis/death and plays a detrimental role in disease development and progression. Stressors alter the miRNA expression profile and miRNAs play a role in the cell response to stress. We previously showed that miR-711 is significantly over-expressed in extended cold ischemia reperfusion injured hearts in heart transplant. In this study, we aimed to investigate the role of miR-711 in cardiac cell damage in response to oxidative stress and how miR-711 is regulated. Methods Rat cardiac cell line H9c2 cells were cultured and exposed to oxidative conditions (Antimycin A (AA), H2O2, CoCl2, or cold hypoxia/reoxygenation (H/R)) in vitro. H9c2 cells were transfected with miR-711 mimics, miR-711 inhibitors, or small interference RNA, using transfection reagents. The expression of miR-711 was measured by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Cell apoptosis/death was detected by flow cytometry and an IncuCyte system. Mitochondrial damage was detected by measuring the mitochondria membrane potential by flow cytometry. Gene expression was detected by qRT-PCR at the mRNA level and Western blotting and immunocytochemistry staining at the protein level. Results We found that miR-711 was significantly up-regulated in cells treated with H2O2, AA, CoCl2, and cold H/R. Over-expression of miR-711 increased cell apoptosis/death induced by AA and H/R whereas cell death was reduced by miR-711 inhibitors. MiR-711 induced cell death through negative regulation of angiopoietin 1 (Ang-1), fibroblast growth factor 14 (FGF14) and calcium voltage-gated channel subunit alpha1C (Cacna1c) genes. Both knockdown of hypoxia inducible factor 1α (HIF-1α) and inactivation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFКB) pathway inhibited over-expression of miR-711. Conclusion Oxidative stress increases the expression of miR-711. Over-expression of miR-711 induces cell apoptosis/death. HIF-1α and NFКB regulate miR-711 in H9c2 cells during oxidative stress. miR-711 is a new target for preventing oxidative stress.
Collapse
Affiliation(s)
- Duo Zhao
- 1Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, 130041 China.,2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada.,3Department of Cardiovascular Surgery, The First People's Hospital of Foshan, Foshan, Guangdong China
| | - Hao Zheng
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Adam Greasley
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Fengjun Ling
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Qinfeng Zhou
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada.,Department of Laboratory Medicine, Zhangjiagang TCM Hospital Affiliated to Nanking University of Chinese Medicine, Zhangjiagang, Jiangsu China
| | - Bowen Wang
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Tiffany Ni
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Ishita Topiwala
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Cuilin Zhu
- 1Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, 130041 China.,2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada
| | - Tina Mele
- 5Department of Surgery, Western University, Ontario, London Canada
| | - Kexiang Liu
- 1Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, 130041 China
| | - Xiufen Zheng
- 2Department of Pathology and Laboratory Medicine, Western University, London, Ontario Canada.,5Department of Surgery, Western University, Ontario, London Canada.,6London Health Sciences Centre, London, Ontario Canada.,7Department of Oncology, Western University, Ontario, London Canada.,8Lawson Health Research Institute, Ontario, London Canada
| |
Collapse
|
11
|
Rabinovich-Nikitin I, Kirshenbaum LA. Hypoxia-inducible factor 1 regulates SERCA2 in the heart by modulating miR-29c levels. Am J Physiol Heart Circ Physiol 2019; 316:H1211-H1213. [PMID: 30848679 DOI: 10.1152/ajpheart.00019.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Inna Rabinovich-Nikitin
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Lorrie A Kirshenbaum
- Institute of Cardiovascular Sciences, Saint Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba , Winnipeg, Manitoba , Canada.,Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba , Canada R2H 2H6
| |
Collapse
|
12
|
Sousa Fialho MDL, Abd Jamil AH, Stannard GA, Heather LC. Hypoxia-inducible factor 1 signalling, metabolism and its therapeutic potential in cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:831-843. [DOI: 10.1016/j.bbadis.2018.09.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/24/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022]
|
13
|
Yao HC, Zhou M, Zhou YH, Wang LH, Zhang DY, Han QF, Liu T, Wu L, Tian KL, Zhang M. Intravenous high mobility group box 1 upregulates the expression of HIF-1α in the myocardium via a protein kinase B-dependent pathway in rats following acute myocardial ischemia. Mol Med Rep 2015; 13:1211-9. [PMID: 26648172 PMCID: PMC4732844 DOI: 10.3892/mmr.2015.4648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/10/2015] [Indexed: 12/23/2022] Open
Abstract
The effects of intravenous high mobility group box 1 (HMGB1) on myocardial ischemia/reperfusion (I/R) injury remains to be elucidated. The purpose of the present study was to investigate the effects of intravenous HMGB1 on the expression of hypoxia inducible factor-1α (HIF-1α) in the myocardium of rats following acute myocardial ischemia, and to examine the effects of intravenous HMGB1 on myocardial I/R injury. Male Wistar rats were divided into the following groups: Sham operation group (n=10), a group exposed to ischemia for 30 min and reperfusion for 4 h (I/R group) as a control (n=10), an HMGB group, in which 100 ng/kg HMGB was administered intravenously 30 min prior to ischemia (n=10), an LY group, in whic LY294002, an inhibitor of phosphoinositide 3-kinase (PI3K), was administered intravenously (0.3 mg/kg) 40 min prior to ischemia (n=10), and the HMGB1+LY group, in which HMGB1 (100 ng/kg) and LY294002 (0.3 mg/kg) were administered intravenously 30 min and 40 min prior to ischemia, respectively (n=10). The serum levels of cardiac troponin I (cTnI) and tumor necrosis factor-α (TNF-α), and myocardial infarct size were measured. The expression levels of phosphorylated Akt and HIF-1α were investigated using western blot analyses. The results showed that pre-treatment with HMGB1 significantly decreased serum levels of cTnI, and TNF-α, and reduced myocardial infarct size following 4 h reperfusion (all P<0.05). HMGB1 also increased the expression levels of HIF-1α and p-Akt induced by I/R (P<0.05). LY294002 was found to eliminate the effects of intravenous HMGB1 on myocardial I/R injury (P<0.05). These results suggest that intravenous pre-treatment with HMGB1 may exert its cardioprotective effects via the upregulation of the myocardial expression of HIF-1α, which may be regulated by the PI3K/Akt signaling pathway, in rats following acute myocardial I/R.
Collapse
Affiliation(s)
- Heng-Chen Yao
- Department of Cardiology, Qilu Hospital of Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Min Zhou
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Yan-Hong Zhou
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lan-Hua Wang
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - De-Yong Zhang
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Qian-Feng Han
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Tao Liu
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Lei Wu
- Department of Cardiology, Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Ke-Li Tian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Mei Zhang
- Department of Cardiology, Qilu Hospital of Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
14
|
Yamagami K, Oka T, Wang Q, Ishizu T, Lee JK, Miwa K, Akazawa H, Naito AT, Sakata Y, Komuro I. Pirfenidone exhibits cardioprotective effects by regulating myocardial fibrosis and vascular permeability in pressure-overloaded hearts. Am J Physiol Heart Circ Physiol 2015; 309:H512-22. [DOI: 10.1152/ajpheart.00137.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/28/2015] [Indexed: 12/12/2022]
Abstract
Although cardiac fibrosis causes heart failure, its molecular mechanisms remain elusive. In this study, we investigated the mechanisms of cardiac fibrosis and examined the effects of the antifibrotic drug pirfenidone (PFD) on chronic heart failure. To understand the responsible mechanisms, we generated an in vivo pressure-overloaded heart failure model via transverse aortic constriction (TAC) and examined the effects of PFD on chronic-phase cardiac fibrosis and function. In the vehicle group, contractile dysfunction and left ventricle fibrosis progressed further from 4 to 8 wk after TAC but were prevented by PFD treatment beginning 4 wk after TAC. We isolated cardiac fibroblasts and vascular endothelial cells from the left ventricles of adult male mice and investigated the cell-type-specific effects of PFD. Transforming growth factor-β induced upregulated collagen 1 expression via p38 phosphorylation and downregulated claudin 5 (Cldn5) expression in cardiac fibroblasts and endothelial cells, respectively; both processes were inhibited by PFD. Moreover, PFD inhibited changes in the collagen 1 and Cldn5 expression levels, resulting in reduced fibrosis and serum albumin leakage into the interstitial space during the chronic phase in TAC hearts. In conclusion, PFD inhibited cardiac fibrosis by suppressing both collagen expression and the increased vascular permeability induced by pressure overload.
Collapse
Affiliation(s)
- Kiyoshi Yamagami
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toru Oka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Qi Wang
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takamaru Ishizu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Keiko Miwa
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Atsuhiko T. Naito
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
15
|
Lu X, Bi YW, Chen KB. Olmesartan restores the protective effect of remote ischemic perconditioning against myocardial ischemia/reperfusion injury in spontaneously hypertensive rats. Clinics (Sao Paulo) 2015; 70. [PMID: 26222820 PMCID: PMC4496757 DOI: 10.6061/clinics/2015(07)07] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Remote ischemic perconditioning is the newest technique used to lessen ischemia/reperfusion injury. However, its effect in hypertensive animals has not been investigated. This study aimed to examine the effect of remote ischemic perconditioning in spontaneously hypertensive rats and determine whether chronic treatment with Olmesartan could influence the effect of remote ischemic perconditioning. METHODS Sixty rats were randomly divided into six groups: vehicle-sham, vehicle-ischemia/reperfusion injury, vehicle-remote ischemic perconditioning, olmesartan-sham, olmesartan-ischemia/reperfusion and olmesartan-remote ischemic perconditioning. The left ventricular mass index, creatine kinase concentration, infarct size, arrhythmia scores, HIF-1α mRNA expression, miR-21 expression and miR-210 expression were measured. RESULTS Olmesartan significantly reduced the left ventricular mass index, decreased the creatine kinase concentration, limited the infarct size and reduced the arrhythmia score. The infarct size, creatine kinase concentration and arrhythmia score during reperfusion were similar for the vehicle-ischemia/reperfusion group and vehicle-remote ischemic perconditioning group. However, these values were significantly decreased in the olmesartan-remote ischemic perconditioning group compared to the olmesartan-ischemia/reperfusion injury group. HIF-1α, miR-21 and miR-210 expression were markedly down-regulated in the Olmesartan-sham group compared to the vehicle-sham group and significantly up-regulated in the olmesartan-remote ischemic perconditioning group compared to the olmesartan-ischemia/reperfusion injury group. CONCLUSION The results indicate that (1) the protective effect of remote ischemic perconditioning is lost in vehicle-treated rats and that chronic treatment with Olmesartan restores the protective effect of remote ischemic perconditioning; (2) chronic treatment with Olmesartan down-regulates HIF-1α, miR-21 and miR-210 expression and reduces hypertrophy, thereby limiting ischemia/reperfusion injury; and (3) recovery of the protective effect of remote ischemic perconditioning is related to the up-regulation of HIF-1α, miR-21 and miR-210 expression.
Collapse
Affiliation(s)
- Xin Lu
- Shandong University, Qilu Hospital, Department of Cardiovascular Surgery, Jinan, China
| | - Yan-Wen Bi
- Shandong University, Qilu Hospital, Department of Cardiovascular Surgery, Jinan, China
- Corresponding Author: E-mail:
| | - Ke-Biao Chen
- Taian City Central Hospital, Department of Cardiovascular Surgery, Taian, China
| |
Collapse
|
16
|
Galectin-3 is expressed in the myocardium very early post–myocardial infarction. Cardiovasc Pathol 2015; 24:213-23. [DOI: 10.1016/j.carpath.2014.12.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 12/17/2022] Open
|
17
|
Rose AH, Hoffmann PR. Selenoproteins and cardiovascular stress. Thromb Haemost 2014; 113:494-504. [PMID: 25354851 DOI: 10.1160/th14-07-0603] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/18/2014] [Indexed: 02/07/2023]
Abstract
Dietary selenium (Se) is an essential micronutrient that exerts its biological effects through its incorporation into selenoproteins. This family of proteins contains several antioxidant enzymes such as the glutathione peroxidases, redox-regulating enzymes such as thioredoxin reductases, a methionine sulfoxide reductase, and others. In this review, we summarise the current understanding of the roles these selenoproteins play in protecting the cardiovascular system from different types of stress including ischaemia-reperfusion, homocysteine dysregulation, myocardial hypertrophy, doxirubicin toxicity, Keshan disease, and others.
Collapse
Affiliation(s)
| | - Peter R Hoffmann
- Peter R. Hoffmann, University of Hawaii, John A. Burns School of Medicine, 651 Ilalo Street, Honolulu, HI 96813, USA, Fax: +1 808 692 1968, E-mail:
| |
Collapse
|
18
|
Gao W, Ferguson G, Connell P, Walshe T, O'Brien C, Redmond EM, Cahill PA. Glucose attenuates hypoxia-induced changes in endothelial cell growth by inhibiting HIF-1α expression. Diab Vasc Dis Res 2014; 11:270-280. [PMID: 24853909 DOI: 10.1177/1479164114533356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Hyperglycaemia and hypoxia play essential pathophysiological roles in diabetes. We determined whether hyperglycaemia influences endothelial cell growth under hypoxic conditions in vitro. Using a Ruskinn Invivo2 400 Hypoxia Workstation, bovine aortic endothelial cells (BAEC) were exposed to high glucose concentrations (25 mM glucose) under normoxic or hypoxic conditions before cell growth (balance of proliferation and apoptosis) was assessed by fluorescence-activated cell sorting (FACS) analysis, proliferating cell nuclear antigen (pCNA), Bcl-xL and caspase-3 protein expression and activity. Hypoxia increased hypoxia response element (HRE) transactivation and induced hypoxia-inducible factor-1α (HIF-1α) expression when compared to normoxic controls concomitant with a significant decrease in cell growth. High glucose (25 mM) concentrations attenuated HRE transactivation and HIF-1α protein expression while concurrently reducing hypoxia-induced changes in BAEC growth. Knockdown of HIF-1α expression significantly decreased hypoxia-induced changes in growth and attenuated the modulatory effects of glucose. These results provide evidence that hypoxia-induced control of BAEC growth can be altered by the presence of glucose via inhibition of HIF-1α expression and activation.
Collapse
Affiliation(s)
- Wei Gao
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Gail Ferguson
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Paul Connell
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland Mater Misericordiae Hospital, Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Tony Walshe
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Colm O'Brien
- Mater Misericordiae Hospital, Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, USA
| | - Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
19
|
Catrina SB. Impaired hypoxia-inducible factor (HIF) regulation by hyperglycemia. J Mol Med (Berl) 2014; 92:1025-34. [PMID: 25027070 DOI: 10.1007/s00109-014-1166-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 05/06/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
The mechanisms that contribute to the development of diabetes complications remain unclear. A defective reaction of tissues to hypoxia has recently emerged as a new pathogenic mechanism and consists of a complex repression of hypoxia-inducible factor (HIF), which is the main regulator of the adaptive response to hypoxia. This paper discusses the mechanisms by which hyperglycaemia contributes to HIF repression in diabetes. Furthermore, a comprehensive analysis of the functional relevance of these new findings to the development of chronic diabetes complications is provided, along with examples from animal models and clinics.
Collapse
Affiliation(s)
- Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital, L1:01, 17176, Stockholm, Sweden,
| |
Collapse
|
20
|
Gallo S, Gatti S, Sala V, Albano R, Costelli P, Casanova E, Comoglio PM, Crepaldi T. Agonist antibodies activating the Met receptor protect cardiomyoblasts from cobalt chloride-induced apoptosis and autophagy. Cell Death Dis 2014; 5:e1185. [PMID: 24743740 PMCID: PMC4001309 DOI: 10.1038/cddis.2014.155] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/31/2022]
Abstract
Met, the tyrosine kinase receptor for hepatocyte growth factor (HGF), mainly activates prosurvival pathways, including protection from apoptosis. In this work, we investigated the cardioprotective mechanisms of Met activation by agonist monoclonal antibodies (mAbs). Cobalt chloride (CoCl2), a chemical mimetic of hypoxia, was used to induce cardiac damage in H9c2 cardiomyoblasts, which resulted in reduction of cell viability by (i) caspase-dependent apoptosis and (ii) – surprisingly – autophagy. Blocking either apoptosis with the caspase inhibitor benzyloxycarbonyl-VAD-fluoromethylketone or autophagosome formation with 3-methyladenine prevented loss of cell viability, which suggests that both processes contribute to cardiomyoblast injury. Concomitant treatment with Met-activating antibodies or HGF prevented apoptosis and autophagy. Pro-autophagic Redd1, Bnip3 and phospho-AMPK proteins, which are known to promote autophagy through inactivation of the mTOR pathway, were induced by CoCl2. Mechanistically, Met agonist antibodies or HGF prevented the inhibition of mTOR and reduced the flux of autophagosome formation. Accordingly, their anti-autophagic function was completely blunted by Temsirolimus, a specific mTOR inhibitor. Targeted Met activation was successful also in the setting of low oxygen conditions, in which Met agonist antibodies or HGF demonstrated anti-apoptotic and anti-autophagic effects. Activation of the Met pathway is thus a promising novel therapeutic tool for ischaemic injury.
Collapse
Affiliation(s)
- S Gallo
- Department of Oncology, University of Turin, Turin, Italy
| | - S Gatti
- Department of Oncology, University of Turin, Turin, Italy
| | - V Sala
- Department of Oncology, University of Turin, Turin, Italy
| | - R Albano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - P Costelli
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - E Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - P M Comoglio
- 1] Department of Oncology, University of Turin, Turin, Italy [2] Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - T Crepaldi
- Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Al-Salam S, Hashmi S. Galectin-1 in early acute myocardial infarction. PLoS One 2014; 9:e86994. [PMID: 24498007 PMCID: PMC3909026 DOI: 10.1371/journal.pone.0086994] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/17/2013] [Indexed: 02/02/2023] Open
Abstract
Myocardial infarction (MI) is the most serious manifestation of coronary artery disease and the cause of significant mortality and morbidity worldwide. Galectin-1(GAL-1), a divalent 14.5-kDa protein, is present both inside and outside cells, and has both intracellular and extracellular functions. Hypoxia inducible factor-1 alpha (HIF-1α) is a transcription factor mediating early and late responses to myocardial ischemia. Identification of the pattern of expression of GAL-1 and HIF-1α in the heart during the first 24 hours following acute MI will help in understanding early molecular changes in this event and may provide methods to overcome serious complications. Mouse model of MI was used and heart samples were processed for immunohistochemical and immunofluorescent labeling and Enzyme linked immunosorbent assay to identify GAL-1 and HIF 1α levels in the heart during the first 24 hours following MI. There was significant increase in left ventricular GAL-1 at 20 (p = 0.001) and 30 minutes (p = 0.004) following MI. There was also a significant increase in plasma GAL-1 at 4 hours (p = 0.012) and 24 hours (p = 0.001) following MI. A significant increase in left ventricular HIF-1 α was seen at 20 minutes (p = 0.047) following MI. In conclusion, we show for the first time that GAL-1 level in the left ventricle is increased in early ischemic period. We also report for the first time that HIF-1 α is significantly increased at 20 minutes following MI. In addition we report for the first time that mouse plasma GAL-1 level is significantly raised as early as 4 hours following MI.
Collapse
Affiliation(s)
- Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- * E-mail:
| | - Satwat Hashmi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
22
|
Shibata T, Yamagata H, Uchida S, Otsuki K, Hobara T, Higuchi F, Abe N, Watanabe Y. The alteration of hypoxia inducible factor-1 (HIF-1) and its target genes in mood disorder patients. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:222-9. [PMID: 23333658 DOI: 10.1016/j.pnpbp.2013.01.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 01/01/2023]
Abstract
Recent studies suggest that the dysfunction of neural plasticity is associated with mood disorders. Hypoxia-inducible factor-1 (HIF-1), which is a transcriptional activator of vascular endothelial growth factor (VEGF), activates the cellular response to hypoxia. HIF-1 is ubiquitously expressed in all cells, including peripheral leukocytes. However, little is known about the role of HIF-1 in mood disorder. In the present study, we investigated the mRNA expression levels of HIF-1 (α and β) and its target genes (VEGF, GLUT1, PGK1, PFKFB3, and LDHA) in the peripheral white blood cells of patients with major depressive disorder (MDD) and bipolar disorder (BPD). We found increased expression of HIF- 1α and HIF-1β mRNA, as well as the target genes, VEGF, and PFKFB3 in both MDD and BPD patients in a depressive state compared to healthy control subjects. Furthermore, the mRNA expression levels of GLUT1, PGK1, and LDHA were increased in MDD patients in a depressive state compared to healthy control subjects. We also found increased expression of HIF-1α and LDHA mRNA in MDD patients in a remissive state, whereas the mRNA expression levels of other genes in a remissive state were comparable to those in healthy control subjects. There was no significant difference in mRNA expression levels of the genes examined among patients receiving any type of antidepressant or mood stabilizer. Our data suggest that altered expression of HIF-1 and its target genes mRNA in peripheral blood cells are associated-mainly in a state-dependent manner-with mood disorders (especially with MDD). In addition, altered expression of HIF-1 and its target genes may be associated with the pathophysiology of depression.
Collapse
Affiliation(s)
- Tomohiko Shibata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | |
Collapse
|