1
|
Di Loria A, Ferravante C, D'Agostino Y, Giurato G, Tursi M, Grego E, Perego M, Weisz A, Ciaramella P, Santilli R. Gene-expression profiling of endomyocardial biopsies from dogs with dilated cardiomyopathy phenotype. J Vet Cardiol 2024; 52:78-89. [PMID: 38508121 DOI: 10.1016/j.jvc.2024.02.008] [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: 02/22/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION The employment of advanced molecular biology technologies has expanded the diagnostic investigation of cardiomyopathies in dogs; these technologies have predominantly been performed on postmortem samples, although the recent use of endomyocardial biopsy in living dogs has enabled a better premortem diagnostic approach to study the myocardial injury. ANIMALS, MATERIALS, AND METHODS Endomyocardial biopsies were collected in nine dogs with a dilated cardiomyopathy phenotype (DCM-p) and congestive heart failure and submitted to histologic examination, next-generation sequencing (NGS), and polymerase chain reaction analysis. Data from three healthy dogs (Fastq files) were retrieved from a previously approved study and used as a control group for ribonucleic acid sequencing. RESULTS Histologic examination revealed endocardial fibrosis in 6 of 9 dogs, whereas lymphocytic interstitial infiltrates were detected in 2 of 9 dogs, and lymphoplasmacytic and macrophage infiltrates were detected in 1 of 9 dogs. On polymerase chain reaction analysis, two dogs tested positive for canine parvovirus 2 and one dog for canine distemper virus. Gene-expression pathways involved in cellular energy metabolism (especially carbohydrates-insulin) and cardiac structural proteins were different in all DCM-p dogs compared to those in the control group. When dogs with lymphocytic interstitial infiltrates were compared to those in the control group, NGS analysis revealed the predominant role of genes related to inflammation and pathogen infection. CONCLUSIONS NGS technology performed on in vivo endomyocardial biopsies has identified different molecular and genetic factors that could play a role in the development and/or progression of DCM-p in dogs.
Collapse
Affiliation(s)
- A Di Loria
- Department of Veterinary Medicine and Animal Productions, University Federico II, Napoli, 80130, Italy
| | - C Ferravante
- Department of Veterinary Medicine and Animal Productions, University Federico II, Napoli, 80130, Italy; Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, SA, Italy; Medical Genomics Program, AOU 'SS. Giovanni di Dio e Ruggi d'Aragona', University of Salerno, 84131 Salerno, Italy
| | - Y D'Agostino
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, SA, Italy; Medical Genomics Program, AOU 'SS. Giovanni di Dio e Ruggi d'Aragona', University of Salerno, 84131 Salerno, Italy
| | - G Giurato
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, SA, Italy; Genome Research Center for Health, Campus of Medicine, University of Salerno, 84081 Baronissi, Italy
| | - M Tursi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| | - E Grego
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| | - M Perego
- Clinica Veterinaria Malpensa, Viale Marconi 27, Samarate, 21017 Varese, Italy
| | - A Weisz
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, SA, Italy; Medical Genomics Program, AOU 'SS. Giovanni di Dio e Ruggi d'Aragona', University of Salerno, 84131 Salerno, Italy; Genome Research Center for Health, Campus of Medicine, University of Salerno, 84081 Baronissi, Italy
| | - P Ciaramella
- Department of Veterinary Medicine and Animal Productions, University Federico II, Napoli, 80130, Italy.
| | - R Santilli
- Clinica Veterinaria Malpensa, Viale Marconi 27, Samarate, 21017 Varese, Italy; Department of Clinical Sciences, Cornell University, 930, Campus Road, 14853, Ithaca, NY, USA
| |
Collapse
|
2
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. Data-Independent Acquisition Proteomics and N-Terminomics Methods Reveal Alterations in Mitochondrial Function and Metabolism in Ischemic-Reperfused Hearts. J Proteome Res 2024; 23:844-856. [PMID: 38264990 PMCID: PMC10846531 DOI: 10.1021/acs.jproteome.3c00754] [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: 11/11/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
Myocardial ischemia-reperfusion (IR) (stunning) injury triggers changes in the proteome and degradome of the heart. Here, we utilize quantitative proteomics and comprehensive degradomics to investigate the molecular mechanisms of IR injury in isolated rat hearts. The control group underwent aerobic perfusion, while the IR injury group underwent 20 min of ischemia and 30 min of reperfusion to induce a stunning injury. As MMP-2 activation has been shown to contribute to myocardial injury, hearts also underwent IR injury with ARP-100, an MMP-2-preferring inhibitor, to dissect the contribution of MMP-2 to IR injury. Using data-independent acquisition (DIA) and mass spectroscopy, we quantified 4468 proteins in ventricular extracts, whereby 447 proteins showed significant alterations among the three groups. We then used subtiligase-mediated N-terminomic labeling to identify more than a hundred specific cleavage sites. Among these protease substrates, 15 were identified following IR injury. We identified alterations in numerous proteins involved in mitochondrial function and metabolism following IR injury. Our findings provide valuable insights into the biochemical mechanisms of myocardial IR injury, suggesting alterations in reactive oxygen/nitrogen species handling and generation, fatty acid metabolism, mitochondrial function and metabolism, and cardiomyocyte contraction.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
3
|
Gurler B, Gencay G, Baloglu E. Hypoxia and HIF-1α Regulate the Activity and Expression of Na,K-ATPase Subunits in H9c2 Cardiomyoblasts. Curr Issues Mol Biol 2023; 45:8277-8288. [PMID: 37886965 PMCID: PMC10605391 DOI: 10.3390/cimb45100522] [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: 09/23/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The optimal function of the Na,K-ATPase (NKA) pump is essential for the heart. In ischemic heart disease, NKA activity decreases due to the decreased expression of the pump subunits. Here, we tested whether the hypoxia-inducible transcription factor (HIF-1α), the key signaling molecule regulating the adaptation of cells to hypoxia, is involved in controlling the expression and cellular dynamics of α1- and β1-NKA isoforms and of NKA activity in in-vitro hypoxic H9c2 cardiomyoblasts. HIF-1α was silenced through adenoviral infection, and cells were kept in normoxia (19% O2) or hypoxia (1% O2) for 24 h. We investigated the mRNA and protein expression of α1-, β1-NKA using RT-qPCR and Western blot in whole-cell lysates, cell membranes, and cytoplasmic fractions after labeling the cell surface with NHS-SS-biotin and immunoprecipitation. NKA activity and intracellular ATP levels were also measured. We found that in hypoxia, silencing HIF-1α prevented the decreased mRNA expression of α1-NKA but not of β1-NKA. Hypoxia decreased the plasma membrane expression of α1-NKA and β1- NKA compared to normoxic cells. In hypoxic cells, HIF-1α silencing prevented this effect by inhibiting the internalization of α1-NKA. Total protein expression was not affected. The decreased activity of NKA in hypoxic cells was fully prevented by silencing HIF-1α independent of cellular ATP levels. This study is the first to show that in hypoxic H9c2 cardiomyoblasts, HIF-1α controls the internalization and membrane insertion of α1-NKA subunit and of NKA activity. The mechanism behind this regulation needs further investigation.
Collapse
Affiliation(s)
- Beyza Gurler
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Gizem Gencay
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| |
Collapse
|
4
|
Wu HX, He PM, Jia R. Effects of µ-Conotoxin GIIIB on the cellular activity of mouse skeletal musculoblast: combined transcriptome and proteome analysis. Proteome Sci 2023; 21:17. [PMID: 37828502 PMCID: PMC10568904 DOI: 10.1186/s12953-023-00221-w] [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: 06/25/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
µ-Conotoxin GIIIB (µ-CTX GIIIB) is a polypeptide containing three disulfide bridges, produced by the sea snail Conus geographus. This study was aimed to explored the cytotoxic effects of µ-CTX GIIIB on mouse skeletal musculoblast (Sol8). Sol8 cells were exposed to ouabain and veratridine to establish the cell injury model, and then treated with µ-CTX GIIIB. CCK-8 was adopted to evaluate the cytotoxicity of µ-CTX GIIIB. Then, proteomics and transcriptome were conducted, and the explore the differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) affected by µ-CTX GIIIB were found. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was used to investigate the affected signaling pathways. µ-CTX GIIIB increased the cell survival rate of injured Sol8 cells. We found and identified 1,663 DEGs and 444 DEPs influenced by µ-CTX GIIIB. 106 pairs of correlated DEGs and DEPs were selected by combining transcriptome and proteome data. The results of KEGG and GO analysis showed that µ-CTX GIIB affected the cell cycle, apoptosis, DNA damage and repair, lipid metabolism and other biological processes of Sol8 cells. µ-CTX GIIIB could affected cell cycle regulation, DNA damage repair, and activation of tumor factors, with potential carcinogenic effects. Our results provide an important basis for the study of in vitro toxicity, the mechanism of toxicity and injury prevention by µ-CTX GIIIB.
Collapse
Affiliation(s)
- Han-Xi Wu
- College of Marine Ecology and Environment, Shanghai Ocean University, No.999, Huchenghuan Rd, Nanhui New City, Shanghai, 201306, P.R. China
| | - Pei-Min He
- College of Marine Ecology and Environment, Shanghai Ocean University, No.999, Huchenghuan Rd, Nanhui New City, Shanghai, 201306, P.R. China
| | - Rui Jia
- College of Marine Ecology and Environment, Shanghai Ocean University, No.999, Huchenghuan Rd, Nanhui New City, Shanghai, 201306, P.R. China.
| |
Collapse
|
5
|
Kutz LC, Cui X, Xie JX, Mukherji ST, Terrell KC, Huang M, Wang X, Wang J, Martin AJ, Pessoa MT, Cai L, Zhu H, Heiny JA, Shapiro JI, Blanco G, Xie Z, Pierre SV. The Na/K-ATPase α1/Src interaction regulates metabolic reserve and Western diet intolerance. Acta Physiol (Oxf) 2021; 232:e13652. [PMID: 33752256 PMCID: PMC8570534 DOI: 10.1111/apha.13652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
AIM Highly prevalent diseases such as insulin resistance and heart failure are characterized by reduced metabolic flexibility and reserve. We tested whether Na/K-ATPase (NKA)-mediated regulation of Src kinase, which requires two NKA sequences specific to the α1 isoform, is a regulator of metabolic capacity that can be targeted pharmacologically. METHODS Metabolic capacity was challenged functionally by Seahorse metabolic flux analyses and glucose deprivation in LLC-PK1-derived cells expressing Src binding rat NKA α1, non-Src-binding rat NKA α2 (the most abundant NKA isoform in the skeletal muscle), and Src binding gain-of-function mutant rat NKA α2. Mice with skeletal muscle-specific ablation of NKA α1 (skα1-/-) were generated using a MyoD:Cre-Lox approach and were subjected to treadmill testing and Western diet. C57/Bl6 mice were subjected to Western diet with or without pharmacological inhibition of NKA α1/Src modulation by treatment with pNaKtide, a cell-permeable peptide designed by mapping one of the sites of NKA α1/Src interaction. RESULTS Metabolic studies in mutant cell lines revealed that the Src binding regions of NKA α1 are required to maintain metabolic reserve and flexibility. Skα1-/- mice had decreased exercise endurance and mitochondrial Complex I dysfunction. However, skα1-/- mice were resistant to Western diet-induced insulin resistance and glucose intolerance, a protection phenocopied by pharmacological inhibition of NKA α1-mediated Src regulation with pNaKtide. CONCLUSIONS These results suggest that NKA α1/Src regulatory function may be targeted in metabolic diseases. Because Src regulatory capability by NKA α1 is exclusive to endotherms, it may link the aerobic scope hypothesis of endothermy evolution to metabolic dysfunction.
Collapse
Affiliation(s)
- Laura C Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jeffrey X. Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Shreya T Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Kayleigh C Terrell
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Minqi Huang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Adam J Martin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Marco T Pessoa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Hua Zhu
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
6
|
Wang J, Wang X, Gao Y, Lin Z, Chen J, Gigantelli J, Shapiro JI, Xie Z, Pierre SV. Stress Signal Regulation by Na/K-ATPase As a New Approach to Promote Physiological Revascularization in a Mouse Model of Ischemic Retinopathy. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 33275652 PMCID: PMC7718810 DOI: 10.1167/iovs.61.14.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The identification of target pathways to block excessive angiogenesis while simultaneously restoring physiological vasculature is an unmet goal in the therapeutic management of ischemic retinopathies. pNaKtide, a cell-permeable peptide that we have designed by mapping the site of α1 Na/K-ATPase (NKA)/Src binding, blocks the formation of α1 NKA/Src/reactive oxygen species (ROS) amplification loops and restores physiological ROS signaling in a number of oxidative disease models. The aim of this study was to evaluate the importance of the NKA/Src/ROS amplification loop and the effect of pNaKtide in experimental ischemic retinopathy. Methods Human retinal microvascular endothelial cells (HRMECs) and retinal pigment epithelium (ARPE-19) cells were used to evaluate the effect of pNaKtide on viability, proliferation, and angiogenesis. Retinal toxicity and distribution were assessed in those cells and in the mouse. Subsequently, the role and molecular mechanism of NKA/Src in ROS stress signaling were evaluated biochemically in the retinas of mice exposed to the well-established protocol of oxygen-induced retinopathy (OIR). Finally, pNaKtide efficacy was assessed in this model. Results The results suggest a key role of α1 NKA in the regulation of ROS stress and the Nrf2 pathway in mouse OIR retinas. Inhibition of α1 NKA/Src by pNaKtide reduced pathologic ROS signaling and restored normal expression of hypoxia-inducible factor 1-α/vascular endothelial growth factor (VEGF). Unlike anti-VEGF agents, pNaKtide did promote retinal revascularization while inhibiting neovascularization and inflammation. Conclusions Targeting α1 NKA represents a novel strategy to develop therapeutics that not only inhibit neovascularization but also promote physiological revascularization in ischemic eye diseases.
Collapse
Affiliation(s)
- Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Zhucheng Lin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James Gigantelli
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Joseph I Shapiro
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
7
|
Qu X, Zhang Z, Hu W, Lou M, Zhai B, Mei S, Hu Z, Zhang L, Liu D, Liu Z, Chen J, Wang Y. Attenuation of the Na/K‑ATPase/Src/ROS amplification signaling pathway by astaxanthin ameliorates myocardial cell oxidative stress injury. Mol Med Rep 2020; 22:5125-5134. [PMID: 33173978 PMCID: PMC7646965 DOI: 10.3892/mmr.2020.11613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/27/2020] [Indexed: 12/27/2022] Open
Abstract
The 3S, 3′S-ASTaxanthin (3S, 3′S-AST) isomer has strong antioxidant activity; however, its protective roles and potential mechanisms against oxidative stress damage in cardiomyocytes have not been investigated. Na+/K+-ATPase (NKA)/Src signal activation has an important role in increasing reactive oxygen species (ROS) production. The aim of the present study was to investigate the protective effects and mechanism of 3S, 3′S-AST on hydrogen peroxide (H2O2)-induced oxidative stress injury in H9c2 myocardial cells. The protective effects of 3S, 3′S-AST on H2O2-induced H9c2 cell injury was observed by measuring lactate dehydrogenase and creatine kinase myocardial band content, cell viability and nuclear morphology. The antioxidant effect was investigated by analyzing ROS accumulation and malondialdehyde, glutathione (GSH) peroxidase, GSH and glutathione reductase activity levels. The protein expression levels of Bax, Bcl-2, caspase-3 and cleaved caspase-3 were analyzed using western blotting to determine cardiomyocyte apoptosis. Western blot analysis of the phosphorylation levels of Src and Erk1/2 were also performed to elucidate the molecular mechanism involved. The results showed that 3S, 3′S-AST reduced the release of LDH and promoted cell viability, and attenuated ROS accumulation and cell apoptosis induced by H2O2. Furthermore, 3S, 3′S-AST also restored apoptosis-related Bax and Bcl-2 protein expression levels in H2O2-treated H9c2 cells. The phosphorylation levels of Src and Erk1/2 were significantly higher in the H2O2 treatment group, whereas 3S, 3′S-AST pretreatment significantly decreased the levels of phosphorylated (p)-Src and p-ERK1/2. The results provided evidence that 3S, 3′S-AST exhibited a cardioprotective effect against oxidative stress injury by attenuating NKA/Src/Erk1/2-modulated ROS amplification.
Collapse
Affiliation(s)
- Xuefeng Qu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhouyi Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Wenli Hu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Minhan Lou
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Bingzhong Zhai
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Song Mei
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhihang Hu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Lijing Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Dongying Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhen Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Jianguo Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Yin Wang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
8
|
Lowry CA, Golod ME, Andrew RD, Bennett BM. Expression of Neuronal Na +/K +-ATPase α Subunit Isoforms in the Mouse Brain Following Genetically Programmed or Behaviourally-induced Oxidative Stress. Neuroscience 2020; 442:202-215. [PMID: 32653541 DOI: 10.1016/j.neuroscience.2020.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/21/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
The Na+/K+-ATPase is a transmembrane ion pump that has a critical homeostatic role within every mammalian cell; however, it is vulnerable to the effects of increased oxidative stress. Understanding how expression of this transporter is influenced by oxidative stress may yield insight into its role in the pathophysiology of neurological and neuropsychiatric diseases. In this study we investigated whether increased oxidative stress could influence Na+/K+-ATPase expression in various brain regions of mice. We utilized two different models of oxidative stress: a behavioural chronic unpredictable stress protocol and the Aldh2-/- mouse model of oxidative stress-based and age-related cognitive impairment. We identified distinct regional baseline mRNA and protein expression patterns of the Na+/K+-ATPase α1 and α3 isoforms within the neocortex, hippocampus, and brainstem of wildtype mice. Consistent with previous studies, there was a higher proportion of α3 expression relative to α1 in the brainstem versus neocortex, but a higher proportion of α1 expression relative to α3 in the neocortex versus the brainstem. The hippocampus had similar expression levels of both α1 and α3. Despite increased staining for oxidative stress in higher brain, no differences in α1 or α3 expression were noted in Aldh2-/- mice versus wildtype, or in mice exposed to a 28-day chronic unpredictable stress protocol. In both models of oxidative stress, gene and protein expression of Na+/K+-ATPase α1 and α3 isoforms within the higher and lower brain was remarkably stable. Thus, Na+/K+-ATPase function previously reported as altered by oxidative stress is not through induced changes in the expression of pump isoforms.
Collapse
Affiliation(s)
- Chloe A Lowry
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - Michael E Golod
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - R David Andrew
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada; Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - Brian M Bennett
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada; Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
9
|
Qi Y, Yang C, Jiang Z, Wang Y, Zhu F, Li T, Wan X, Xu Y, Xie Z, Li D, Pierre SV. Epicatechin-3-Gallate Signaling and Protection against Cardiac Ischemia/Reperfusion Injury. J Pharmacol Exp Ther 2019; 371:663-674. [DOI: 10.1124/jpet.119.260117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
|
10
|
Abstract
Ouabain preconditioning (OPC) initiated by low concentrations of the cardiac glycoside (CG) ouabain binding to Na/K-ATPase is relayed by a unique intracellular signaling and protects cardiac myocytes against ischemia/reperfusion injury. To explore more clinically applicable protocols based on CG properties, we tested whether the FDA-approved CG digoxin could trigger cardioprotective effects comparable with those of ouabain using PC, preconditioning and PostC, postconditioning protocols in the Langendorff-perfused mouse heart subjected to global ischemia and reperfusion. Ouabain or digoxin at 10 μmol/L inhibited Na/K-ATPase activity by approximately 30% and activated PKCε translocation by approximately 50%. Digoxin-induced PC (DigPC), initiated by a transient exposure before 40 minutes of ischemia, was as effective as OPC as suggested by the recovery of left ventricular developed pressure, end-diastolic pressure, and cardiac Na/K-ATPase activity after 30 minutes of reperfusion. DigPC also significantly decreased lactate dehydrogenase release and reduced infarct size, comparable with OPC. PostC protocols consisting of a single bolus injection of 100 nmoles of ouabain or digoxin in the coronary tree at the beginning of reperfusion both improved significantly the recovery of left ventricular developed pressure and decreased lactate dehydrogenase release, demonstrating a functional and structural protection comparable with the one provided by OPC. Given the unique signaling triggered by OPC, these results suggest that DigPostC could be considered for patients with risk factors and/or concurrent treatments that may limit effectiveness of ischemic PostC.
Collapse
|
11
|
Kumaş M, Eşrefoğlu M, Karataş E, Duymaç N, Kanbay S, Ergün IS, Üyüklü M, Koçyiğit A. Investigation of dose-dependent effects of berberine against renal ischemia/reperfusion injury in experimental diabetic rats. Nefrologia 2019; 39:411-423. [PMID: 30712966 DOI: 10.1016/j.nefro.2018.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/23/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Ischemia-reperfusion injury causes various severe morphological and functional changes in diabetic patients. To date, numerous antidiabetic and antioxidant agents have been used for treatment of the disease-related changes. OBJECTIVES We aimed to examine effective therapeutic doses or doses of berberine against renal ischemia/reperfusion injury (IRI) in a streptozotocin (STZ)-induced diabetic rat model by histopathological and biochemical analysis. METHODS Thirty male Sprague Dawley rats were treated with STZ injection for the development of diabetes, and divided into the following groups: STZ-induced diabetic group (STZ); IRI-induced diabetic group (STZ+IRI); 50mg/kg berberine (BRB) treated diabetic group after inducing IRI (STZ+IRI+BRB1); 100mg/kg BRB treated diabetic group after IRI (STZ+IRI+BRB2); 150mg/kg BRB treated diabetic group after IRI (STZ+IRI+BRB3). Bilateral renal ischemia model was applied for 45min, then reperfusion was allowed for 14 days in STZ-induced diabetic rats. Renal injury was detected histopathologically. Blood urea nitrogen (BUN), creatinine and lactate dehydrogenase (LDH) levels were measured in serum using the ELISA method. Total antioxidant status (TAS) and total oxidant status (TOS) of renal tissue was studied by spectrophotometric assay. Oxidative stress index (OSI) was calculated as TOS-to-TAS ratio. Tumor necrosis factor alpha (TNF-α), C-reactive protein (CRP), Na+/K+-ATPase (sodium pump), and Ca2+-ATPase (calcium ATPase) enzyme levels were measured in tissues using the ELISA method. Anti-apoptotic Bax and pro-apoptotic Bcl-2 protein levels were detected by Western blot analysis. All data were evaluated statistically. RESULTS The highest histopathological score was detected in the STZ+IRI group compared to the other group. BRB administration at the doses of 100mg/kg and 150mg/kg markedly improved renal injury. BUN and creatinine levels significantly increased in the STZ+IRI group compared to the STZ group (p<0.001). 100mg/kg and 150mg/kg BRB administration significantly decreased those levels (p<0.01). The highest TOS and the lowest TAS levels were detected in the STZ+IRI group (p<0.001). IRI markedly aggravated inflammation via increasing levels of TNF-α and CRP (<0.001), and caused apoptosis via inducing Bcl-2 protein, and suppressing Bax protein (p<0.001). BRB administration at the doses of 100mg/kg and 150mg/kg showed anti-oxidant, anti-inflammatory and anti-apoptotic effects (p<0.01). The LDH enzyme, was used as a necrosis marker, was higher in the STZ+IRI group than other groups. BRB administration at all of the doses, resulted in the decline of LDH enzyme level (p<0.001). Ca2+-ATPase and Na+/K+-ATPase enzyme activities decreased in the STZ+IRI group compared to the STZ group (p<0.001), while BRB administration at the doses of 100mg/kg and 150mg/kg significantly increased those of enzyme activities, respectively (p<0.05). CONCLUSION Ischemia with diabetes caused severe histopathological and biochemical damage in renal tissue. The high doses of berberine markedly improved histopathological findings, regulated kidney function via decreasing BUN and creatinine levels, and rearranged intercellular ion concentration via increasing Na+/K+-ATPase and Ca2+- ATPase levels. Berberine showed anti-oxidant, anti-apoptotic, and anti-inflammatory effects. According to these data, we suggest that berberine at the doses of 100 and 150mg may be used as a potential therapeutic agent to prevent renal ischemic injury.
Collapse
Affiliation(s)
- Meltem Kumaş
- Department of Histology and Embryology, Faculty of Medicine, Bezmialem Vakif University, 34093 Istanbul, Turkey.
| | - Mukaddes Eşrefoğlu
- Department of Histology and Embryology, Faculty of Medicine, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Ersin Karataş
- Department of Molecular Biology and Genetics, Gebze Technical University, Kocaeli, Turkey
| | - Nurcihan Duymaç
- Department of Pathology Laboratory Techniques, Vocational School of Health Services, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Songül Kanbay
- Department of Pathology Laboratory Techniques, Vocational School of Health Services, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Ilyas Samet Ergün
- Department of Pathology Laboratory Techniques, Vocational School of Health Services, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Mehmet Üyüklü
- Department of Physiology, Faculty of Medicine, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Abdurrahim Koçyiğit
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, 34093 Istanbul, Turkey
| |
Collapse
|
12
|
Marck PV, Pierre SV. Na/K-ATPase Signaling and Cardiac Pre/Postconditioning with Cardiotonic Steroids. Int J Mol Sci 2018; 19:ijms19082336. [PMID: 30096873 PMCID: PMC6121447 DOI: 10.3390/ijms19082336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The first reports of cardiac Na/K-ATPase signaling, published 20 years ago, have opened several major fields of investigations into the cardioprotective action of low/subinotropic concentrations of cardiotonic steroids (CTS). This review focuses on the protective cardiac Na/K-ATPase-mediated signaling triggered by low concentrations of ouabain and other CTS, in the context of the enduring debate over the use of CTS in the ischemic heart. Indeed, as basic and clinical research continues to support effectiveness and feasibility of conditioning interventions against ischemia/reperfusion injury in acute myocardial infarction (AMI), the mechanistic information available to date suggests that unique features of CTS-based conditioning could be highly suitable, alone /or as a combinatory approach.
Collapse
Affiliation(s)
- Pauline V Marck
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, WV 25701, USA.
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, WV 25701, USA.
| |
Collapse
|
13
|
Buzaglo N, Golomb M, Rosen H, Beeri R, Ami HCB, Langane F, Pierre S, Lichtstein D. Augmentation of Ouabain-Induced Increase in Heart Muscle Contractility by Akt Inhibitor MK-2206. J Cardiovasc Pharmacol Ther 2018; 24:78-89. [DOI: 10.1177/1074248418788301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac steroids (CSs), such as ouabain and digoxin, increase the force of contraction of heart muscle and are used for the treatment of congestive heart failure (CHF). However, their small therapeutic window limits their use. It is well established that Na+, K+-ATPase inhibition mediates CS-induced increase in heart contractility. Recently, the involvement of intracellular signal transduction was implicated in this effect. The aim of the present study was to test the hypothesis that combined treatment with ouabain and Akt inhibitor (MK-2206) augments ouabain-induced inotropy in mammalian models. We demonstrate that the combined treatment led to an ouabain-induced increase in contractility at concentrations at which ouabain alone was ineffective. This was shown in 3 experimental systems: neonatal primary rat cardiomyocytes, a Langendorff preparation, and an in vivo myocardial infarction induced by left anterior descending coronary artery (LAD) ligation. Furthermore, cell viability experiments revealed that this treatment protected primary cardiomyocytes from MK-2206 toxicity and in vivo reduced the size of scar tissue 10 days post-LAD ligation. We propose that Akt activity imposes a constant inhibitory force on muscle contraction, which is attenuated by low concentrations of MK-2206, resulting in potentiation of the ouabain effect. This demonstration of the increase in the CS effect advocates the development of the combined treatment in CHF.
Collapse
Affiliation(s)
- Nahum Buzaglo
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Mordechai Golomb
- The Heart Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haim Rosen
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Ronen Beeri
- The Heart Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hagit Cohen-Ben Ami
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Fattal Langane
- Marshall Institute for Interdisciplinary Research, Huntington, WV, USA
| | - Sandrine Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV, USA
| | - David Lichtstein
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
14
|
Ferdinandy P, Baczkó I, Bencsik P, Giricz Z, Görbe A, Pacher P, Varga ZV, Varró A, Schulz R. Definition of hidden drug cardiotoxicity: paradigm change in cardiac safety testing and its clinical implications. Eur Heart J 2018; 40:1771-1777. [PMID: 29982507 PMCID: PMC6554653 DOI: 10.1093/eurheartj/ehy365] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Unexpected cardiac adverse effects are the leading causes of discontinuation of clinical trials and withdrawal of drugs from the market. Since the original observations in the mid-90s, it has been well established that cardiovascular risk factors and comorbidities (such as ageing, hyperlipidaemia, and diabetes) and their medications (e.g. nitrate tolerance, adenosine triphosphate-dependent potassium inhibitor antidiabetic drugs, statins, etc.) may interfere with cardiac ischaemic tolerance and endogenous cardioprotective signalling pathways. Indeed drugs may exert unwanted effects on the diseased and treated heart that is hidden in the healthy myocardium. Hidden cardiotoxic effects may be due to (i) drug-induced enhancement of deleterious signalling due to ischaemia/reperfusion injury and/or the presence of risk factors and/or (ii) inhibition of cardioprotective survival signalling pathways, both of which may lead to ischaemia-related cell death and/or pro-arrhythmic effects. This led to a novel concept of ‘hidden cardiotoxicity’, defined as cardiotoxity of a drug that manifests only in the diseased heart with e.g. ischaemia/reperfusion injury and/or in the presence of its major comorbidities. Little is known on the mechanism of hidden cardiotoxocity, moreover, hidden cardiotoxicity cannot be revealed by the routinely used non-clinical cardiac safety testing methods on healthy animals or tissues. Therefore, here, we emphasize the need for development of novel cardiac safety testing platform involving combined experimental models of cardiac diseases (especially myocardial ischaemia/reperfusion and ischaemic conditioning) in the presence and absence of major cardiovascular comorbidities and/or cotreatments.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | | | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, Giessen, Germany
| |
Collapse
|
15
|
Kutz LC, Mukherji ST, Wang X, Bryant A, Larre I, Heiny JA, Lingrel JB, Pierre SV, Xie Z. Isoform-specific role of Na/K-ATPase α1 in skeletal muscle. Am J Physiol Endocrinol Metab 2018; 314:E620-E629. [PMID: 29438630 PMCID: PMC6032065 DOI: 10.1152/ajpendo.00275.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The distribution of Na/K-ATPase α-isoforms in skeletal muscle is unique, with α1 as the minor (15%) isoform and α2 comprising the bulk of the Na/K-ATPase pool. The acute and isoform-specific role of α2 in muscle performance and resistance to fatigue is well known, but the isoform-specific role of α1 has not been as thoroughly investigated. In vitro, we reported that α1 has a role in promoting cell growth that is not supported by α2. To assess whether α1 serves this isoform-specific trophic role in the skeletal muscle, we used Na/K-ATPase α1-haploinsufficient (α1+/-) mice. A 30% decrease of Na/K-ATPase α1 protein expression without change in α2 induced a modest yet significant decrease of 10% weight in the oxidative soleus muscle. In contrast, the mixed plantaris and glycolytic extensor digitorum longus weights were not significantly affected, likely because of their very low expression level of α1 compared with the soleus. The soleus mass reduction occurred without change in total Na/K-ATPase activity or glycogen metabolism. Serum analytes including K+, fat tissue mass, and exercise capacity were not altered in α1+/- mice. The impact of α1 content on soleus muscle mass is consistent with a Na/K-ATPase α1-specific role in skeletal muscle growth that cannot be fulfilled by α2. The preserved running capacity in α1+/- is in sharp contrast with previously reported consequences of genetic manipulation of α2. Taken together, these results lend further support to the concept of distinct isoform-specific functions of Na/K-ATPase α1 and α2 in skeletal muscle.
Collapse
Affiliation(s)
- Laura C Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Shreya T Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Amber Bryant
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| |
Collapse
|
16
|
Yan X, Xun M, Wu L, Du X, Zhang F, Zheng J. DRm217 attenuates myocardial ischemia-reperfusion injury via stabilizing plasma membrane Na + -K + -ATPase, inhibiting Na + -K + -ATPase/ROS pathway and activating PI3K/Akt and ERK1/2. Toxicol Appl Pharmacol 2018; 349:62-71. [DOI: 10.1016/j.taap.2018.04.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 11/17/2022]
|
17
|
Hu Y, Wang Z, Ge N, Huang T, Zhang M, Wang H. Sodium pump alpha-2 subunit (ATP1A2) alleviates cardiomyocyte anoxia-reoxygenation injury via inhibition of endoplasmic reticulum stress-related apoptosis. Can J Physiol Pharmacol 2018; 96:515-520. [PMID: 29394489 DOI: 10.1139/cjpp-2017-0349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have found decreased functional capacity of the sodium pump (Na+-K+-ATPase) alpha and beta subunits and recovery of Na+-K+-ATPase activity significantly decreased myocyte apoptosis in myocardial ischemia-reperfusion (I/R) injury. However, the potential role of the Na+-K+-ATPase α-2 subunit (ATP1A2) in cardiomyocyte anoxia-reoxygenation (A/R) injury has not been elucidated. Rat myocardial cells were subjected to siRNA transfection followed by A/R injury. Apoptosis and expression of endoplasmic reticulum (ER) stress proteins CHOP, GRP78, and caspase-12 were detected in 4 groups of cells: ATP1A2 siRNA + A/R, control siRNA + A/R, control, and A/R injury model. We found that apoptosis was significantly elevated in the ATP1A2 siRNA + A/R group as compared with control siRNA + A/R, control, and A/R injury model groups (p < 0.05, p < 0.01, and p < 0.05). Furthermore, expression of CHOP, GRP78, and caspase-12 were significantly elevated in the ATP1A2 siRNA + A/R group as compared with control siRNA + A/R, control, and A/R injury model groups (p < 0.05, p < 0.01, and p < 0.05). Our findings suggest that cardiomyocyte ATP1A2 is a target of A/R injury, and its cardioprotective function may be mediated via inhibiting the ER-stress-related apoptosis.
Collapse
Affiliation(s)
- Yulong Hu
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zheng Wang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Nannan Ge
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Ting Huang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Mingchao Zhang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Hegui Wang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China.,Department of Cardiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
18
|
Belliard A, Gulati GK, Duan Q, Alves R, Brewer S, Madan N, Sottejeau Y, Wang X, Kalisz J, Pierre SV. Ischemia/reperfusion-induced alterations of enzymatic and signaling functions of the rat cardiac Na+/K+-ATPase: protection by ouabain preconditioning. Physiol Rep 2017; 4:4/19/e12991. [PMID: 27702882 PMCID: PMC5064143 DOI: 10.14814/phy2.12991] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/12/2016] [Indexed: 11/24/2022] Open
Abstract
Cardiac glycosides (CG) are traditionally known as positive cardiac inotropes that inhibit Na+/K+‐ATPase‐dependent ion transport. CG also trigger‐specific signaling pathways through the cardiac Na+/K+‐ATPase, with beneficial effects in ischemia/reperfusion (I/R) injury (e.g., ouabain preconditioning, known as OPC) and hypertrophy. Our current understanding of hypersensitivity to CG and subsequent toxicity in the ischemic heart is mostly based on specific I/R‐induced alterations of the Na+/K+‐ATPase enzymatic function and has remained incomplete. The primary goal of this study was to investigate and compare the impact of I/R on Na+/K+‐ATPase enzymatic and signaling functions. Second, we assessed the impact of OPC on both functions. Langendorff‐perfused rat hearts were exposed to 30 min of ischemia and 30 min of reperfusion. At the inotropic concentration of 50 μmol/L, ouabain increased ERK and Akt phosphorylation in control hearts. In I/R hearts, this concentration did not induced positive inotropy and failed to induce Akt or ERK phosphorylation. The inotropic response to dobutamine as well as insulin signaling persisted, suggesting specific alterations of Na+/K+‐ATPase. Indeed, Na+/K+‐ATPase protein expression was intact, but the enzyme activity was decreased by 60% and the enzymatic function of the isoform with high affinity for ouabain was abolished following I/R. Strikingly, OPC prevented all I/R‐induced alterations of the receptor. Further studies are needed to reveal the respective roles of I/R‐induced modulations of Na+/K+‐ATPase enzymatic and signaling functions in cardiomyocyte death.
Collapse
Affiliation(s)
- Aude Belliard
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Gaurav K Gulati
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Qiming Duan
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Rosana Alves
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio Marshall Institute for Interdisciplinary Research, Huntington, West Virginia
| | - Shannon Brewer
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Namrata Madan
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia
| | - Yoann Sottejeau
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia
| | - Jennifer Kalisz
- Department of Biochemistry and Cancer Biology, College of Medicine, University of Toledo, Toledo, Ohio
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia
| |
Collapse
|
19
|
Petrushanko IY, Mitkevich VA, Lakunina VA, Anashkina AA, Spirin PV, Rubtsov PM, Prassolov VS, Bogdanov NB, Hänggi P, Fuller W, Makarov AA, Bogdanova A. Cysteine residues 244 and 458-459 within the catalytic subunit of Na,K-ATPase control the enzyme's hydrolytic and signaling function under hypoxic conditions. Redox Biol 2017; 13:310-319. [PMID: 28601781 PMCID: PMC5470536 DOI: 10.1016/j.redox.2017.05.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/16/2017] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
Our previous findings suggested that reversible thiol modifications of cysteine residues within the actuator (AD) and nucleotide binding domain (NBD) of the Na,K-ATPase may represent a powerful regulatory mechanism conveying redox- and oxygen-sensitivity of this multifunctional enzyme. S-glutathionylation of Cys244 in the AD and Cys 454-458-459 in the NBD inhibited the enzyme and protected cysteines' thiol groups from irreversible oxidation under hypoxic conditions. In this study mutagenesis approach was used to assess the role these cysteines play in regulation of the Na,K-ATPase hydrolytic and signaling functions. Several constructs of mouse α1 subunit of the Na,K-ATPase were produced in which Cys244, Cys 454-458-459 or Cys 244-454-458-459 were replaced by alanine. These constructs were expressed in human HEK293 cells. Non-transfected cells and those expressing murine α1 subunit were exposed to hypoxia or treated with oxidized glutathione (GSSG). Both conditions induced inhibition of the wild type Na,K-ATPase. Enzymes containing mutated mouse α1 lacking Cys244 or all four cysteines (Cys 244-454-458-459) were insensitive to hypoxia. Inhibitory effect of GSSG was observed for wild type murine Na,K-ATPase, but was less pronounced in Cys454-458-459Ala mutant and completely absent in the Cys244Ala and Cys 244-454-458-459Ala mutants. In cells, expressing wild type enzyme, ouabain induced activation of Src and Erk kinases under normoxic conditions, whereas under hypoxic conditions this effect was inversed. Cys454-458-459Ala substitution abolished Src kinase activation in response to ouabain treatment, uncoupled Src from Erk signaling, and interfered with O2-sensitivity of Na,K-ATPase signaling function. Moreover, modeling predicted that S-glutathionylation of Cys 458 and 459 should prevent inhibitory binding of Src to NBD. Our data indicate for the first time that cysteine residues within the AD and NBD influence hydrolytic as well as receptor function of the Na,K-ATPase and alter responses of the enzyme to hypoxia or upon treatment with cardiotonic steroids.
Collapse
Affiliation(s)
- Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Valentina A Lakunina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia A Anashkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Pavel V Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Peter M Rubtsov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir S Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay B Bogdanov
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Pascal Hänggi
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - William Fuller
- Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna Bogdanova
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
On the Many Actions of Ouabain: Pro-Cystogenic Effects in Autosomal Dominant Polycystic Kidney Disease. Molecules 2017; 22:molecules22050729. [PMID: 28467389 PMCID: PMC5688955 DOI: 10.3390/molecules22050729] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/30/2017] [Indexed: 02/07/2023] Open
Abstract
Ouabain and other cardenolides are steroidal compounds originally discovered in plants. Cardenolides were first used as poisons, but after finding their beneficial cardiotonic effects, they were rapidly included in the medical pharmacopeia. The use of cardenolides to treat congestive heart failure remained empirical for centuries and only relatively recently, their mechanisms of action became better understood. A breakthrough came with the discovery that ouabain and other cardenolides exist as endogenous compounds that circulate in the bloodstream of mammals. This elevated these compounds to the category of hormones and opened new lines of investigation directed to further study their biological role. Another important discovery was the finding that the effect of ouabain was mediated not only by inhibition of the activity of the Na,K-ATPase (NKA), but by the unexpected role of NKA as a receptor and a signal transducer, which activates a complex cascade of intracellular second messengers in the cell. This broadened the interest for ouabain and showed that it exerts actions that go beyond its cardiotonic effect. It is now clear that ouabain regulates multiple cell functions, including cell proliferation and hypertrophy, apoptosis, cell adhesion, cell migration, and cell metabolism in a cell and tissue type specific manner. This review article focuses on the cardenolide ouabain and discusses its various in vitro and in vivo effects, its role as an endogenous compound, its mechanisms of action, and its potential use as a therapeutic agent; placing especial emphasis on our findings of ouabain as a pro-cystogenic agent in autosomal dominant polycystic kidney disease (ADPKD).
Collapse
|
21
|
Friedenberg SG, Chdid L, Keene B, Sherry B, Motsinger-Reif A, Meurs KM. Use of RNA-seq to identify cardiac genes and gene pathways differentially expressed between dogs with and without dilated cardiomyopathy. Am J Vet Res 2017; 77:693-9. [PMID: 27347821 DOI: 10.2460/ajvr.77.7.693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To identify cardiac tissue genes and gene pathways differentially expressed between dogs with and without dilated cardiomyopathy (DCM). ANIMALS 8 dogs with and 5 dogs without DCM. PROCEDURES Following euthanasia, samples of left ventricular myocardium were collected from each dog. Total RNA was extracted from tissue samples, and RNA sequencing was performed on each sample. Samples from dogs with and without DCM were grouped to identify genes that were differentially regulated between the 2 populations. Overrepresentation analysis was performed on upregulated and downregulated gene sets to identify altered molecular pathways in dogs with DCM. RESULTS Genes involved in cellular energy metabolism, especially metabolism of carbohydrates and fats, were significantly downregulated in dogs with DCM. Expression of cardiac structural proteins was also altered in affected dogs. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested that RNA sequencing may provide important insights into the pathogenesis of DCM in dogs and highlight pathways that should be explored to identify causative mutations and develop novel therapeutic interventions.
Collapse
|
22
|
Zhao Q, Wu J, Hua Q, Lin Z, Ye L, Zhang W, Wu G, Du J, Xia J, Chu M, Hu X. Resolvin D1 mitigates energy metabolism disorder after ischemia-reperfusion of the rat lung. J Transl Med 2016; 14:81. [PMID: 27009328 PMCID: PMC4806414 DOI: 10.1186/s12967-016-0835-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/16/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Energy metabolism disorder is a critical process in lung ischemia-reperfusion injury (LIRI). This study was aimed to determine the effects of resolvin D1 (RvD1) on the energy metabolism in LIRI. METHODS Forty Sprague-Dawley rats were divided into the following groups: Sham group; untreated ischemia-reperfusion (IR) control; IR treated with normal saline (IR-NS); and IR treated with RvD1 (IR-RV) (100 μg/kg, iv). LIRI and energy metabolism disorder were determined in these rats. RESULTS The results revealed that the levels of interleukin (IL)-1β, tumor necrosis factor-α, IL-10, monocyte chemoattractant protein-1, macrophage inflammatory protein-2, cytokine-induced neutrophil chemoattractant-1, injured alveoli rate, apoptosis index, pulmonary permeability index, malondialdehyde, ADP, and lactic acid were increased, whereas the levels of ATP, ATP/ADP, glycogen, Na(+)-K(+)-ATPase, superoxide dismutase, glutathione peroxidase activity, pulmonary surfactant associated protein-A, and oxygenation index were decreased in rats with LIRI. Except for IL-10, all these biomarkers of LIRI and its related energy metabolism disorder were significantly inhibited by RvD1 treatment. In addition, histological analysis via hematoxylin-eosin staining, and transmission electron microscopy confirmed that IR-induced structure damages of lung tissues were reduced by RvD1. CONCLUSION RvD1 improves the energy metabolism of LIRI disturbance, protects the mitochondrial structure and function, increases the ATP, glycogen content and Na(+)-K(+)-ATPase activity of lung tissue, balances the ratio of ATP/ADP and finally decreases the rate of apoptosis, resulting in the protection of IR-induced lung injury. The improved energy metabolism after LIRI may be related to the reduced inflammatory response, the balance of the oxidative/antioxidant and the pro-inflammatory/anti-inflammatory systems in rats.
Collapse
Affiliation(s)
- Qifeng Zhao
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Ji Wu
- Wuhan Medical & Healthcare Center for Woman and Children, 430015, Wuhan, People's Republic of China
| | - Qingwang Hua
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Zhiyong Lin
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Leping Ye
- The Department of Children's Respiration Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Weixi Zhang
- The Department of Children's Respiration Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Guowei Wu
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Jie Du
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Jie Xia
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Maoping Chu
- The Department of Children's Cardiovascular Medicine, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Xingti Hu
- The Department of Children's Cardiovascular and Thoracic Surgery, Children's Heart Center, the Second Affiliated Hospital, Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, 325000, Wenzhou, People's Republic of China.
| |
Collapse
|
23
|
Role of phosphoinositide 3-kinase IA (PI3K-IA) activation in cardioprotection induced by ouabain preconditioning. J Mol Cell Cardiol 2015; 80:114-25. [PMID: 25575882 DOI: 10.1016/j.yjmcc.2014.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/06/2014] [Accepted: 12/26/2014] [Indexed: 11/23/2022]
Abstract
Acute myocardial infarction, the clinical manifestation of ischemia-reperfusion (IR) injury, is a leading cause of death worldwide. Like ischemic preconditioning (IPC) induced by brief episodes of ischemia and reperfusion, ouabain preconditioning (OPC) mediated by Na/K-ATPase signaling protects the heart against IR injury. Class I PI3K activation is required for IPC, but its role in OPC has not been investigated. While PI3K-IB is critical to IPC, studies have suggested that ouabain signaling is PI3K-IA-specific. Hence, a pharmacological approach was used to test the hypothesis that OPC and IPC rely on distinct PI3K-I isoforms. In Langendorff-perfused mouse hearts, OPC was initiated by 4 min of ouabain 10 μM and IPC was triggered by 4 cycles of 5 min ischemia and reperfusion prior to 40 min of global ischemia and 30 min of reperfusion. Without affecting PI3K-IB, ouabain doubled PI3K-IA activity and Akt phosphorylation at Ser(473). IPC and OPC significantly preserved cardiac contractile function and tissue viability as evidenced by left ventricular developed pressure and end-diastolic pressure recovery, reduced lactate dehydrogenase release, and decreased infarct size. OPC protection was blunted by the PI3K-IA inhibitor PI-103, but not by the PI3K-IB inhibitor AS-604850. In contrast, IPC-mediated protection was not affected by PI-103 but was blocked by AS-604850, suggesting that PI3K-IA activation is required for OPC while PI3K-IB activation is needed for IPC. Mechanistically, PI3K-IA activity is required for ouabain-induced Akt activation but not PKCε translocation. However, in contrast to PKCε translocation which is critical to protection, Akt activity was not required for OPC. Further studies shall reveal the identity of the downstream targets of this new PI3K IA-dependent branch of OPC. These findings may be of clinical relevance in patients at risk for myocardial infarction with underlying diseases and/or medication that could differentially affect the integrity of cardiac PI3K-IA and IB pathways.
Collapse
|
24
|
Lin MJ, Fine M, Lu JY, Hofmann SL, Frazier G, Hilgemann DW. Massive palmitoylation-dependent endocytosis during reoxygenation of anoxic cardiac muscle. eLife 2013; 2:e01295. [PMID: 24282237 PMCID: PMC3839539 DOI: 10.7554/elife.01295] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In fibroblasts, large Ca transients activate massive endocytosis (MEND) that involves membrane protein palmitoylation subsequent to mitochondrial permeability transition pore (PTP) openings. Here, we characterize this pathway in cardiac muscle. Myocytes with increased expression of the acyl transferase, DHHC5, have decreased Na/K pump activity. In DHHC5-deficient myocytes, Na/K pump activity and surface area/volume ratios are increased, the palmitoylated regulatory protein, phospholemman (PLM), and the cardiac Na/Ca exchanger (NCX1) show greater surface membrane localization, and MEND is inhibited in four protocols. Both electrical and optical methods demonstrate that PTP-dependent MEND occurs during reoxygenation of anoxic hearts. Post-anoxia MEND is ablated in DHHC5-deficient hearts, inhibited by cyclosporine A (CsA) and adenosine, promoted by staurosporine (STS), reduced in hearts lacking PLM, and correlates with impaired post-anoxia contractile function. Thus, the MEND pathway appears to be deleterious in severe oxidative stress but may constitutively contribute to cardiac sarcolemma turnover in dependence on metabolic stress. DOI:http://dx.doi.org/10.7554/eLife.01295.001 Many people who survive a stroke or heart attack experience substantial tissue damage when the blood supply is restored. Much of this damage can be caused by the mitochondria inside the cells releasing a protein called cytochrome c that can cause cells to die in a process called apoptosis. The cytochrome c is released as the outer membrane of the mitochondria becomes permeable and pores called permeability transition pores open up in the inner membrane. Now Lin et al. explore if additional molecules released from the mitochondria might also initiate important cellular responses during the reoxygenation of oxygen-deprived tissue. Lin and co-workers recently showed that the mitochondria of some cells can release a small enzyme cofactor, coenzyme A, which then promotes a cellular response called massive endocytosis. This process can cause up to 70% of the cell surface membrane to be absorbed into the interior of the cell in the form of membrane vesicles. Most forms of endocytosis involve a much smaller fraction of the cell membrane and employ a set of well-known endocytic proteins that are not involved in massive endocytosis. Now, Lin et al. investigate the role of massive endocytosis in cardiac muscle. Electrical and optical measurements reveal that massive endocytosis occurs as cardiac cells that have been deprived of oxygen are reoxygenated. Lin et al. also find that an enzyme called DHHC5 must be present to allow endocytosis to take place during reoxygenation. DHHC5 is an enzyme that catalyzes a process called acylation – the transfer of acyl groups to proteins at the cell surface. Moreover, the deletion of DHHC5 has a beneficial impact on the performance of cardiac muscle after oxygen deprivation, which implies that molecules that inhibit protein acylation might protect the heart from damage during reoxygenation. Together, these results establish new pathological and physiological roles for the acylation, which is one of the most common biochemical modifications made to membrane proteins after they are synthesized. DOI:http://dx.doi.org/10.7554/eLife.01295.002
Collapse
Affiliation(s)
- Mei-Jung Lin
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States
| | | | | | | | | | | |
Collapse
|
25
|
Lipoxin a4 preconditioning and postconditioning protect myocardial ischemia/reperfusion injury in rats. Mediators Inflamm 2013; 2013:231351. [PMID: 23956501 PMCID: PMC3730367 DOI: 10.1155/2013/231351] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/15/2013] [Accepted: 06/16/2013] [Indexed: 12/23/2022] Open
Abstract
This study aims to investigate the pre- and postconditioning effects of lipoxin A4 (LXA4) on myocardial damage caused by ischemia/reperfusion (I/R) injury. Seventy-two rats were divided into 6 groups: sham groups (C1 and C2), I/R groups (I/R1 and I/R2), and I/R plus LXA4 preconditioning and postconditioning groups (LX1 and LX2). The serum levels of IL-1β, IL-6, IL-8, IL-10, TNF-α, and cardiac troponin I (cTnI) were measured. The content and the activity of Na+-K+-ATPase as well as the superoxide dismutase (SOD), and malondialdehyde (MDA) levels were determined. Along with the examination of myocardium ultrastructure and ventricular arrhythmia scores (VAS), connexin 43 (Cx43) expression were also detected. Lower levels of IL-1β, IL-6, IL-8, TNF-α, cTnI, MDA content, and VAS and higher levels of IL-10, SOD activity, Na+-K+-ATPase content and activity, and Cx43 expression appeared in LX groups than I/R groups. Besides, H&E staining, TEM examination as well as analysis of gene, and protein confirmed that LXA4 preconditioning was more effective than postconditioning in preventing arrhythmogenesis via the upregulation of Cx43. That is, LXA4 postconditioning had better protective effect on Na+-K+-ATPase and myocardial ultrastructure.
Collapse
|
26
|
Bueno-Orovio A, Sánchez C, Pueyo E, Rodriguez B. Na/K pump regulation of cardiac repolarization: insights from a systems biology approach. Pflugers Arch 2013; 466:183-93. [PMID: 23674099 DOI: 10.1007/s00424-013-1293-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/02/2013] [Accepted: 05/03/2013] [Indexed: 11/26/2022]
Abstract
The sodium-potassium pump is widely recognized as the principal mechanism for active ion transport across the cellular membrane of cardiac tissue, being responsible for the creation and maintenance of the transarcolemmal sodium and potassium gradients, crucial for cardiac cell electrophysiology. Importantly, sodium-potassium pump activity is impaired in a number of major diseased conditions, including ischemia and heart failure. However, its subtle ways of action on cardiac electrophysiology, both directly through its electrogenic nature and indirectly via the regulation of cell homeostasis, make it hard to predict the electrophysiological consequences of reduced sodium-potassium pump activity in cardiac repolarization. In this review, we discuss how recent studies adopting the systems biology approach, through the integration of experimental and modeling methodologies, have identified the sodium-potassium pump as one of the most important ionic mechanisms in regulating key properties of cardiac repolarization and its rate dependence, from subcellular to whole organ levels. These include the role of the pump in the biphasic modulation of cellular repolarization and refractoriness, the rate control of intracellular sodium and calcium dynamics and therefore of the adaptation of repolarization to changes in heart rate, as well as its importance in regulating pro-arrhythmic substrates through modulation of dispersion of repolarization and restitution. Theoretical findings are consistent across a variety of cell types and species including human, and widely in agreement with experimental findings. The novel insights and hypotheses on the role of the pump in cardiac electrophysiology obtained through this integrative approach could eventually lead to novel therapeutic and diagnostic strategies.
Collapse
Affiliation(s)
- Alfonso Bueno-Orovio
- Department of Computer Science, University of Oxford, Wolfson Building, Parks Road, Oxford, OX1 3QD, UK,
| | | | | | | |
Collapse
|