1
|
Li Y, Wright NT, Bloch RJ. The juxtamembrane sequence of small ankyrin 1 mediates the binding of its cytoplasmic domain to SERCA1 and is required for inhibitory activity. J Biol Chem 2025; 301:108216. [PMID: 39863105 PMCID: PMC11927728 DOI: 10.1016/j.jbc.2025.108216] [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: 12/11/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+-ATPase1 (SERCA1) is responsible for the clearance of cytosolic Ca2+ in skeletal muscle. Due to its vital importance in regulating Ca2+ homeostasis, the regulation of SERCA1 has been intensively studied. Small ankyrin 1 (sAnk1, Ank1.5), a 17 kDa muscle-specific isoform of ANK1, binds to SERCA1 directly via both its transmembrane and cytoplasmic domains and inhibits SERCA1's ATPase activity. Here, we characterize the interaction between the cytoplasmic domain of sAnk1 (sAnk1(29-155)) and SERCA1. The binding affinity for sAnk1 (29-155) to SERCA1 was 444 nM by blot overlay, about 7-fold weaker than the binding of sAnk1(29-155) to obscurin, a giant protein of the muscle cytoskeleton. Site-directed mutagenesis identified K38, H39, and H41, in the juxtamembrane region, as residues likely to mediate binding to SERCA1. These residues are not required for obscurin binding. Residues R64-K73, which do contribute to obscurin binding, are also required for binding to SERCA1, but only the hydrophobic residues in this sequence are required, not the positively charged residues necessary for obscurin binding. Circular dichroism analysis of sAnk1(29-155) indicates that most mutants show significant structural changes, with the exception of those containing alanines in place of K38, H39 and H41. Although the cytoplasmic domain of sAnk1 does not inhibit SERCA1's Ca2+-ATPase activity, with or without mutations in the juxtamembrane sequence, the inhibitory activity of full-length sAnk1 requires the WT juxtamembrane sequence. We used these data to model sAnk1 and the sAnk1-SERCA1 complex. Our results suggest that, in addition to its transmembrane domain, sAnk1 uses its juxtamembrane sequence and perhaps part of its obscurin binding site to bind to SERCA1, and that this binding contributes to their robust association in situ, as well as regulation of SERCA1's activity.
Collapse
Affiliation(s)
- Yi Li
- Program in Biochemistry and Molecular Biology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, USA
| | - Robert J Bloch
- Department of Physiology, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA.
| |
Collapse
|
2
|
Desai DA, Baby A, Ananthamohan K, Green LC, Arif M, Duncan BC, Kumar M, Singh RR, Koch SE, Natesan S, Rubinstein J, Jegga AG, Sadayappan S. Roles of cMyBP-C phosphorylation on cardiac contractile dysfunction in db/db mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100075. [PMID: 38957358 PMCID: PMC11218625 DOI: 10.1016/j.jmccpl.2024.100075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease and comorbidity associated with several conditions, including cardiac dysfunction leading to heart failure with preserved ejection fraction (HFpEF), in turn resulting in T2DM-induced cardiomyopathy (T2DM-CM). However, the molecular mechanisms underlying the development of T2DM-CM are poorly understood. It is hypothesized that molecular alterations in myopathic genes induced by diabetes promote the development of HFpEF, whereas cardiac myosin inhibitors can rescue the resultant T2DM-mediated cardiomyopathy. To test this hypothesis, a Leptin receptor-deficient db/db homozygous (Lepr db/db) mouse model was used to define the pathogenesis of T2DM-CM. Echocardiographic studies at 4 and 6 months revealed that Lepr db/db hearts started developing cardiac dysfunction by four months, and left ventricular hypertrophy with diastolic dysfunction was evident at 6 months. RNA-seq data analysis, followed by functional enrichment, revealed the differential regulation of genes related to cardiac dysfunction in Lepr db/db heart tissues. Strikingly, the level of cardiac myosin binding protein-C phosphorylation was significantly increased in Lepr db/db mouse hearts. Finally, using isolated skinned papillary muscles and freshly isolated cardiomyocytes, CAMZYOS ® (mavacamten, MYK-461), a prescription heart medicine used for symptomatic obstructive hypertrophic cardiomyopathy treatment, was tested for its ability to rescue T2DM-CM. Compared with controls, MYK-461 significantly reduced force generation in papillary muscle fibers and cardiomyocyte contractility in the db/db group. This line of evidence shows that 1) T2DM-CM is associated with hyperphosphorylation of cardiac myosin binding protein-C and 2) MYK-461 significantly lessened disease progression in vitro, suggesting its promise as a treatment for HFpEF.
Collapse
Affiliation(s)
- Darshini A. Desai
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Akhil Baby
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Kalyani Ananthamohan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Lisa C. Green
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohammed Arif
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Brittany C. Duncan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohit Kumar
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rohit R. Singh
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sheryl E. Koch
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Jack Rubinstein
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sakthivel Sadayappan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
3
|
Guo XB, Deng X, Wang J, Qi Y, Zhao W, Guan S. HAX-1 interferes in assembly of NLRP3-ASC to block microglial pyroptosis in cerebral I/R injury. Cell Death Discov 2024; 10:264. [PMID: 38811533 PMCID: PMC11136987 DOI: 10.1038/s41420-024-02005-3] [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: 11/26/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
Acute cerebral ischemia has a high rate of disability and death. Although timely recanalization therapy may rescue the ischemic brain tissue, cerebral ischemia-reperfusion injury has been shown to limit the therapeutic effects of vascular recanalization. Protein HAX-1 has been reported as a pro-survival protein that plays an important role in various disorders, particularly in association with the nervous system. However, the effects and mechanisms of HAX-1 in cerebral IR injury have yet to be elucidated. So, we aimed to investigate the effect of HAX-1 on microglial pyroptosis and explore its potential neuroprotective effects in ischemia-reperfusion injury. Our results show that the expression of HAX-1 decreased after cerebral IR injury, accompanied by an increase in pyroptosis pathway activation. In addition, HAX-1 could inhibit microglial pyroptosis both in vivo and in vitro and reduce the release of inflammatory mediators. The above neuroprotective effects might be partially mediated by inhibiting of interaction of NLRP3 and ASC through competitive binding, followed by the attenuation of NLRP3 inflammasome formation. In conclusion, Our findings support that HAX-1 exhibits a protective role in cerebral I/R injury, and further study on HAX-1 expression regulation will contribute to cerebral infarction therapy.
Collapse
Affiliation(s)
- Xin-Bin Guo
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, 450052, Zhengzhou, China
| | - Xin Deng
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, 450052, Zhengzhou, China
| | - Jingjing Wang
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, 450052, Zhengzhou, China
| | - Yuruo Qi
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Co-innovation Center of Henan Province for New Drug R & D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, 450001, Zhengzhou, Henan, China
| | - Wen Zhao
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Co-innovation Center of Henan Province for New Drug R & D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, 450001, Zhengzhou, Henan, China.
| | - Sheng Guan
- Department of Neuro-interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, 450052, Zhengzhou, China.
| |
Collapse
|
4
|
Adamcova M, Parova H, Lencova-Popelova O, Kollarova-Brazdova P, Baranova I, Slavickova M, Stverakova T, Mikyskova PS, Mazurova Y, Sterba M. Cardiac miRNA expression during the development of chronic anthracycline-induced cardiomyopathy using an experimental rabbit model. Front Pharmacol 2024; 14:1298172. [PMID: 38235109 PMCID: PMC10791979 DOI: 10.3389/fphar.2023.1298172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Background: Anthracycline cardiotoxicity is a well-known complication of cancer treatment, and miRNAs have emerged as a key driver in the pathogenesis of cardiovascular diseases. This study aimed to investigate the expression of miRNAs in the myocardium in early and late stages of chronic anthracycline induced cardiotoxicity to determine whether this expression is associated with the severity of cardiac damage. Method: Cardiotoxicity was induced in rabbits via daunorubicin administration (daunorubicin, 3 mg/kg/week; for five and 10 weeks), while the control group received saline solution. Myocardial miRNA expression was first screened using TaqMan Advanced miRNA microfluidic card assays, after which 32 miRNAs were selected for targeted analysis using qRT-PCR. Results: The first subclinical signs of cardiotoxicity (significant increase in plasma cardiac troponin T) were observed after 5 weeks of daunorubicin treatment. At this time point, 10 miRNAs (including members of the miRNA-34 and 21 families) showed significant upregulation relative to the control group, with the most intense change observed for miRNA-1298-5p (29-fold change, p < 0.01). After 10 weeks of daunorubicin treatment, when a further rise in cTnT was accompanied by significant left ventricle systolic dysfunction, only miR-504-5p was significantly (p < 0.01) downregulated, whereas 10 miRNAs were significantly upregulated relative to the control group; at this time-point, the most intense change was observed for miR-34a-5p (76-fold change). Strong correlations were found between the expression of multiple miRNAs (including miR-34 and mir-21 family and miR-1298-5p) and quantitative indices of toxic damage in both the early and late phases of cardiotoxicity development. Furthermore, plasma levels of miR-34a-5p were strongly correlated with the myocardial expression of this miRNA. Conclusion: To the best of our knowledge, this is the first study that describes alterations in miRNA expression in the myocardium during the transition from subclinical, ANT-induced cardiotoxicity to an overt cardiotoxic phenotype; we also revealed how these changes in miRNA expression are strongly correlated with quantitative markers of cardiotoxicity.
Collapse
Affiliation(s)
| | - Helena Parova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | | | | | - Ivana Baranova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Marcela Slavickova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Tereza Stverakova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Petra Sauer Mikyskova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Yvona Mazurova
- Department of Histology and Embryology, Charles University in Prague, Hradec Kralove, Czechia
| | | |
Collapse
|
5
|
Diny NL, Wood MK, Won T, Talor MV, Lukban C, Bedja D, Wang N, Kalinoski H, Daoud A, Talbot CC, Leei Lin B, Čiháková D. Hypereosinophilia causes progressive cardiac pathologies in mice. iScience 2023; 26:107990. [PMID: 37829205 PMCID: PMC10565781 DOI: 10.1016/j.isci.2023.107990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/02/2023] [Accepted: 09/16/2023] [Indexed: 10/14/2023] Open
Abstract
Hypereosinophilic syndrome is a progressive disease with extensive eosinophilia that results in organ damage. Cardiac pathologies are the main reason for its high mortality rate. A better understanding of the mechanisms of eosinophil-mediated tissue damage would benefit therapeutic development. Here, we describe the cardiac pathologies that developed in a mouse model of hypereosinophilic syndrome. These IL-5 transgenic mice exhibited decreased left ventricular function at a young age which worsened with age. Mechanistically, we demonstrated infiltration of activated eosinophils into the heart tissue that led to an inflammatory environment. Gene expression signatures showed tissue damage as well as repair and remodeling processes. Cardiomyocytes from IL-5Tg mice exhibited significantly reduced contractility relative to wild type (WT) controls. This impairment may result from the inflammatory stress experienced by the cardiomyocytes and suggest that dysregulation of contractility and Ca2+ reuptake in cardiomyocytes contributes to cardiac dysfunction at the whole organ level in hypereosinophilic mice.
Collapse
Affiliation(s)
- Nicola Laura Diny
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Megan Kay Wood
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Monica Vladut Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Clarisse Lukban
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Djahida Bedja
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nadan Wang
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian Leei Lin
- Department of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Trębińska-Stryjewska A, Wakula M, Chmielarczyk M, Grzybowska EA. HAX1: A versatile, intrinsically disordered regulatory protein. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119538. [PMID: 37454914 DOI: 10.1016/j.bbamcr.2023.119538] [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: 04/17/2023] [Revised: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
HAX1 is a relatively small, ubiquitously expressed, predominantly mitochondrial, intrinsically disordered protein. It has been implicated in the regulation of apoptosis, cell migration, calcium cycling, proteostasis, angiogenesis, autophagy and translation. A wide spectrum of functions, numerous interactions and still elusive molecular mechanisms of action make HAX1 an intriguing subject of research. Moreover, HAX1 is involved in the pathogenesis of diseases; its deficiency leads to neutropenia and its overexpression is associated with cancer. In this review we aim to describe the characteristics of HAX1 gene and protein, and comprehensively discuss its multiple functions, highlighting the emerging role of HAX1 in protection from stress and apoptosis through maintaining cellular proteostasis and homeostasis.
Collapse
Affiliation(s)
| | - Maciej Wakula
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Ewa A Grzybowska
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| |
Collapse
|
7
|
Mehdi RR, Kumar M, Mendiola EA, Sadayappan S, Avazmohammadi R. Machine learning-based classification of cardiac relaxation impairment using sarcomere length and intracellular calcium transients. Comput Biol Med 2023; 163:107134. [PMID: 37379617 PMCID: PMC10525035 DOI: 10.1016/j.compbiomed.2023.107134] [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: 04/06/2023] [Revised: 05/29/2023] [Accepted: 06/04/2023] [Indexed: 06/30/2023]
Abstract
Impaired relaxation of cardiomyocytes leads to diastolic dysfunction in the left ventricle. Relaxation velocity is regulated in part by intracellular calcium (Ca2+) cycling, and slower outflux of Ca2+ during diastole translates to reduced relaxation velocity of sarcomeres. Sarcomere length transient and intracellular calcium kinetics are integral parts of characterizing the relaxation behavior of the myocardium. However, a classifier tool that can separate normal cells from cells with impaired relaxation using sarcomere length transient and/or calcium kinetics remains to be developed. In this work, we employed nine different classifiers to classify normal and impaired cells, using ex-vivo measurements of sarcomere kinematics and intracellular calcium kinetics data. The cells were isolated from wild-type mice (referred to as normal) and transgenic mice expressing impaired left ventricular relaxation (referred to as impaired). We utilized sarcomere length transient data with a total of n = 126 cells (n = 60 normal cells and n = 66 impaired cells) and intracellular calcium cycling measurements with a total of n = 116 cells (n = 57 normal cells and n = 59 impaired cells) from normal and impaired cardiomyocytes as inputs to machine learning (ML) models for classification. We trained all ML classifiers with cross-validation method separately using both sets of input features, and compared their performance metrics. The performance of classifiers on test data showed that our soft voting classifier outperformed all other individual classifiers on both sets of input features, with 0.94 and 0.95 area under the receiver operating characteristic curves for sarcomere length transient and calcium transient, respectively, while multilayer perceptron achieved comparable scores of 0.93 and 0.95, respectively. However, the performance of decision tree, and extreme gradient boosting was found to be dependent on the set of input features used for training. Our findings highlight the importance of selecting appropriate input features and classifiers for the accurate classification of normal and impaired cells. Layer-wise relevance propagation (LRP) analysis demonstrated that the time to 50% contraction of the sarcomere had the highest relevance score for sarcomere length transient, whereas time to 50% decay of calcium had the highest relevance score for calcium transient input features. Despite the limited dataset, our study demonstrated satisfactory accuracy, suggesting that the algorithm can be used to classify relaxation behavior in cardiomyocytes when the potential relaxation impairment of the cells is unknown.
Collapse
Affiliation(s)
- Rana Raza Mehdi
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Mohit Kumar
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, Cincinnati, OH, USA; Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Emilio A Mendiola
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Sakthivel Sadayappan
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, Cincinnati, OH, USA; Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Reza Avazmohammadi
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA; Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA; J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
8
|
Kumar M, Haghighi K, Koch S, Rubinstein J, Stillitano F, Hajjar RJ, Kranias EG, Sadayappan S. Myofilament Alterations Associated with Human R14del-Phospholamban Cardiomyopathy. Int J Mol Sci 2023; 24:2675. [PMID: 36768995 PMCID: PMC9917359 DOI: 10.3390/ijms24032675] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Phospholamban (PLN) is a major regulator of cardiac contractility, and human mutations in this gene give rise to inherited cardiomyopathies. The deletion of Arginine 14 is the most-prevalent cardiomyopathy-related mutation, and it has been linked to arrhythmogenesis and early death. Studies in PLN-humanized mutant mice indicated an increased propensity to arrhythmias, but the underlying cellular mechanisms associated with R14del-PLN cardiac dysfunction in the absence of any apparent structural remodeling remain unclear. The present study addressed the specific role of myofilaments in the setting of R14del-PLN and the long-term effects of R14del-PLN in the heart. Maximal force was depressed in skinned cardiomyocytes from both left and right ventricles, but this effect was more pronounced in the right ventricle of R14del-PLN mice. In addition, the Ca2+ sensitivity of myofilaments was increased in both ventricles of mutant mice. However, the depressive effects of R14del-PLN on contractile parameters could be reversed with the positive inotropic drug omecamtiv mecarbil, a myosin activator. At 12 months of age, corresponding to the mean symptomatic age of R14del-PLN patients, contractile parameters and Ca2+ transients were significantly depressed in the right ventricular R14del-PLN cardiomyocytes. Echocardiography did not reveal any alterations in cardiac function or remodeling, although histological and electron microscopy analyses indicated subtle alterations in mutant hearts. These findings suggest that both aberrant myocyte calcium cycling and aberrant contractility remain specific to the right ventricle in the long term. In addition, altered myofilament activity is an early characteristic of R14del-PLN mutant hearts and the positive inotropic drug omecamtiv mecarbil may be beneficial in treating R14del-PLN cardiomyopathy.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sheryl Koch
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jack Rubinstein
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Francesca Stillitano
- Division Heart and Lung, Department of Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Roger J. Hajjar
- Phospholamban Heart Foundation, Postbus 66, 1775 ZH Middenmeer, The Netherlands
| | - Evangelia G. Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
9
|
Ernst P, Bidwell PA, Dora M, Thomas DD, Kamdar F. Cardiac calcium regulation in human induced pluripotent stem cell cardiomyocytes: Implications for disease modeling and maturation. Front Cell Dev Biol 2023; 10:986107. [PMID: 36742199 PMCID: PMC9889838 DOI: 10.3389/fcell.2022.986107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) are based on ground-breaking technology that has significantly impacted cardiovascular research. They provide a renewable source of human cardiomyocytes for a variety of applications including in vitro disease modeling and drug toxicity testing. Cardiac calcium regulation plays a critical role in the cardiomyocyte and is often dysregulated in cardiovascular disease. Due to the limited availability of human cardiac tissue, calcium handling and its regulation have most commonly been studied in the context of animal models. hiPSC-CMs can provide unique insights into human physiology and pathophysiology, although a remaining limitation is the relative immaturity of these cells compared to adult cardiomyocytes Therefore, this field is rapidly developing techniques to improve the maturity of hiPSC-CMs, further establishing their place in cardiovascular research. This review briefly covers the basics of cardiomyocyte calcium cycling and hiPSC technology, and will provide a detailed description of our current understanding of calcium in hiPSC-CMs.
Collapse
Affiliation(s)
- Patrick Ernst
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Philip A. Bidwell
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| | - Michaela Dora
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, United States
| | - David D. Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Forum Kamdar
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
10
|
Aberrant PLN-R14del Protein Interactions Intensify SERCA2a Inhibition, Driving Impaired Ca2+ Handling and Arrhythmogenesis. Int J Mol Sci 2022; 23:ijms23136947. [PMID: 35805951 PMCID: PMC9266971 DOI: 10.3390/ijms23136947] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Phospholamban (PLN), a key modulator of Ca2+-homeostasis, inhibits sarcoplasmic reticulum (SR) calcium-ATPase (SERCA2a) and regulates cardiac contractility. The human PLN mutation R14del has been identified in arrhythmogenic cardiomyopathy patients worldwide and is currently extensively investigated. In search of the molecular mechanisms mediating the pathological phenotype, we examined PLN-R14del associations to known PLN-interacting partners. We determined that PLN-R14del interactions to key Ca2+-handling proteins SERCA2a and HS-1-associated protein X-1 (HAX-1) were enhanced, indicating the super-inhibition of SERCA2a’s Ca2+-affinity. Additionally, histidine-rich calcium binding protein (HRC) binding to SERCA2a was increased, suggesting the inhibition of SERCA2a maximal velocity. As phosphorylation relieves the inhibitory effect of PLN on SERCA2a activity, we examined the impact of phosphorylation on the PLN-R14del/SERCA2a interaction. Contrary to PLN-WT, phosphorylation did not affect PLN-R14del binding to SERCA2a, due to a lack of Ser-16 phosphorylation in PLN-R14del. No changes were observed in the subcellular distribution of PLN-R14del or its co-localization to SERCA2a. However, in silico predictions suggest structural perturbations in PLN-R14del that could impact its binding and function. Our findings reveal for the first time that by increased binding to SERCA2a and HAX-1, PLN-R14del acts as an enhanced inhibitor of SERCA2a, causing a cascade of molecular events contributing to impaired Ca2+-homeostasis and arrhythmogenesis. Relieving SERCA2a super-inhibition could offer a promising therapeutic approach for PLN-R14del patients.
Collapse
|
11
|
Hax-1 Regulates Radiation-Induced Mitochondrial-Dependent Apoptosis of Uveal Melanoma Cells through PI3K/AKT/eNOS Pathway. JOURNAL OF ONCOLOGY 2022; 2022:2956888. [PMID: 35602302 PMCID: PMC9122716 DOI: 10.1155/2022/2956888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/23/2022]
Abstract
Uveal melanoma is an aggressive skin cancer that remains insurmountable and is accompanied by inferior prognostic results. The proliferative and survival mechanisms of uveal melanoma cells need to be further investigated to improve the treatment of uveal melanoma. According to reports, HAX-1 is an antiapoptotic protein vital for multiple malignancies. Nevertheless, the role and causal link of HAX-1 in uveal melanoma are still elusive. The survival diversity of uveal melanoma sufferers with diverse haX-1 expressing levels was studied by TCGA database. Patients in the riskhigh group exhibited greater levels of HAX-1 in contrast to the risklow group, and individuals with higher HAX-1 levels displayed inferior survival times. The outcomes of CCK-8 and clonogenesis revealed that the proliferative rate of haX-1 knockout cells was slower. The result of scratch experiment shows that the ability of scratch recovery after HAX-1 is reduced. Transwell migration and tumor cell pelletization experiments showed that siHAX-1 significantly reduced cell migration and tumor cell pelletization. After haX-1 was knocked out, the loss of MMP was decreased, the transfer of CyT C was elevated, and the protein expression of Bax, Caspase 3, and Bcl2 was elevated, suggesting that mitochondria-induced apoptosis was increased. Sihax-1 treatment remarkably decreased the phosphonation of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR)/endothelial NO synthase (eNOS) in mum-2B and C918. Pretreatment with LY294002 significantly restored iHAX-1-induced decline in PI3K/AKT/mTOR/eNOS phosphorylation. Therefore, our results suggest that haX-1 induces radiation-dependent apoptosis of UM cells via the PI3K/AKT/eNOS signal path.
Collapse
|
12
|
Fadeel B, Garwicz D, Carlsson G, Sandstedt B, Nordenskjöld M. Kostmann disease and other forms of severe congenital neutropenia. Acta Paediatr 2021; 110:2912-2920. [PMID: 34160857 DOI: 10.1111/apa.16005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/05/2021] [Accepted: 06/22/2021] [Indexed: 12/17/2022]
Abstract
Congenital neutropenia with autosomal recessive inheritance was first described by the Swedish paediatrician Rolf Kostmann who coined the term 'infantile genetic agranulocytosis'. The condition is now commonly referred to as Kostmann disease. These patients display a maturation arrest of the myelopoiesis in the bone marrow and reduced neutrophil numbers and suffer from recurrent, often life-threatening infections. The molecular mechanism underlying congenital neutropenia has been intensively investigated, and mutations in genes that impinge on programmed cell death have been identified. The present review provides an overview of these studies.
Collapse
Affiliation(s)
- Bengt Fadeel
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | - Daniel Garwicz
- Department of Medical Sciences Uppsala University Uppsala Sweden
| | - Göran Carlsson
- Department of Woman and Child Health Karolinska University Hospital Stockholm Sweden
| | - Bengt Sandstedt
- Department of Woman and Child Health Karolinska University Hospital Stockholm Sweden
| | - Magnus Nordenskjöld
- Department of Molecular Medicine and Surgery Karolinska Institutet Stockholm Sweden
- Department of Clinical Genetics Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
13
|
Phospholamban and sarcolipin prevent thermal inactivation of sarco(endo)plasmic reticulum Ca2+-ATPases. Biochem J 2020; 477:4281-4294. [PMID: 33111944 DOI: 10.1042/bcj20200346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/15/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022]
Abstract
Na+-K+-ATPase from mice lacking the γ subunit exhibits decreased thermal stability. Phospholamban (PLN) and sarcolipin (SLN) are small homologous proteins that regulate sarco(endo)plasmic reticulum Ca2+-ATPases (SERCAs) with properties similar to the γ subunit, through physical interactions with SERCAs. Here, we tested the hypothesis that PLN and SLN may protect against thermal inactivation of SERCAs. HEK-293 cells were co-transfected with different combinations of cDNAs encoding SERCA2a, PLN, a PLN mutant (N34A) that cannot bind to SERCA2a, and SLN. One-half of the cells were heat stressed at 40°C for 1 h (HS), and one-half were maintained at 37°C (CTL) before harvesting the cells and isolating microsomes. Compared with CTL, maximal SERCA activity was reduced by 25-35% following HS in cells that expressed either SERCA2a alone or SERCA2a and mutant PLN (N34A) whereas no change in maximal SERCA2a activity was observed in cells that co-expressed SERCA2a and either PLN or SLN following HS. Increases in SERCA2a carbonyl group content and nitrotyrosine levels that were detected following HS in cells that expressed SERCA2a alone were prevented in cells co-expressing SERCA2a with PLN or SLN, whereas co-expression of SERCA2a with mutant PLN (N34A) only prevented carbonyl group formation. In other experiments using knock-out mice, we found that thermal inactivation of SERCA was increased in cardiac left ventricle samples from Pln-null mice and in diaphragm samples from Sln-null mice, compared with WT littermates. Our results show that both PLN and SLN form a protective interaction with SERCA pumps during HS, preventing nitrosylation and oxidation of SERCA and thus preserving its maximal activity.
Collapse
|
14
|
Kumar M, Haghighi K, Kranias EG, Sadayappan S. Phosphorylation of cardiac myosin-binding protein-C contributes to calcium homeostasis. J Biol Chem 2020; 295:11275-11291. [PMID: 32554466 DOI: 10.1074/jbc.ra120.013296] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiac myosin-binding protein-C (cMyBP-C) is highly phosphorylated under basal conditions. However, its phosphorylation level is decreased in individuals with heart failure. The necessity of cMyBP-C phosphorylation for proper contractile function is well-established, but the physiological and pathological consequences of decreased cMyBP-C phosphorylation in the heart are not clear. Herein, using intact adult cardiomyocytes from mouse models expressing phospho-ablated (AAA) and phosphomimetic (DDD) cMyBP-C as well as controls, we found that cMyBP-C dephosphorylation is sufficient to reduce contractile parameters and calcium kinetics associated with prolonged decay time of the calcium transient and increased diastolic calcium levels. Isoproterenol stimulation reversed the depressive contractile and Ca2+-kinetic parameters. Moreover, caffeine-induced calcium release yielded no difference between AAA/DDD and controls in calcium content of the sarcoplasmic reticulum. On the other hand, sodium-calcium exchanger function and phosphorylation levels of calcium-handling proteins were significantly decreased in AAA hearts compared with controls. Stress conditions caused increases in both spontaneous aftercontractions in AAA cardiomyocytes and the incidence of arrhythmias in vivo compared with the controls. Treatment with omecamtiv mecarbil, a positive cardiac inotropic drug, rescued the contractile deficit in AAA cardiomyocytes, but not the calcium-handling abnormalities. These findings indicate a cascade effect whereby cMyBP-C dephosphorylation causes contractile defects, which then lead to calcium-cycling abnormalities, resulting in aftercontractions and increased incidence of cardiac arrhythmias under stress conditions. We conclude that improvement of contractile deficits alone without improving calcium handling may be insufficient for effective management of heart failure.
Collapse
Affiliation(s)
- Mohit Kumar
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sakthivel Sadayappan
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA .,Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
15
|
Chen J, Sitsel A, Benoy V, Sepúlveda MR, Vangheluwe P. Primary Active Ca 2+ Transport Systems in Health and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035113. [PMID: 31501194 DOI: 10.1101/cshperspect.a035113] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calcium ions (Ca2+) are prominent cell signaling effectors that regulate a wide variety of cellular processes. Among the different players in Ca2+ homeostasis, primary active Ca2+ transporters are responsible for keeping low basal Ca2+ levels in the cytosol while establishing steep Ca2+ gradients across intracellular membranes or the plasma membrane. This review summarizes our current knowledge on the three types of primary active Ca2+-ATPases: the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pumps, the secretory pathway Ca2+- ATPase (SPCA) isoforms, and the plasma membrane Ca2+-ATPase (PMCA) Ca2+-transporters. We first discuss the Ca2+ transport mechanism of SERCA1a, which serves as a reference to describe the Ca2+ transport of other Ca2+ pumps. We further highlight the common and unique features of each isoform and review their structure-function relationship, expression pattern, regulatory mechanisms, and specific physiological roles. Finally, we discuss the increasing genetic and in vivo evidence that links the dysfunction of specific Ca2+-ATPase isoforms to a broad range of human pathologies, and highlight emerging therapeutic strategies that target Ca2+ pumps.
Collapse
Affiliation(s)
- Jialin Chen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Aljona Sitsel
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Veronick Benoy
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
16
|
Larsen EK, Weber DK, Wang S, Gopinath T, Blackwell DJ, Dalton MP, Robia SL, Gao J, Veglia G. Intrinsically disordered HAX-1 regulates Ca 2+ cycling by interacting with lipid membranes and the phospholamban cytoplasmic region. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183034. [PMID: 31400305 PMCID: PMC6899184 DOI: 10.1016/j.bbamem.2019.183034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/09/2019] [Accepted: 07/30/2019] [Indexed: 01/14/2023]
Abstract
Hematopoietic-substrate-1 associated protein X-1 (HAX-1) is a 279 amino acid protein expressed ubiquitously. In cardiac muscle, HAX-1 was found to modulate the sarcoendoplasmic reticulum calcium ATPase (SERCA) by shifting its apparent Ca2+ affinity (pCa). It has been hypothesized that HAX-1 binds phospholamban (PLN), enhancing its inhibitory function on SERCA. HAX-1 effects are reversed by cAMP-dependent protein kinase A that phosphorylates PLN at Ser16. To date, the molecular mechanisms for HAX-1 regulation of the SERCA/PLN complex are still unknown. Using enzymatic, in cell assays, circular dichroism, and NMR spectroscopy, we found that in the absence of a binding partner HAX-1 is essentially disordered and adopts a partial secondary structure upon interaction with lipid membranes. Also, HAX-1 interacts with the cytoplasmic region of monomeric and pentameric PLN as detected by NMR and in cell FRET assays, respectively. We propose that the regulation of the SERCA/PLN complex by HAX-1 is mediated by its interactions with lipid membranes, adding another layer of control in Ca2+ homeostatic balance in the heart muscle.
Collapse
Affiliation(s)
- Erik K Larsen
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel K Weber
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Songlin Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tata Gopinath
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Michael P Dalton
- Department of Physiology, Loyola University, Maywood, IL 60153, USA
| | - Seth L Robia
- Department of Physiology, Loyola University, Maywood, IL 60153, USA
| | - Jiali Gao
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; School of Chemical Biology and Technology, Beijing University Graduate School, Shenzhen 518055, China
| | - Gianluigi Veglia
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
17
|
Liu Y, Duan C, Liu W, Chen X, Wang Y, Liu X, Yue J, Yang J, Zhou X. Upregulation of let-7f-2-3p by long noncoding RNA NEAT1 inhibits XPO1-mediated HAX-1 nuclear export in both in vitro and in vivo rodent models of doxorubicin-induced cardiotoxicity. Arch Toxicol 2019; 93:3261-3276. [PMID: 31570982 DOI: 10.1007/s00204-019-02586-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022]
Abstract
Clinical application of doxorubicin (Dox) is limited due to its undesirable side effects, especially cardiotoxicity. Several microRNAs (miRNAs) such as microRNA-140-5p and miR-23a aggravate Dox-induced cardiotoxicity. Here we demonstrate that upregulation of miRNA let-7f-2-3p by long noncoding RNA (lncRNA) NEAT1 inhibits exportin-1 (XPO1)-mediated nuclear export of hematopoietic-substrate-1 associated protein X-1 (HAX-1) in Dox-induced cardiotoxicity. Treatment of the H9c2 cells with the Dox (1 μM) for 6 h inhibited HAX-1 nuclear export and decreased XPO1 expression. Overexpression of XPO1 significantly attenuated the Dox-induced leakage of myocardial enzymes (creatine phosphokinase, creatine kinase-MB and lactate dehydrogenase) and cardiomyocyte apoptosis with the increased HAX-1 nuclear export. Differentially expressed miRNAs including let-7f-2-3p were selected from the Dox or vehicle-treated cardiomyocytes. TargetScan and luciferase assay showed that let-7f-2-3p targeted XPO1 3' UTR. Inhibition of let-7f-2-3p reduced Dox-induced cardiotoxicity and apoptosis by inhibiting XPO1-mediated HAX-1 nuclear export, whereas let-7f-2-3p overexpression aggravated these effects. In addition, lncRNA NEAT1 was identified as an endogenous sponge RNA to repress let-7f-2-3p expression. Overexpression of lncRNA NEAT1 abolished the increased let-7f-2-3p expression by Dox, and thereby attenuated cardiotoxicity. The loss function of let-7f-2-3p increased XPO1-mediated HAX-1 nuclear export and reduced myocardial injury in Dox (20 mg/kg)-treated rats. Importantly, let-7f-2-3p inhibition in mice alleviated Dox-induced cardiotoxicity and preserved the antitumor efficacy. Together, let-7f-2-3p regulated by lncRNA NEAT1 aggravates Dox-induced cardiotoxicity through inhibiting XPO1-mediated HAX-1 nuclear export, and may serve as a potential therapeutic target against Dox-induced cardiotoxicity.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/therapeutic use
- Antibiotics, Antineoplastic/toxicity
- Apoptosis/drug effects
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Cardiotoxicity
- Cell Line
- Doxorubicin/therapeutic use
- Doxorubicin/toxicity
- Heart/drug effects
- Intracellular Signaling Peptides and Proteins/metabolism
- Karyopherins/genetics
- Male
- Mice, Inbred C57BL
- MicroRNAs/genetics
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA, Long Noncoding/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/genetics
- Up-Regulation
- Exportin 1 Protein
Collapse
Affiliation(s)
- Yanzhuo Liu
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, 430060, China
| | - Chenfan Duan
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, 430060, China
| | - Wen Liu
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuewei Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yang Wang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoxiao Liu
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiang Yue
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jing Yang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoyang Zhou
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
18
|
Gamu D, Juracic ES, Fajardo VA, Rietze BA, Tran K, Bombardier E, Tupling AR. Phospholamban deficiency does not alter skeletal muscle SERCA pumping efficiency or predispose mice to diet-induced obesity. Am J Physiol Endocrinol Metab 2019; 316:E432-E442. [PMID: 30601702 DOI: 10.1152/ajpendo.00288.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump is a major contributor to skeletal muscle Ca2+ homeostasis and metabolic rate. SERCA activity can become adaptively uncoupled by its regulator sarcolipin (SLN) to increase the energy demand of Ca2+ pumping, preventing excessive obesity and glucose intolerance in mice. Several other SERCA regulators bear structural and functional resemblance to SLN, including phospholamban (PLN). Here, we sought to examine whether endogenous levels of skeletal muscle PLN control SERCA Ca2+ pumping efficiency and whole body metabolism. Using PLN-null mice ( Pln-/-), we found that soleus (SOL) muscle's SERCA pumping efficiency (measured as an apparent coupling ratio: Ca2+ uptake/ATP hydrolysis) was unaffected by PLN. Expression of Ca2+-handling proteins within the SOL, including SLN, were comparable between Pln-/- and wild-type (WT) littermates, as were fiber-type characteristics. Not surprisingly then, Pln-/- mice developed a similar degree of diet-induced obesity and glucose intolerance as WT controls when given a "Western" high-fat diet. Lack of an excessively obesogenic phenotype of Pln-/- could not be explained by compensation from skeletal muscle SLN or brown adipose tissue uncoupling protein-1 content. In agreement with several other reports, our study lends support to the notion that PLN serves a functionally distinct role from that of SLN in skeletal muscle physiology.
Collapse
Affiliation(s)
- Daniel Gamu
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Emma Sara Juracic
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Val A Fajardo
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | | | - Khanh Tran
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Eric Bombardier
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| |
Collapse
|