1
|
Schwarz KG, Pereyra KV, Díaz-Jara E, Vicencio SC, Del Rio R. Brainstem C1 neurons mediate heart failure decompensation and mortality during acute salt loading. Cardiovasc Res 2025; 121:241-253. [PMID: 39775485 PMCID: PMC12012444 DOI: 10.1093/cvr/cvae261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
AIMS Heart failure (HF) is an emerging epidemic worldwide. Despite advances in treatment, the morbidity and mortality rate of HF remain high, and the global prevalence continues to rise. Common clinical features of HF include cardiac sympathoexcitation, disordered breathing, and kidney dysfunction; kidney dysfunction strongly contributes to sodium retention and fluid overload, leading to poor outcomes of HF patients. We have previously shown that brainstem pre-sympathetic neurons (C1) from the rostral ventrolateral medulla (RVLM) play a key role in sympathetic regulation in experimental models of HF. However, the role of RVLM-C1 neurons during salt-loading in the context of HF is unknown. This study tests whether RVLM C1 neurons drive cardiorespiratory decompensation and ultimately lead to sudden death in HF rats. METHODS AND RESULTS Adult male Sprague-Dawley rats underwent arteriovenous shunt to induce HF with preserved ejection fraction (HFpEF). Two weeks after HFpEF induction, bilateral selective ablation of RVLM C1 neurons was performed using anti-dopamine β-hydroxylase-saporin toxin. Animals were then fed a high Na+ diet (3% Na+ in food and 2% Na+ in water) for 3 weeks to induce compensated-to-decompensated HF state transition. Echocardiography, cardiac autonomic function, breathing function, and survival were assessed during the progression of HF. Salt loading resulted in marked decompensation in HF rats, as evidenced by a significant decrease in survival rates (survival: 10% vs. 100% HFpEF + Na+ vs. HFpEF). Furthermore, HFpEF + Na+ animals showed a further increase in cardiac sympathetic drive and more severe disordered breathing, including higher hypoxia-related epochs (i.e. apnoeas/hypopnoeas), compared with HF. Ablation of RVLM C1 neurons partly reduced the excessive cardiac sympathoexcitation during salt loading in HF, improved the exaggerated disordered breathing in HFpEF+ Na+ rats, and reduced decompensation-linked mortality. We found that hypoxia, but not high sodium, was the major contributor to impaired calcium handling in isolated adult cardiomyocytes. CONCLUSION Our results strongly suggest that RVLM C1 neurons contribute to acute HF decompensation during salt loading by a mechanism encompassing further increases in sympathetic outflow and hypoxia-related breathing disorders. This mechanism may ultimately impact cardiac contractility through cardiomyocyte calcium mishandling, increasing morbidity and mortality.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Sinay C Vicencio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Av. los Flamencos 01364, Punta Arenas 6210005, Chile
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd HLSIC-2091, Kansas City, KS 66160, USA
| |
Collapse
|
2
|
Fang GH, Zhou XL, Ran CL, Jin CX, Bu SY, Chen Y, Gong Y, Hu ZT, Song FB, Luo J, Sun JL. Chronic intermittent hypoxia modulates energy metabolic pathways and improves hypoxia tolerance capacity in golden pompano, Trachinotus blochii. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 969:178921. [PMID: 40022974 DOI: 10.1016/j.scitotenv.2025.178921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Hypoxia is one of the most significant abiotic stresses affecting organisms in aquatic environments. However, dissolved oxygen in water is not always at the low concentrations that cause hypoxia, but their levels often fluctuate. Here, golden pompano (Trachinotus blochii) was subjected to low oxygen concentrations for 28 days (intermittent hypoxia, 2 h per day), and their metabolic indexes were systematically evaluated. RNA-seq was used to construct a regulatory network to elucidate the transcriptional regulation of golden pompano metabolism under intermittent hypoxia. We found that the liver lactic acid content, as well as hexokinase and phosphofructokinase activities, were elevated during the first 7 days, suggesting that anaerobic glycolysis was enhanced during the preceding period. In addition, triglyceride, lipoprotein lipase, and carnitine palmitoyltransferase-1 levels were elevated in the liver after 14 days, suggesting that lipid utilization was activated after 14 days. Intermittent hypoxia increased the activity of aspartate aminotransferase and alanine aminotransferase in the liver, decreased total plasma protein and amino acid levels, and enhanced the metabolism of proteins and amino acids. Decreased levels of oxidative stress and LOEcrit (the O2 tension for loss of equilibrium) were observed in golden pompano after 28 days of intermittent hypoxia. Transcriptome analysis showed that the fatty acid metabolism, PPAR signaling pathway, fatty acid degradation, D-amino acid metabolism, and cholesterol metabolism pathway were activated. These results suggest that intermittent hypoxia improves the metabolic activities of golden pompano, increases its hypoxia tolerance, and promotes its adaptation to hypoxic environments.
Collapse
Affiliation(s)
- Geng Hui Fang
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Xiao Li Zhou
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Chun Li Ran
- Guangdong Evergreen Conglomerate Co., Ltd., Key Laboratory of Aquatic Animal Breeding and Culturing in South China Sea, Ministry of Agriculture and Rural Affairs, Zhanjiang 524000, China
| | - Chun Xiu Jin
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Shao Yang Bu
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Yue Chen
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Yong Gong
- Guangdong Evergreen Conglomerate Co., Ltd., Key Laboratory of Aquatic Animal Breeding and Culturing in South China Sea, Ministry of Agriculture and Rural Affairs, Zhanjiang 524000, China
| | - Zeng Tan Hu
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Fei Biao Song
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China
| | - Jian Luo
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China.
| | - Jun Long Sun
- School of Marine Biology and Fisheries, Sanya Nanfan Research Institute of Hainan University, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Hainan University, Haikou 570228, China.
| |
Collapse
|
3
|
Amirrad F, La V, Ohadi S, Albotaif M, Webster S, Pru JK, Shamloo K, Mohieldin AM, Nauli SM. PGRMC2 is a pressure-volume regulator critical for myocardial responses to stress in mice. Nat Commun 2025; 16:2422. [PMID: 40069180 PMCID: PMC11897200 DOI: 10.1038/s41467-025-57707-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 03/01/2025] [Indexed: 03/14/2025] Open
Abstract
Progesterone receptors are classified into nuclear and membrane-bound receptor families. Previous unbiased proteomic studies indicate a potential association between cardiac diseases and the progesterone receptor membrane-bound component-2 (PGRMC2); however, the role of PGRMC2 in the heart remains unknown. In this study, we use a heart-specific knockout (KO) mouse model (MyH6•Pgrmc2flox/flox) in which the Pgrmc2 gene was selectively deleted in cardiomyocytes. Here we show that PGRMC2 serves as a mediator of steroid hormones for rapid calcium signaling in cardiomyocytes to maintain cardiac contraction, sufficient stroke volume, and adequate cardiac output by regulating the cardiac pressure-volume relationship. The KO hearts from male and female mice exhibit an impairment in pressure-volume relationship. Under hypoxic conditions, this pressure-volume dysregulation progresses to congestive left and right ventricular failure in the KO hearts. Overall, we propose that PGRMC2 is a cardiac pressure-volume regulator to maintain normal cardiac physiology, especially during hypoxic stress.
Collapse
Affiliation(s)
- Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
- Department of Pharmaceutical Sciences, Marshall B. Ketchum University, Fullerton, USA
| | - Vivian La
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Sharareh Ohadi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Miram Albotaif
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Sha Webster
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - James K Pru
- Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Kiumars Shamloo
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Ashraf M Mohieldin
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
- College of Graduate Studies, California Northstate University, Elk Grove, CA, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA.
- Department of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
4
|
Bambor C, Daunheimer S, Raffort C, Koedel J, Salameh A, Raßler B. Effects of a Three-Day vs. Six-Day Exposure to Normobaric Hypoxia on the Cardiopulmonary Function of Rats. Curr Issues Mol Biol 2025; 47:125. [PMID: 39996846 PMCID: PMC11854188 DOI: 10.3390/cimb47020125] [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: 01/08/2025] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
In rats, normobaric hypoxia significantly reduced left ventricular (LV) inotropic function while right ventricular (RV) function was not impaired. In parallel, the animals developed pulmonary edema and inflammation. In the present study, we investigated whether cardiac function and pulmonary injury would aggravate after three and six days of hypoxia exposure or whether cardiopulmonary reactions to prolonged hypoxia would become weaker due to hypoxic acclimatization. Sixty-four female rats were exposed for 72 or 144 h to normoxia. They received a low-rate infusion (0.1 mL/h) with 0.9% NaCl solution. We evaluated indicators of the general condition, blood gas parameters, and hemodynamic function of the rats. In addition, we performed histological and immunohistochemical analyses of the lung. Despite a significant increase in hemoglobin concentration, the LV function deteriorated with prolonged hypoxia. In contrast, the RV systolic pressure and contractility steadily increased by six days of hypoxia. The pulmonary edema and inflammation persisted and rather increased with prolonged hypoxia. Furthermore, elevated protein concentration in the pleural fluid indicated capillary wall stress, which may have aggravated the pulmonary edema. In conclusion, six days of hypoxia and NaCl infusion place significant stress on the cardiopulmonary system of rats, as is also reflected by the 33% of premature deaths in this rat group.
Collapse
Affiliation(s)
- Charly Bambor
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| | - Sarah Daunheimer
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| | - Coralie Raffort
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Julia Koedel
- Institute of Pathology, University of Leipzig, 04103 Leipzig, Germany;
| | - Aida Salameh
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Beate Raßler
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (C.B.); (S.D.)
| |
Collapse
|
5
|
Wu C, Huang J, Huang M, Tan Y, Chen C, Zheng M, Zhao W, Xu Y, Guo L, Wu X, Xue Y, Deng H, Liu X. Association of electrocardiogram features with risk of obstructed sleep apnea: a population-based cohort study. Sleep Breath 2025; 29:96. [PMID: 39934598 DOI: 10.1007/s11325-025-03266-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/05/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND There is limited investigation on the longitudinal association between common electrocardiogram (ECG) features and the incidence of obstructive sleep apnea (OSA). This study aimed to examine the association of common ECG features with the incidence of OSA in a prospective cohort. METHODS 2,563 participants aged 60 years or more were selected from the baseline survey of the Guangzhou Heart Study. OSA was evaluated by the Berlin Questionnaire. Eight electrocardiogram features including PR interval, QRS duration, QT interval, QTc interval, heart rate, P-wave, R-wave, and T-wave were extracted from 24-hour single-lead Holter. Relative risk (RR) with 95% confidence interval (CI) was estimated using the multivariate logistic regression model. A receiver operating characteristic (ROC) curve analysis was used to evaluate the predictive ability of ECG features. RESULTS 397 (15.5%) participants were divided into the OSA group and 2,166 (84.5%) into the OSA non-group. When comparing the highest with the lowest quartiles, heart rate was related to a 30% reduced risk of OSA (RR: 0.70, 95%CI: 0.51-0.97) after adjustment for possible confounders. Participants with prolonged PR interval were more likely to be at risk of OSA (RR: 2.68, 95%CI: 1.02-6.55). No significant association was found between the other six ECG features and OSA risk. Area under ROC curve was 0.676 (95% CI: 0.648-0.704), 0.676 (95%CI: 0.648-0.704), and 0.678 (95%CI: 0.651-0.706) for heart rate, PR interval, and their combination, respectively. CONCLUSIONS The results suggest that heart rate and PR interval are related to OSA incidence. Future studies should be carried out in different populations, and consider the use of portable monitors together with scales to comprehensively determine OSA, and comprehensively elucidate the relationship of various ECG features and their changes with OSA occurrence.
Collapse
Affiliation(s)
- Chuchu Wu
- School of Public Health, Guangdong Pharmaceutical University, No. 283 Jianghai Avenue, Haizhu District, Guangzhou, 510310, China
| | - Jun Huang
- Department of Geriatrics, Institute of Geriatrics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Southern Medical University, Guangzhou, 510080, China
| | - Minjing Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science,Southern Medical University, Guangzhou, 510080, China
| | - Yiting Tan
- School of Public Health, Guangdong Pharmaceutical University, No. 283 Jianghai Avenue, Haizhu District, Guangzhou, 510310, China
| | - Chuanjiang Chen
- School of Public Health, Guangdong Pharmaceutical University, No. 283 Jianghai Avenue, Haizhu District, Guangzhou, 510310, China
| | - Murui Zheng
- Faculty of Health Sciences, University of Macau, Macau, 999078, SAR, China
| | - Wenjing Zhao
- School of Public Health and Emergency Management, Southern University of Science and Technology, No.1088, Xueyuan Avenue, Nanshan District, Shenzhen, 518055, China.
| | - Yangjie Xu
- Guangzhou Xinzao Town Community Health Service Center, Guangzhou, 511442, China
| | - Lili Guo
- Guangzhou Xinzao Town Community Health Service Center, Guangzhou, 511442, China
| | - Xiuyi Wu
- Guangzhou Nancun Town Community Health Service Center, Guangzhou, 511442, China
| | - Yumei Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science,Southern Medical University, Guangzhou, 510080, China
| | - Hai Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Southern Medical University, 5/F, Ying Tung Building, No.106, Zhongshan Second Road, Guangzhou, 518055, China.
| | - Xudong Liu
- School of Public Health, Guangdong Pharmaceutical University, No. 283 Jianghai Avenue, Haizhu District, Guangzhou, 510310, China.
| |
Collapse
|
6
|
Dwyer KD, Snyder CA, Coulombe KLK. Cardiomyocytes in Hypoxia: Cellular Responses and Implications for Cell-Based Cardiac Regenerative Therapies. Bioengineering (Basel) 2025; 12:154. [PMID: 40001674 PMCID: PMC11851968 DOI: 10.3390/bioengineering12020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Myocardial infarction (MI) is a severe hypoxic event, resulting in the loss of up to one billion cardiomyocytes (CMs). Due to the limited intrinsic regenerative capacity of the heart, cell-based regenerative therapies, which feature the implantation of stem cell-derived cardiomyocytes (SC-CMs) into the infarcted myocardium, are being developed with the goal of restoring lost muscle mass, re-engineering cardiac contractility, and preventing the progression of MI into heart failure (HF). However, such cell-based therapies are challenged by their susceptibility to oxidative stress in the ischemic environment of the infarcted heart. To maximize the therapeutic benefits of cell-based approaches, a better understanding of the heart environment at the cellular, tissue, and organ level throughout MI is imperative. This review provides a comprehensive summary of the cardiac pathophysiology occurring during and after MI, as well as how these changes define the cardiac environment to which cell-based cardiac regenerative therapies are delivered. This understanding is then leveraged to frame how cell culture treatments may be employed to enhance SC-CMs' hypoxia resistance. In this way, we synthesize both the complex experience of SC-CMs upon implantation and the engineering techniques that can be utilized to develop robust SC-CMs for the clinical translation of cell-based cardiac therapies.
Collapse
Affiliation(s)
| | | | - Kareen L. K. Coulombe
- Institute for Biology, Engineering, and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA; (K.D.D.); (C.A.S.)
| |
Collapse
|
7
|
Waleczek FJG, Cipriano G, Haas JA, Garg A, Pfanne A, Just A, Neumüller S, Hegermann J, Pich A, Radocaj A, Xiao K, Weber N, Thum T. Prolonged Hypoxia in Rat Living Myocardial Slices Affects Function, Expression, and Structure. Int J Mol Sci 2024; 26:218. [PMID: 39796086 PMCID: PMC11720517 DOI: 10.3390/ijms26010218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Ischemic heart disease is the leading cause of death worldwide. Reduced oxygen supply and myocardial hypoxia lead to tissue damage and impairment of the heart function. To the best of our knowledge, the primary functional effects of hypoxia in the multicellular model of living myocardial slices (LMSs) have not been investigated so far. In this study, we analyzed force generation, ultrastructure, gene expression, and proteome changes in rat LMS after 24 h of ex vivo culture in normal and reduced levels of oxygen (O2). We observed a significant reduction in absolute force and a slowdown of force kinetics as well as an increase in cardiomyocyte apoptosis and myofibrillar and mitochondrial damage, as well as transcriptomic changes. Proteome analysis revealed the deregulation of proteins involved in metabolic processes, hypoxic response, and neutralizing of reactive oxygen species. Our results indicate that hypoxia induces substantial primary changes in heart tissue, which are independent of perfusion and immune responses. Our new LMS model could serve as a screening system for drug development and new mechanistic insights.
Collapse
Affiliation(s)
- Florian J. G. Waleczek
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Giuseppe Cipriano
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Jonas A. Haas
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Ankita Garg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Angelika Pfanne
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Susanne Neumüller
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany;
| | - Andreas Pich
- Institute of Toxicology and Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany;
| | - Ante Radocaj
- Institute of Molecular and Cell Physiology, Hannover Medical School, 30625 Hannover, Germany;
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Natalie Weber
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (F.J.G.W.); (G.C.); (J.A.H.); (A.G.); (A.P.); (A.J.); (S.N.); (K.X.)
| |
Collapse
|
8
|
Zhao M, Wu Q, Duanmu W, Shen J, Yuan W, Sun Y, Zhang X, Zhang J, He S. Clinical Analysis of Myocardial Injury in Highlanders with Pulmonary Hypertension. High Alt Med Biol 2024; 25:205-211. [PMID: 38900692 DOI: 10.1089/ham.2023.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Background: Pulmonary hypertension (PH) is a prevalent adverse cardiovascular event at high-altitude environments. Prolonged exposure to high altitudes may result in myocardial injury, which is associated with poor clinical outcomes. This study aims to investigate the clinical characteristics of myocardial injury in patients with PH at high altitude. Methods: Consecutive patients admitted to a general tertiary hospital at the altitude of 3,650 m were selected into this retrospective study. Clinical and biochemical data were collected, as well as based on cardiac troponin I (cTnI) and echocardiography, patients were divided into myocardial injury group and non-myocardial injury group. Results: A total of 231 patients were enrolled, among whom 29 (12.6%) had myocardial injury. We found that body mass index, left ventricular end-diastolic dimension, and serum level of creatine kinase-MB (CK-MB) in myocardial injury group were significantly higher than non-myocardial injury group. Spearman correlation analysis revealed that cTnI has a significant positive correlation with CK-MB and lactic dehydrogenase instead of aspartate aminotransferase. A receiver operating characteristic curve was drawn to demonstrate that CK-MB could significantly predict the occurrence of myocardial injury with an area under the curve of 0.749, and a level of 3.035 (sensitivity = 59.3%, specificity = 90.5%) was optimal cutoff value. Conclusion: The incidence of myocardial injury in highlanders with PH is significant. CK-MB, as a convenient and efficient marker, has been found to be closely associated with cTnI and plays a predictive role in the occurrence of myocardial injury with PH in individuals exposed to high altitude.
Collapse
Affiliation(s)
- Maolin Zhao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, The General Hospital of Western Theater Command, Chengdu, China
| | - Qianjin Wu
- Department of Health Service, Tibetan Military General Hospital, Lhasa, China
| | - Wangsheng Duanmu
- Department of Neurology, Tibetan Military General Hospital, Lhasa, China
| | - Junxian Shen
- Department of Neurology, Tibetan Military General Hospital, Lhasa, China
| | - Weixin Yuan
- Department of Neurology, Tibetan Military General Hospital, Lhasa, China
| | - Yingbin Sun
- Department of Cardiology, Tibetan Military General Hospital, Lhasa, China
| | - Xu Zhang
- Department of Cardiology, Tibetan Military General Hospital, Lhasa, China
| | - Jinbao Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, The General Hospital of Western Theater Command, Chengdu, China
| | - Siyi He
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
9
|
Wang Y, Zhang R, Chen Q, Lei Z, Shi C, Pang Y, Zhang S, He L, Xu L, Xing J, Guo H. PPARγ Agonist Pioglitazone Prevents Hypoxia-induced Cardiac Dysfunction by Reprogramming Glucose Metabolism. Int J Biol Sci 2024; 20:4297-4313. [PMID: 39247816 PMCID: PMC11379067 DOI: 10.7150/ijbs.98387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 09/10/2024] Open
Abstract
The heart relies on various defense mechanisms, including metabolic plasticity, to maintain its normal structure and function under high-altitude hypoxia. Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ), sensitizes insulin, which in turn regulates blood glucose levels. However, its preventive effects against hypoxia-induced cardiac dysfunction at high altitudes have not been reported. In this study, pioglitazone effectively prevented cardiac dysfunction in hypoxic mice for 4 weeks, independent of its effects on insulin sensitivity. In vitro experiments demonstrated that pioglitazone enhanced the contractility of primary cardiomyocytes and reduced the risk of QT interval prolongation under hypoxic conditions. Additionally, pioglitazone promoted cardiac glucose metabolic reprogramming by increasing glycolytic capacity; enhancing glucose oxidation, electron transfer, and oxidative phosphorylation processes; and reducing mitochondrial reactive ROS production, which ultimately maintained mitochondrial membrane potential and ATP production in cardiomyocytes under hypoxic conditions. Notably, as a PPARγ agonist, pioglitazone promoted hypoxia-inducible factor 1α (HIF-1α) expression in hypoxic myocardium. Moreover, KC7F2, a HIF-1α inhibitor, disrupted the reprogramming of cardiac glucose metabolism and reduced cardiac function in pioglitazone-treated mice under hypoxic conditions. In conclusion, pioglitazone effectively prevented high-altitude hypoxia-induced cardiac dysfunction by reprogramming cardiac glucose metabolism.
Collapse
Affiliation(s)
- Yijin Wang
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Ru Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Fourth Military Medical University, 710069, China
| | - Qian Chen
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Zhangwen Lei
- School of Medicine, Northwest University, Xi'an, 710069, China
| | - Caiyu Shi
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yifei Pang
- School of Medicine, Northwest University, Xi'an, 710069, China
| | - Shan'an Zhang
- College of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Linjie He
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Longtao Xu
- College of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Haitao Guo
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
10
|
Zhang X, Zheng W, Sun S, Du Y, Xu W, Sun Z, Liu F, Wang M, Zhao Z, Liu J, Liu Q. Cadmium contributes to cardiac metabolic disruption by activating endothelial HIF1A-GLUT1 axis. Cell Signal 2024; 119:111170. [PMID: 38604344 DOI: 10.1016/j.cellsig.2024.111170] [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: 01/21/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Cadmium (Cd) is an environmental risk factor of cardiovascular diseases. Researchers have found that Cd exposure causes energy metabolic disorders in the heart decades ago. However, the underlying molecular mechanisms are still elusive. In this study, male C57BL/6 J mice were exposed to cadmium chloride (CdCl2) through drinking water for 4 weeks. We found that exposure to CdCl2 increased glucose uptake and utilization, and disrupted normal metabolisms in the heart. In vitro studies showed that CdCl2 specifically increased endothelial glucose uptake without affecting cardiomyocytic glucose uptake and endothelial fatty acid uptake. The glucose transporter 1 (GLUT1) as well as its transcription factor HIF1A was significantly increased after CdCl2 treatment in endothelial cells. Further investigations found that CdCl2 treatment upregulated HIF1A expression by inhibiting its degradation through ubiquitin-proteasome pathway, thereby promoted its transcriptional activation of SLC2A1. Administration of HIF1A small molecule inhibitor echinomycin and A-485 reversed CdCl2-mediated increase of glucose uptake in endothelial cells. In accordance with this, intravenous injection of echinomycin effectively ameliorated CdCl2-mediated metabolic disruptions in the heart. Our study uncovered the molecular mechanisms of Cd in contributing cardiac metabolic disruption by inhibiting HIF1A degradation and increasing GLUT1 transcriptional expression. Inhibition of HIF1A could be a potential strategy to ameliorate Cd-mediated cardiac metabolic disorders and Cd-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Wendan Zheng
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Shiyu Sun
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Yang Du
- Department of Personnel, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Wenjuan Xu
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering Laboratory for Health Management, Ji'nan, Shandong, China
| | - Zongguo Sun
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Fuhong Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Manzhi Wang
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Zuohui Zhao
- Department of Pediatric Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Ju Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Qiang Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Ji'nan, Shandong, China.
| |
Collapse
|
11
|
Iwoń Z, Krogulec E, Kierlańczyk A, Wojasiński M, Jastrzębska E. Hypoxia and re-oxygenation effects on human cardiomyocytes cultured on polycaprolactone and polyurethane nanofibrous mats. J Biol Eng 2024; 18:37. [PMID: 38844979 PMCID: PMC11157810 DOI: 10.1186/s13036-024-00432-5] [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: 03/28/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Heart diseases are caused mainly by chronic oxygen insufficiency (hypoxia), leading to damage and apoptosis of cardiomyocytes. Research into the regeneration of a damaged human heart is limited due to the lack of cellular models that mimic damaged cardiac tissue. Based on the literature, nanofibrous mats affect the cardiomyocyte morphology and stimulate the growth and differentiation of cells cultured on them; therefore, nanofibrous materials can support the production of in vitro models that faithfully mimic the 3D structure of human cardiac tissue. Nanofibrous mats were used as scaffolds for adult primary human cardiomyocytes (HCM) and immature human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). This work focuses on understanding the effects of hypoxia and re-oxygenation on human cardiac cells cultured on polymer nanofibrous mats made of poly(ε-caprolactone) (PCL) and polyurethane (PU). The expression of selected genes and proteins in cardiomyocytes during hypoxia and re-oxygenation were evaluated. In addition, the type of cell death was analyzed. To the best of our knowledge, there are no studies on the effects of hypoxia on cardiomyocyte cells cultured on nanofibrous mats. The present study aimed to use nanofiber mats as scaffolds that structurally could mimic cardiac extracellular matrix. Understanding the impact of 3D structural properties in vitro cardiac models on different human cardiomyocytes is crucial for advancing cardiac tissue engineering and regenerative medicine. Observing how 3D scaffolds affect cardiomyocyte function under hypoxic conditions is necessary to understand the functioning of the entire human heart.
Collapse
Affiliation(s)
- Zuzanna Iwoń
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Ewelina Krogulec
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Aleksandra Kierlańczyk
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Michał Wojasiński
- Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland.
- Centre for Advanced Materials and Technologies, CEZAMAT Warsaw University of Technology, Warsaw, Poland.
| |
Collapse
|
12
|
Condori LDM, Vivas CV, Barreto YB, Gomes LF, Alencar AM, Bloise AC. Effects of Hypoxia and Reoxygenation on Metabolic Profiles of Cardiomyocytes. Cell Biochem Biophys 2024; 82:969-985. [PMID: 38498099 DOI: 10.1007/s12013-024-01249-1] [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] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
In vitro cellular models provide valuable insights into the adaptive biochemical mechanisms triggered by cells to cope with the stress situation induced by hypoxia and reoxygenation cycles. The first biological data generated in studies based on this micrometric life-scale has the potential to provide us a global overview about the main biochemical phenomena presented in some reported preconditioning therapies in life-scale of higher dimensions. Thus, in this study, a cell incubator was designed and manufactured to produce a cellular model of heart hypoxia followed by reoxygenation (HfR) through consecutive repetitions of hypoxia-normoxia gas exchange. Samples of cellular extracts and culture media were obtained from non-proliferative cardiomyocytes (CMs) cultivated under challenging HfR (stressed CMs) and regular cultivation (unstressed CMs) in rounds of four days for each case. Metabolomic based on proton magnetic resonance spectroscopy (1H-MRS) was used as an analytical approach to identify and quantify the metabolomes of these samples, the endo- and exo-metabolome. Despite the stressed CMs presented over 90% higher cellular death rate compared to the unstressed CMs, the metabolic profiles indicates that the surviving cells up-regulate their amino acid metabolism either by active protein degradation or by the consumption of culture media components to increase coenzyme A-dependent metabolic pathways. This cell auto-regulation mechanism could be well characterized in the first two days when the difference smears off under once the metabolomes become similar. The metabolic adaptations of stressed CMs identified the relevance of the cyclic oxidation/reduction reactions of nicotinamide adenine dinucleotide phosphate molecules, NADP+/NADPH, and the increased tricarboxylic acid cycle activity in an environment overloaded with such a powerful antioxidant agent to survive an extreme HfR challenge. Thus, the combination of cellular models based on CMs, investigative methods, such as metabolomic and 1H-MRS, and the instrumental development of hypoxia incubator shown in this work were able to provide the first biochemical evidences behind therapies of gaseous exchanges paving the way to future assays.
Collapse
Affiliation(s)
| | | | - Yan Borges Barreto
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil
| | - Ligia Ferreira Gomes
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil.
| | | | - Antonio Carlos Bloise
- Universidade de Sao Paulo, Instituto de Fisica, Rua do Matao 1371, Sao Paulo, Brazil.
| |
Collapse
|
13
|
Ostrowska-Leśko M, Herbet M, Pawłowski K, Korga-Plewko A, Poleszak E, Dudka J. Pathological Changes and Metabolic Adaptation in the Myocardium of Rats in Response to Chronic Variable Mild Stress. Int J Mol Sci 2024; 25:5899. [PMID: 38892086 PMCID: PMC11172974 DOI: 10.3390/ijms25115899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic variable mild stress (CVS) in rats is a well-established paradigm for inducing depressive-like behaviors and has been utilized extensively to explore potential therapeutic interventions for depression. While the behavioral and neurobiological effects of CVS have been extensively studied, its impact on myocardial function remains largely unexplored. To induce the CVS model, rats were exposed to various stressors over 40 days. Behavioral assessments confirmed depressive-like behavior. Biochemical analyses revealed alterations in myocardial metabolism, including changes in NAD+ and NADP+, and NADPH concentrations. Free amino acid analysis indicated disturbances in myocardial amino acid metabolism. Evaluation of oxidative DNA damage demonstrated an increased number of abasic sites in the DNA of rats exposed to CVS. Molecular analysis showed significant changes in gene expression associated with glucose metabolism, oxidative stress, and cardiac remodeling pathways. Histological staining revealed minor morphological changes in the myocardium of CVS-exposed rats, including increased acidophilicity of cells, collagen deposition surrounding blood vessels, and glycogen accumulation. This study provides novel insights into the impact of chronic stress on myocardial function and metabolism, highlighting potential mechanisms linking depression and cardiovascular diseases. Understanding these mechanisms may aid in the development of targeted therapeutic strategies to mitigate the adverse cardiovascular effects of depression.
Collapse
Affiliation(s)
- Marta Ostrowska-Leśko
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Mariola Herbet
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Kamil Pawłowski
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodźko Street, 20-093 Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (M.H.); (J.D.)
| |
Collapse
|
14
|
Jin Y, Ren W, Liu J, Tang X, Shi X, Pan D, Hou L, Yang L. Identification and validation of potential hypoxia-related genes associated with coronary artery disease. Front Physiol 2023; 14:1181510. [PMID: 37637145 PMCID: PMC10447898 DOI: 10.3389/fphys.2023.1181510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction: Coronary artery disease (CAD) is one of the most life-threatening cardiovascular emergencies with high mortality and morbidity. Increasing evidence has demonstrated that the degree of hypoxia is closely associated with the development and survival outcomes of CAD patients. However, the role of hypoxia in CAD has not been elucidated. Methods: Based on the GSE113079 microarray dataset and the hypoxia-associated gene collection, differential analysis, machine learning, and validation of the screened hub genes were carried out. Results: In this study, 54 differentially expressed hypoxia-related genes (DE-HRGs), and then 4 hub signature genes (ADM, PPFIA4, FAM162A, and TPBG) were identified based on microarray datasets GSE113079 which including of 93 CAD patients and 48 healthy controls and hypoxia-related gene set. Then, 4 hub genes were also validated in other three CAD related microarray datasets. Through GO and KEGG pathway enrichment analyses, we found three upregulated hub genes (ADM, PPFIA4, TPBG) were strongly correlated with differentially expressed metabolic genes and all the 4 hub genes were mainly enriched in many immune-related biological processes and pathways in CAD. Additionally, 10 immune cell types were found significantly different between the CAD and control groups, especially CD8 T cells, which were apparently essential in cardiovascular disease by immune cell infiltration analysis. Furthermore, we compared the expression of 4 hub genes in 15 cell subtypes in CAD coronary lesions and found that ADM, FAM162A and TPBG were all expressed at higher levels in endothelial cells by single-cell sequencing analysis. Discussion: The study identified four hypoxia genes associated with coronary heart disease. The findings provide more insights into the hypoxia landscape and, potentially, the therapeutic targets of CAD.
Collapse
Affiliation(s)
- Yuqing Jin
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Weiyan Ren
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Jiayi Liu
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xuejiao Tang
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xinrui Shi
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Dongchen Pan
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lianguo Hou
- Biochemistry Research Laboratory, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Lei Yang
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
15
|
Cao Y, Redd MA, Fang C, Mizikovsky D, Li X, Macdonald PS, King GF, Palpant NJ. New Drug Targets and Preclinical Modelling Recommendations for Treating Acute Myocardial Infarction. Heart Lung Circ 2023:S1443-9506(23)00139-7. [PMID: 37230806 DOI: 10.1016/j.hlc.2022.12.015] [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: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 05/27/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide and the primary underlying risk factor for heart failure. Despite decades of research and clinical trials, there are no drugs currently available to prevent organ damage from acute ischaemic injuries of the heart. In order to address the increasing global burden of heart failure, drug, gene, and cell-based regeneration technologies are advancing into clinical testing. In this review we highlight the burden of disease associated with AMI and the therapeutic landscape based on market analyses. New studies revealing the role of acid-sensitive cardiac ion channels and other proton-gated ion channels in cardiac ischaemia are providing renewed interest in pre- and post-conditioning agents with novel mechanisms of action that may also have implications for gene- and cell-based therapeutics. Furthermore, we present guidelines that couple new cell technologies and data resources with traditional animal modelling pipelines to help de-risk drug candidates aimed at treating AMI. We propose that improved preclinical pipelines and increased investment in drug target identification for AMI is critical to stem the increasing global health burden of heart failure.
Collapse
Affiliation(s)
- Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Chen Fang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Peter S Macdonald
- Cardiopulmonary Transplant Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
16
|
Naryzhnaya NV, Derkachev IA, Kurbatov BK, Sirotina MA, Kilin M, Maslov LN. Decrease in Infarct-Limiting Effect of Chronic Normobaric Hypoxia in Rats with Induced Metabolic Syndrome Is Associated with Disturbances of Carbohydrate and Lipid Metabolism. Bull Exp Biol Med 2023; 174:723-727. [PMID: 37171712 DOI: 10.1007/s10517-023-05779-1] [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: 06/21/2022] [Indexed: 05/13/2023]
Abstract
We studied the infarct-limiting effect of adaptation to chronic normobaric hypoxia in rats with induced metabolic syndrome and the relationship between disturbances of adaptive cardioprotection and disorders of carbohydrate and lipid metabolism. Adaptation to chronic normobaric hypoxia was carried out for 21 days at 12% O2 and 0.3% CO2. The metabolic syndrome was modeled with a high-carbohydrate high-fat diet for 84 days with replacement of drinking water with a 20% fructose solution. The infarct size in rats exposed to chronic normobaric hypoxia was 38% smaller than in control animals. In rats with induced metabolic syndrome, hypertension, obesity, decreased glucose tolerance, increased serum triglyceride, and no infarction-limiting effect of chronic normobaric hypoxia were observed. Infarct size showed a direct correlation with impaired glucose tolerance and serum triglyceride levels. The study allows us to conclude that the lack of cardioprotection in chronic normobaric hypoxia in rats with induced metabolic syndrome is associated with impaired carbohydrate and lipid metabolism.
Collapse
Affiliation(s)
- N V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.
| | - I A Derkachev
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - B K Kurbatov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - M A Sirotina
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - M Kilin
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - L N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
17
|
Del Coco L, Greco M, Inguscio A, Munir A, Danieli A, Cossa L, Musarò D, Coscia MR, Fanizzi FP, Maffia M. Blood Metabolite Profiling of Antarctic Expedition Members: An 1H NMR Spectroscopy-Based Study. Int J Mol Sci 2023; 24:ijms24098459. [PMID: 37176166 PMCID: PMC10179003 DOI: 10.3390/ijms24098459] [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: 03/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Serum samples from eight participants during the XV winter-over at Concordia base (Antarctic expedition) collected at defined time points, including predeparture, constituted the key substrates for a specific metabolomics study. To ascertain acute changes and chronic adaptation to hypoxia, the metabolic profiles of the serum samples were analyzed using NMR spectroscopy, with principal components analysis (PCA) followed by partial least squares and orthogonal partial least squares discriminant analyses (PLS-DA and OPLS-DA) used as supervised classification methods. Multivariate data analyses clearly highlighted an adaptation period characterized by an increase in the levels of circulating glutamine and lipids, mobilized to supply the body energy needs. At the same time, a reduction in the circulating levels of glutamate and N-acetyl glycoproteins, stress condition indicators, and proinflammatory markers were also found in the NMR data investigation. Subsequent pathway analysis showed possible perturbations in metabolic processes, potentially related to the physiological adaptation, predominantly found by comparing the baseline (at sea level, before mission onset), the base arrival, and the mission ending collected values.
Collapse
Affiliation(s)
- Laura Del Coco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Alessandra Inguscio
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Anas Munir
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
- Department of Mathematics and Physics "E. De Giorgi", University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy
| | - Antonio Danieli
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Luca Cossa
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Maria Rosaria Coscia
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Via P. Castellino 111, 80131 Naples, Italy
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
18
|
Ulaganathan T, Perales S, Mani S, Baskhairoun BA, Rajasingh J. Pathological implications of cellular stress in cardiovascular diseases. Int J Biochem Cell Biol 2023; 158:106397. [PMID: 36931385 PMCID: PMC10124590 DOI: 10.1016/j.biocel.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Cellular stress has been a key factor in the development of cardiovascular diseases. Major types of cellular stress such as mitochondrial stress, endoplasmic reticulum stress, hypoxia, and replicative stress have been implicated in clinical complications of cardiac patients. The heart is the central regulator of the body by supplying oxygenated blood throughout the system. Impairment of cellular function could lead to heart failure, myocardial infarction, ischemia, and even stroke. Understanding the effect of these distinct types of cellular stress on cardiac function is crucial for the scientific community to understand and develop novel therapeutic approaches. This review will comprehensively explain the different mechanisms of cellular stress and the most recent findings related to stress-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Thennavan Ulaganathan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Selene Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Saiprahalad Mani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Boula A Baskhairoun
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
19
|
Marzook H, Gupta A, Tomar D, Saleh MA, Patil K, Semreen MH, Hamoudi R, Soares NC, Qaisar R, Ahmad F. Nicotinamide riboside kinase-2 regulates metabolic adaptation in the ischemic heart. J Mol Med (Berl) 2023; 101:311-326. [PMID: 36808555 DOI: 10.1007/s00109-023-02296-6] [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: 11/22/2022] [Revised: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/23/2023]
Abstract
Ischemia-induced metabolic remodeling plays a critical role in the pathogenesis of adverse cardiac remodeling and heart failure however, the underlying molecular mechanism is largely unknown. Here, we assess the potential roles of nicotinamide riboside kinase-2 (NRK-2), a muscle-specific protein, in ischemia-induced metabolic switch and heart failure through employing transcriptomic and metabolomic approaches in ischemic NRK-2 knockout mice. The investigations revealed NRK-2 as a novel regulator of several metabolic processes in the ischemic heart. Cardiac metabolism and mitochondrial function and fibrosis were identified as top dysregulated cellular processes in the KO hearts post-MI. Several genes linked to mitochondrial function, metabolism, and cardiomyocyte structural proteins were severely downregulated in the ischemic NRK-2 KO hearts. Analysis revealed significantly upregulated ECM-related pathways which was accompanied by the upregulation of several key cell signaling pathways including SMAD, MAPK, cGMP, integrin, and Akt in the KO heart post-MI. Metabolomic studies identified profound upregulation of metabolites mevalonic acid, 3,4-dihydroxyphenylglycol, 2-penylbutyric acid, and uridine. However, other metabolites stearic acid, 8,11,14-eicosatrienoic acid, and 2-pyrrolidinone were significantly downregulated in the ischemic KO hearts. Taken together, these findings suggest that NRK-2 promotes metabolic adaptation in the ischemic heart. The aberrant metabolism in the ischemic NRK-2 KO heart is largely driven by dysregulated cGMP and Akt and mitochondrial pathways. KEY MESSAGES: Post-myocardial infarction metabolic switch critically regulates the pathogenesis of adverse cardiac remodeling and heart failure. Here, we report NRK-2 as a novel regulator of several cellular processes including metabolism and mitochondrial function post-MI. NRK-2 deficiency leads to downregulation of genes important for mitochondrial pathway, metabolism, and cardiomyocyte structural proteins in the ischemic heart. It was accompanied by upregulation of several key cell signaling pathways including SMAD, MAPK, cGMP, integrin, and Akt and dysregulation of numerous metabolites essential for cardiac bioenergetics. Taken together, these findings suggest that NRK-2 is critical for metabolic adaptation of the ischemic heart.
Collapse
Affiliation(s)
- Hezlin Marzook
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Anamika Gupta
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Dhanendra Tomar
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mohamed A Saleh
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Kiran Patil
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
| | - Mohammad H Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. 27272, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Division of Surgery and Interventional Science, University College London, London, W1W 7EJ, UK
| | - Nelson C Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. 27272, Sharjah, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av.a Padre Cruz, Lisbon, 1649-016, Portugal
| | - Rizwan Qaisar
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Firdos Ahmad
- Research Institute of Medical and Health Sciences, University of Sharjah, P.O. 27272 , Sharjah, United Arab Emirates.
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, 59911, Abu Dhabi, United Arab Emirates.
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240, USA.
| |
Collapse
|
20
|
Ghosh R, Gillaspie JJ, Campbell KS, Symons JD, Boudina S, Pattison JS. Chaperone-mediated autophagy protects cardiomyocytes against hypoxic-cell death. Am J Physiol Cell Physiol 2022; 323:C1555-C1575. [PMID: 35584327 PMCID: PMC9829466 DOI: 10.1152/ajpcell.00369.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 05/02/2022] [Accepted: 05/16/2022] [Indexed: 01/22/2023]
Abstract
Chaperone-mediated autophagy (CMA) is a chaperone-dependent process of selective cytosolic protein turnover that targets specific proteins to lysosomes for degradation. Enhancing protein degradation mechanisms has been shown to be beneficial in multiple models of cardiac disease, including myocardial infarction (MI) and ischemia-reperfusion (I/R) injury. However, the causal role of CMA in cardiomyocyte injury and death is largely unknown. Hypoxia is an important contributor to both MI and I/R damage, which are major, precedent causes of heart failure. Upregulating CMA was hypothesized to protect against hypoxia-induced cardiomyocyte death. Lysosome-associated membrane protein 2a (Lamp2a) overexpression and knockdown were used to causally study CMA's role in hypoxically stressed cardiomyocytes. LAMP2a protein levels were used as both a primary indicator and driver of CMA function. Hypoxic stress was stimulated by CoCl2 treatment, which increased LAMP2a protein levels (+1.4-fold) and induced cardiomyocyte apoptosis (+3.2-4.0-fold). Lamp2a siRNA knockdown (-3.2-fold) of control cardiomyocytes increased apoptosis (+1.8-fold) suggesting that loss of CMA is detrimental for cardiomyocyte survival. However, there was neither an additive nor a synergistic effect on cell death when Lamp2a-silenced cells were treated with CoCl2. Conversely, Lamp2a overexpression (+3.0-fold) successfully reduced hypoxia-induced apoptosis by ∼50%. LAMP2a was also significantly increased (+1.7-fold) in ischemic heart failure patient samples, similar to hypoxically stressed cardiomyocytes. The failing ischemic hearts may have had insufficient CMA activation. To our knowledge, this study for the first time establishes a protective role for CMA (via Lamp2a overexpression) against hypoxia-induced cardiomyocyte loss and reveals the intriguing possibility that CMA activation may offer a cardioprotective treatment for ischemic heart disease.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
- Department of Nutrition and Integrative Physiology Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Jennifer Jason Gillaspie
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Kenneth S Campbell
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - J David Symons
- Department of Nutrition and Integrative Physiology Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - James Scott Pattison
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
21
|
Zhang Z, Wu L, Cui T, Ahmed RZ, Yu H, Zhang R, Wei Y, Li D, Zheng Y, Chen W, Jin X. Oxygen sensors mediated HIF-1α accumulation and translocation: A pivotal mechanism of fine particles-exacerbated myocardial hypoxia injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 300:118937. [PMID: 35114305 DOI: 10.1016/j.envpol.2022.118937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/13/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Epidemiological studies have demonstrated a strong association of ambient fine particulate matter (PM2.5) exposure with the increasing mortality by ischemic heart disease (IHD), but the involved mechanisms remain poorly understood. Herein, we found that the chronic exposure of real ambient PM2.5 led to the upregulation of hypoxia-inducible factor-1 alpha (HIF-1α) protein in the myocardium of mice, accompanied by obvious myocardial injury and hypertrophy. Further data from the hypoxia-ischemia cellular model indicated that PM2.5-induced HIF-1α accumulation was responsible for the promotion of myocardial hypoxia injury. Moreover, the declined ATP level due to the HIF-1α-mediated energy metabolism remodeling from β-oxidation to glycolysis had a critical role in the PM2.5-increased myocardial hypoxia injury. The in-depth analysis delineated that PM2.5 exposure decreased the binding of prolyl hydroxylase domain 2 (PHD2) and HIF-1α and subsequent ubiquitin protease levels, thereby leading to the accumulation of HIF-1α. Meanwhile, factor-inhibiting HIF1 (FIH1) expression was down-regulated by PM2.5, resulting in the enhanced translocation of HIF-1α to the nucleus. Overall, our study provides valuable insight into the regulatory role of oxygen sensor-mediated HIF-1α stabilization and translocation in PM-exacerbated myocardial hypoxia injury, we suggest this adds significantly to understanding the mechanisms of haze particles-caused burden of cardiovascular disease.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Liu Wu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Tenglong Cui
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | | | - Haiyi Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yanhong Wei
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuxin Zheng
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
22
|
TKTL1 Knockdown Impairs Hypoxia-Induced Glucose-6-phosphate Dehydrogenase and Glyceraldehyde-3-phosphate Dehydrogenase Overexpression. Int J Mol Sci 2022; 23:ijms23073574. [PMID: 35408935 PMCID: PMC8999113 DOI: 10.3390/ijms23073574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/02/2022] Open
Abstract
Increased expression of transketolase (TKT) and its isoform transketolase-like-1 (TKTL1) has been related to the malignant leukemia phenotype through promoting an increase in the non-oxidative branch of the pentose phosphate pathway (PPP). Recently, it has also been described that TKTL1 can have a role in survival under hypoxic conditions and in the acquisition of radio resistance. However, TKTL1’s role in triggering metabolic reprogramming under hypoxia in leukemia cells has never been characterized. Using THP-1 AML cells, and by combining metabolomics and transcriptomics techniques, we characterized the impact of TKTL1 knockdown on the metabolic reprogramming triggered by hypoxia. Results demonstrated that TKTL1 knockdown results in a decrease in TKT, glucose-6-phosphate dehydrogenase (G6PD) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activities and impairs the hypoxia-induced overexpression of G6PD and GAPDH, all having significant impacts on the redox capacity of NADPH- and NADH-related cells. Moreover, TKTL1 knockdown impedes hypoxia-induced transcription of genes encoding key enzymes and transporters involved in glucose, PPP and amino acid metabolism, rendering cells unable to switch to enhanced glycolysis under hypoxia. Altogether, our results show that TKTL1 plays a key role in the metabolic adaptation to hypoxia in THP-1 AML cells through modulation of G6PD and GAPDH activities, both regulating glucose/glutamine consumption and the transcriptomic overexpression of key players of PPP, glucose and amino acids metabolism.
Collapse
|
23
|
A “Weird” Mitochondrial Fatty Acid Oxidation as a Metabolic “Secret” of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2339584. [PMID: 35178152 PMCID: PMC8847026 DOI: 10.1155/2022/2339584] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022]
Abstract
Cancer metabolism is an extensively studied field since the discovery of the Warburg effect about 100 years ago and continues to be increasingly intriguing and enigmatic so far. It has become clear that glycolysis is not the only abnormally activated metabolic pathway in the cancer cells, but the same is true for the fatty acid synthesis (FAS) and mevalonate pathway. In the last decade, a lot of data have been accumulated on the pronounced mitochondrial fatty acid oxidation (mFAO) in many types of cancer cells. In this article, we discuss how mFAO can escape normal regulation under certain conditions and be overactivated. Such abnormal activation of mitochondrial β-oxidation can also be combined with mutations in certain enzymes of the Krebs cycle that are common in cancer. If overactivated β-oxidation is combined with other common cancer conditions, such as dysfunctions in the electron transport complexes, and/or hypoxia, this may alter the redox state of the mitochondrial matrix. We propose the idea that the altered mitochondrial redox state and/or inhibited Krebs cycle at certain segments may link mitochondrial β-oxidation to the citrate-malate shuttle instead to the Krebs cycle. We call this abnormal metabolic condition “β-oxidation shuttle”. It is unconventional mFAO, a separate metabolic pathway, unexplored so far as a source of energy, as well as a source of cataplerosis, leading to biomass accumulation, accelerated oxygen consumption, and ultimately a source of proliferation. It is inefficient as an energy source and must consume significantly more oxygen per mole of ATP produced when combined with acetyl-CoA consuming pathways, such as the FAS and mevalonate pathway.
Collapse
|
24
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Ma H, Zhang Y, Prasad NR, Singh N, Fu F, Pei JM, Sarybaev A, Sydykov A. The effect of adaptation to hypoxia on cardiac tolerance to ischemia/reperfusion. J Biomed Res 2022:1-25. [PMID: 37183617 PMCID: PMC10387748 DOI: 10.7555/jbr.36.20220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The acute myocardial infarction (AMI) and sudden cardiac death (SCD), both associated with acute cardiac ischemia, are one of the leading causes of adult death in economically developed countries. The development of new approaches for the treatment and prevention of AMI and SCD remains the highest priority for medicine. A study on the cardiovascular effects of chronic hypoxia (CH) may contribute to the development of these methods. Chronic hypoxia exerts both positive and adverse effects. The positive effects are the infarct-reducing, vasoprotective, and antiarrhythmic effects, which can lead to the improvement of cardiac contractility in reperfusion. The adverse effects are pulmonary hypertension and right ventricular hypertrophy. This review presents a comprehensive overview of how CH enhances cardiac tolerance to ischemia/reperfusion. It is an in-depth analysis of the published data on the underlying mechanisms, which can lead to future development of the cardioprotective effect of CH. A better understanding of the CH-activated protective signaling pathways may contribute to new therapeutic approaches in an increase of cardiac tolerance to ischemia/reperfusion.
Collapse
|
25
|
He S, He S, Yang Y, Li B, Gao L, Xie Q, Zhang L. Correlation Between Neutrophil to Lymphocyte Ratio and Myocardial Injury in Population Exposed to High Altitude. Front Cardiovasc Med 2021; 8:738817. [PMID: 34881301 PMCID: PMC8645565 DOI: 10.3389/fcvm.2021.738817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Objective: Myocardial injury is a severe complication in population exposed to high altitude. As a new biomarker for inflammatory response, neutrophil to lymphocyte ratio (NLR) has been widely used to predict the prognosis of various diseases. In this study, we intend to explore the risk factors for myocardial injury at high altitude and examine the relationship between NLR level and development of myocardial injury. Methods: Consecutive patients admitted to a secondary general hospital at high altitude from June 2019 to May 2020 were selected into this retrospective study. Clinical and biochemical data were collected. According to the results of lactate dehydrogenase (LDH), creatine kinase (CK), creatine kinase isoenzymes (CK-MB), and aspartate amino transferase (AST), patients were divided into myocardial injury group and normal group. Results: A total of 476 patients were enrolled in this study. Myocardial injury occurred in 158 patients (33.2%). We found that altitude, NLR, hemoglobin, total bilirubin, total cholesterol, and lipoprotein A in myocardial injury group were significantly higher than that in normal group (P < 0.05), while platelet count in myocardial injury group was significantly lower than that in normal group (P < 0.05). Logistic multivariate regression analysis revealed that there was an independent relationship between myocardial injury and smoke, NLR, hemoglobin (P < 0.05). By using Spearman correlation analysis, NLR was proved to have a significant positive correlation with LDH, CK, and CK-MB (P < 0.05) instead of AST. A receiver operating characteristic (ROC) curve was drawn to demonstrate that NLR could significantly predict the occurrence of myocardial injury with an area under the curve (AUC) of 0.594 (95% CI: 0.537–0.650, P < 0.05), and the level of 2.967 (sensitivity = 38.0%, specificity = 83.6%) was optimal cutoff value. Conclusion: The incidence of myocardial injury is high in population at high altitude. Smoke, hemoglobin, and NLR are independent factors related to myocardial injury. As a convenient and efficient marker, NLR is found to be closely associated with myocardial enzymes and have a predict role in the occurrence of myocardial injury. This study will provide a theoretical basis on NLR for the early diagnosis of myocardial injury at high altitude.
Collapse
Affiliation(s)
- Siyi He
- General Hospital of Western Theater Command, Chengdu, China
| | - Shengdong He
- General Hospital of Western Theater Command, Chengdu, China
| | - Yongxiang Yang
- General Hospital of Western Theater Command, Chengdu, China
| | - Bin Li
- Military Prevention and Control Center for Mountain Sickness, No. 950 Hospital of the Chinese People's Liberation Army, Yecheng, China
| | - Liang Gao
- Military Prevention and Control Center for Mountain Sickness, No. 950 Hospital of the Chinese People's Liberation Army, Yecheng, China
| | - Qingyun Xie
- General Hospital of Western Theater Command, Chengdu, China
| | - Lin Zhang
- General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
26
|
Sen S, Hallee L, Lam CK. The Potential of Gamma Secretase as a Therapeutic Target for Cardiac Diseases. J Pers Med 2021; 11:jpm11121294. [PMID: 34945766 PMCID: PMC8703931 DOI: 10.3390/jpm11121294] [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: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Heart diseases are some of the most common and pressing threats to human health worldwide. The American Heart Association and the National Institute of Health jointly work to annually update data on cardiac diseases. In 2018, 126.9 million Americans were reported as having some form of cardiac disorder, with an estimated direct and indirect total cost of USD 363.4 billion. This necessitates developing therapeutic interventions for heart diseases to improve human life expectancy and economic relief. In this review, we look into gamma-secretase as a potential therapeutic target for cardiac diseases. Gamma-secretase, an aspartyl protease enzyme, is responsible for the cleavage and activation of a number of substrates that are relevant to normal cardiac development and function as found in mutation studies. Some of these substrates are involved in downstream signaling processes and crosstalk with pathways relevant to heart diseases. Most of the substrates and signaling events we explored were found to be potentially beneficial to maintain cardiac function in diseased conditions. This review presents an updated overview of the current knowledge on gamma-secretase processing of cardiac-relevant substrates and seeks to understand if the modulation of gamma-secretase activity would be beneficial to combat cardiac diseases.
Collapse
Affiliation(s)
- Sujoita Sen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Logan Hallee
- Department of Mathematical Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Correspondence: ; Tel.: +1-302-831-3165
| |
Collapse
|