1
|
Zaglia T, Campo A, Moro N, Di Mauro V, Borile G, Menabò R, Antonucci S, Poli L, Campesan M, Carullo P, Martinazzi S, Luciani GB, Hammer K, Pesce P, Bariani R, Faggian G, Maier L, Ventura L, De Stefani D, Mammucari C, Rizzuto R, Catalucci D, Di Lisa F, Mongillo M. Enhancement of mitochondrial calcium uptake is cardioprotective against maladaptive hypertrophy by retrograde signaling uptuning Akt. Proc Natl Acad Sci U S A 2025; 122:e2402639122. [PMID: 40067892 PMCID: PMC11929399 DOI: 10.1073/pnas.2402639122] [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: 02/11/2024] [Accepted: 01/27/2025] [Indexed: 03/25/2025] Open
Abstract
Regulation of mitochondrial Ca2+ uptake is critical in cardiac adaptation to chronic stressors. Abnormalities in Ca2+ handling, including mitochondrial uptake mechanisms, have been implicated in pathological heart hypertrophy. Enhancing mitochondrial Ca2+ uniporter (MCU) expression has been suggested to interfere with maladaptive development of heart failure. Here, we addressed whether MCU modulation affects the cardiac response to pressure overload. MCU content was quantified in human and murine hearts at different phases of myocardial hypertrophy. Cardiac function/structure were analyzed after Transverse Aortic Constriction (TAC) in mice undergone viral-assisted overexpression or downregulation of MCU. In vitro and ex vivo assays determined the effect of MCU modulation on mitochondrial Ca2+ uptake, cellular phenotype and hypertrophic signaling. In human and murine hearts MCU levels increased in the adaptive phase of myocardial hypertrophy and declined in the failing stage. Consistently, modulation of MCU had a cell-autonomous effect in cardiomyocyte/heart adaptation to chronic overload. Indeed, upon TAC MCU-downregulation accelerated development of contractile dysfunction, interstitial fibrosis and heart failure. Conversely, MCU-overexpression prolonged the adaptive phase of hypertrophic response, as, in advanced stages upon TAC, hearts showed preserved contractility, absence of fibrosis and intact vascularization. In vitro and ex vivo analyses indicated that enhancement in mitochondrial Ca2+ uptake in cardiomyocytes entails "mitochondrion-to-cytoplasm" signals leading to ROS-mediated activation of Akt, which may explain the protective effects towards heart response to TAC. Enhanced mitochondrial Ca2+ uptake affects the compensatory response to pressure overload via retrograde mitochondrial-Ca2+/ROS/Akt signaling, thus uncovering a potentially targetable mechanism against maladaptive myocardial hypertrophy.
Collapse
Affiliation(s)
- Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
- Interdepartmental Research Center of Myology, Padova35131, Italy
| | - Antonio Campo
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
| | - Nicola Moro
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
| | - Vittoria Di Mauro
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
| | - Giulia Borile
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
| | - Roberta Menabò
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | | | - Laura Poli
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
| | - Marika Campesan
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Pierluigi Carullo
- Istituti di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Rozzano, Milan20089, Italy
- National Research Council, Institute of Genetics and Biomedical Research, Milan Unit, Milan20138, Italy
| | - Sara Martinazzi
- Division of Cardiac Surgery, University of Verona, Verona37126, Italy
| | | | - Karin Hammer
- Internal Medicine II, University Hospital Regensburg, Regensburg93053, Germany
| | - Paola Pesce
- Department of Medicine, University of Padova, Padova35128, Italy
| | - Riccardo Bariani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova35131, Italy
| | - Giuseppe Faggian
- Division of Cardiac Surgery, University of Verona, Verona37126, Italy
| | - Lars Maier
- Internal Medicine II, University Hospital Regensburg, Regensburg93053, Germany
| | - Laura Ventura
- Department of Statistical Sciences, University of Padova, Padova35121, Italy
| | - Diego De Stefani
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Daniele Catalucci
- Istituti di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Rozzano, Milan20089, Italy
- National Research Council, Institute of Genetics and Biomedical Research, Milan Unit, Milan20138, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- Veneto Institute of Molecular Medicine, Padova35129, Italy
- Interdepartmental Research Center of Myology, Padova35131, Italy
- National Research Council Institute of Neuroscience, Padova35121, Italy
| |
Collapse
|
2
|
Feng Y, Xin Y, Tang W, Zhang P, Jiang Y, Li H, Gong Y, Chen F, Xu Z, Liu Z, Gao L. Repeat administration of human umbilical cord mesenchymal stem cells improves left ventricular diastolic function in mice with heart failure with preserved ejection fraction. Biochem Biophys Res Commun 2024; 737:150525. [PMID: 39142139 DOI: 10.1016/j.bbrc.2024.150525] [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: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
Currently, no therapy is proven to effectively improve heart failure with preserved ejection fraction (HFpEF). Although stem cell therapy has demonstrated promising results in treating ischemic heart disease, the effectiveness of treating HFpEF with human umbilical cord mesenchymal stem cells (hucMSCs) remains unclear. To answer this question, we administered hucMSCs intravenously (i.v.), either once or repetitively, in a mouse model of HFpEF induced by a high-fat diet and NG-nitroarginine methyl ester hydrochloride. hucMSC treatment improved left ventricular diastolic dysfunction, reduced heart weight and pulmonary edema, and attenuated cardiac modeling (inflammation, interstitial fibrosis, and hypertrophy) in HFpEF mice. Repeat hucMSC administration had better outcomes than a single injection. In vitro, hucMSC culture supernatants reduced maladaptive remodeling in neonatal-rat cardiomyocytes. Ribonucleic acid sequencing and protein level analysis of left ventricle (LV) tissues suggested that hucMSCs activated the protein kinase B (Akt)/forkhead box protein O1 (FoxO1) signaling pathway to treat HFpEF. Inhibition of this pathway reversed the efficacy of hucMSC treatment. In conclusion, these findings indicated that hucMSCs could be a viable therapeutic option for HFpEF.
Collapse
Affiliation(s)
- Yunzhen Feng
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenjie Tang
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Pengfei Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Yun Jiang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Hao Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Yanshan Gong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Feng Chen
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Zhifeng Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China; Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| | - Ling Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| |
Collapse
|
3
|
Lee C, Liang LW, Hasegawa K, Maurer MS, Tower-Rader A, Fifer MA, Reilly M, Shimada YJ. Signaling Pathways Associated With Prior Cardiovascular Events in Hypertrophic Cardiomyopathy. J Card Fail 2024; 30:462-472. [PMID: 37562580 PMCID: PMC10853480 DOI: 10.1016/j.cardfail.2023.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiomyopathy. A subset of patients experience major adverse cardiovascular events (MACEs), including arrhythmias, strokes and heart failure. However, the molecular mechanisms underlying MACEs in HCM are still not well understood. Therefore, we conducted a multicenter case-control study of patients with HCM, comparing those with and without prior histories of MACEs to identify dysregulated signaling pathways through plasma proteomics profiling. METHODS We performed plasma proteomics profiling of 4986 proteins. We developed a proteomics-based discrimination model in patients enrolled at 1 institution (training set) and externally validated the model in patients enrolled at another institution (test set). We performed pathway analysis of proteins dysregulated in patients with prior MACEs. RESULTS A total of 402 patients were included, with 278 in the training set and 124 in the test set. In this cohort, 257 (64%) patients had prior MACEs (172 in the training set and 85 in the test set). Using the proteomics-based model from the training set, the area under the receiver operating characteristic curve was 0.82 (95% confidence interval, 0.75-0.90) in the test set. Patients with prior MACEs demonstrated dysregulation in pathways known to be associated with MACEs (eg, TGF-β) and novel pathways (eg, Ras-MAPK and associated pathways). CONCLUSIONS In this multicenter study of 402 patients with HCM, we identified both known and novel pathways dysregulated in a subset of patients with more advanced disease.
Collapse
Affiliation(s)
- Charlotte Lee
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lusha W Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Albree Tower-Rader
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael A Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Muredach Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY; Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
4
|
Yi JS, Perla S, Bennett AM. An Assessment of the Therapeutic Landscape for the Treatment of Heart Disease in the RASopathies. Cardiovasc Drugs Ther 2023; 37:1193-1204. [PMID: 35156148 PMCID: PMC11726350 DOI: 10.1007/s10557-022-07324-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
The RAS/mitogen-activated protein kinase (MAPK) pathway controls a plethora of developmental and post-developmental processes. It is now clear that mutations in the RAS-MAPK pathway cause developmental diseases collectively referred to as the RASopathies. The RASopathies include Noonan syndrome, Noonan syndrome with multiple lentigines, cardiofaciocutaneous syndrome, neurofibromatosis type 1, and Costello syndrome. RASopathy patients exhibit a wide spectrum of congenital heart defects (CHD), such as valvular abnormalities and hypertrophic cardiomyopathy (HCM). Since the cardiovascular defects are the most serious and recurrent cause of mortality in RASopathy patients, it is critical to understand the pathological signaling mechanisms that drive the disease. Therapies for the treatment of HCM and other RASopathy-associated comorbidities have yet to be fully realized. Recent developments have shown promise for the use of repurposed antineoplastic drugs that target the RAS-MAPK pathway for the treatment of RASopathy-associated HCM. However, given the impact of the RAS-MAPK pathway in post-developmental physiology, establishing safety and evaluating risk when treating children will be paramount. As such insight provided by preclinical and clinical information will be critical. This review will highlight the cardiovascular manifestations caused by the RASopathies and will discuss the emerging therapies for treatment.
Collapse
Affiliation(s)
- Jae-Sung Yi
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
5
|
Li X, Sun T, Liu J, Wei S, Yang Y, Liu J, Zhang B, Li W. Phloretin alleviates doxorubicin-induced cardiotoxicity through regulating Hif3a transcription via targeting transcription factor Fos. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155046. [PMID: 37659297 DOI: 10.1016/j.phymed.2023.155046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Doxorubicin (Dox), a chemotherapeutic agent known for its efficacy, has been associated with the development of severe cardiotoxicity, commonly referred to as doxorubicin-induced cardiotoxicity (DIC). The role and mechanism of action of phloretin (Phl) in cardiovascular diseases are well-established; however, its specific function and underlying mechanism in the context of DIC have yet to be fully elucidated. OBJECTIVE This research aimed to uncover the protective effect of Phl against DIC in vivo and in vitro, while also providing a comprehensive understanding of the underlying mechanisms involved. METHODS DIC cell and murine models were established. The action targets and mechanism of Phl against DIC were comprehensively examined by systematic network pharmacology, molecular docking, transcriptomics technologies, transcription factor (TF) prediction, and experimental validation. RESULTS Phl relieved Dox-induced cell apoptosis in vitro and in vivo. Through network pharmacology analysis, a total of 554 co-targeted genes of Phl and Dox were identified. Enrichment analysis revealed several key pathways including the PI3K-Akt signaling pathway, Apoptosis, and the IL-17 signaling pathway. Protein-protein interaction (PPI) analysis identified 24 core co-targeted genes, such as Fos, Jun, Hif1a, which were predicted to bind well to Phl based on molecular docking. Transcriptomics analysis was performed to identify the top 20 differentially expressed genes (DEGs), and 202 transcription factors (TFs) were predicted for these DEGs. Among these TFs, 10 TFs (Fos, Jun, Hif1a, etc.) are also the co-targeted genes, and 3 TFs (Fos, Jun, Hif1a) are also the core co-targeted genes. Further experiments validated the finding that Phl reduced the elevated levels of Hif3a (one of the top 20 DEGs) and Fos (one of Hif3a's predicted TFs) induced by Dox. Moreover, the interaction between Fos protein and the Hif3a promoter was confirmed through luciferase reporter assays. CONCLUSION Phl actively targeted and down-regulated the Fos protein to inhibit its binding to the promoter region of Hif3a, thereby providing protection against DIC.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiaqin Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China.
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
6
|
Shi H, Chen S, Meng FW, Ossip DJ, Yan C, Li D. Epigenome-wide DNA methylation profiling in comparison between pathological and physiological hypertrophy of human cardiomyocytes. Front Genet 2023; 14:1264382. [PMID: 37829282 PMCID: PMC10565041 DOI: 10.3389/fgene.2023.1264382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Background: Physiological and pathological stimuli result in distinct forms of cardiac hypertrophy, but the molecular regulation comparing the two, especially at the DNA methylation level, is not well understood. Methods: We conducted an in vitro study using human cardiomyocytes exposed to angiotensin II (AngII) and insulin-like growth factor 1 (IGF-1) to mimic pathologically and physiologically hypertrophic heart models, respectively. Whole genome DNA methylation patterns were profiled by the Infinium human MethylationEPIC platform with >850 K DNA methylation loci. Two external datasets were used for comparisons and qRT-PCR was performed for examining expression of associated genes of those identified DNA methylation loci. Results: We detected 194 loci that are significantly differentially methylated after AngII treatment, and 206 significant loci after IGF-1 treatment. Mapping the significant loci to genes, we identified 158 genes corresponding to AngII treatment and 175 genes to IGF-1 treatment. Using the gene-set enrichment analysis, the PI3K-Akt signaling pathway was identified to be significantly enriched for both AngII and IGF-1 treatment. The Hippo signaling pathway was enriched after IGF-1 treatment, but not for AngII treatment. CDK6 and RPTOR are components of the PI3K-Akt pathway but have different DNA methylation patterns in response to AngII and IGF-1. qRT-PCR confirmed the different gene expressions of CDK6 and PRTOR. Conclusion: Our study is pioneering in profiling epigenome DNA methylation changes in adult human cardiomyocytes under distinct stress conditions: pathological (AngII) and physiological (IGF-1). The identified DNA methylation loci, genes, and pathways might have the potential to distinguish between pathological and physiological cardiac hypertrophy.
Collapse
Affiliation(s)
- Hangchuan Shi
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Si Chen
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, NY, United States
| | - Fanju W. Meng
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Deborah J. Ossip
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, NY, United States
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
7
|
Nguyen BY, Zhou F, Binder P, Liu W, Hille SS, Luo X, Zi M, Zhang H, Adamson A, Ahmed FZ, Butterworth S, Cartwright EJ, Müller OJ, Guan K, Fitzgerald EM, Wang X. Prolylcarboxypeptidase Alleviates Hypertensive Cardiac Remodeling by Regulating Myocardial Tissue Angiotensin II. J Am Heart Assoc 2023; 12:e028298. [PMID: 37318028 PMCID: PMC10356030 DOI: 10.1161/jaha.122.028298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/25/2023] [Indexed: 06/16/2023]
Abstract
Background Prolonged activation of angiotensin II is the main mediator that contributes to the development of heart diseases, so converting angiotensin II into angiotensin 1-7 has emerged as a new strategy to attenuate detrimental effects of angiotensin II. Prolylcarboxypeptidase is a lysosomal pro-X carboxypeptidase that is able to cleave angiotensin II at a preferential acidic pH optimum. However, insufficient attention has been given to the cardioprotective functions of prolylcarboxylpeptidase. Methods and Results We established a CRISPR/CRISPR-associated protein 9-mediated global prolylcarboxylpeptidase-knockout and adeno-associated virus serotype 9-mediated cardiac prolylcarboxylpeptidase overexpression mouse models, which were challenged with the angiotensin II infusion (2 mg/kg per day) for 4 weeks, aiming to investigate the cardioprotective effect of prolylcarboxylpeptidase against hypertensive cardiac hypertrophy. Prolylcarboxylpeptidase expression was upregulated after 2 weeks of angiotensin II infusion and then became downregulated afterward in wild-type mouse myocardium, suggesting its compensatory function against angiotensin II stress. Moreover, angiotensin II-treated prolylcarboxylpeptidase-knockout mice showed aggravated cardiac remodeling and dampened cardiac contractility independent of hypertension. We also found that prolylcarboxylpeptidase localizes in cardiomyocyte lysosomes, and loss of prolylcarboxylpeptidase led to excessive angiotensin II levels in myocardial tissue. Further screening demonstrated that hypertrophic prolylcarboxylpeptidase-knockout hearts showed upregulated extracellular signal-regulated kinases 1/2 and downregulated protein kinase B activities. Importantly, adeno-associated virus serotype 9-mediated restoration of prolylcarboxylpeptidase expression in prolylcarboxylpeptidase-knockout hearts alleviated angiotensin II-induced hypertrophy, fibrosis, and cell death. Interestingly, the combination of adeno-associated virus serotype 9-mediated prolylcarboxylpeptidase overexpression and an antihypertensive drug, losartan, likely conferred more effective protection than a single treatment protocol to mitigate angiotensin II-induced cardiac dysfunction. Conclusions Our data demonstrate that prolylcarboxylpeptidase protects the heart from angiotensin II-induced hypertrophic remodeling by controlling myocardial angiotensin II levels.
Collapse
Affiliation(s)
- Binh Y. Nguyen
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Fangchao Zhou
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Pablo Binder
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Wei Liu
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Susanne S. Hille
- Department of Internal Medicine IIIUniversity of KielKielGermany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav CarusTechnische Universitaet DresdenDresdenGermany
| | - Min Zi
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Hongyuan Zhang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Antony Adamson
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Fozia Z. Ahmed
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | - Sam Butterworth
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| | | | - Oliver J. Müller
- Department of Internal Medicine IIIUniversity of KielKielGermany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav CarusTechnische Universitaet DresdenDresdenGermany
| | | | - Xin Wang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
8
|
Chakrabarti M, Raut GK, Jain N, Bhadra MP. Prohibitin1 maintains mitochondrial quality in isoproterenol-induced cardiac hypertrophy in H9C2 cells. Biol Cell 2023; 115:e2200094. [PMID: 36453777 DOI: 10.1111/boc.202200094] [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: 10/04/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND INFORMATION Various types of stress initially induce a state of cardiac hypertrophy (CH) in the heart. But, persistent escalation of cardiac stress leads to progression from an adaptive physiological to a maladaptive pathological state. So, elucidating molecular mechanisms that can attenuate CH is imperative in developing cardiac therapies. Previously, we showed that Prohibitin1 (PHB1) has a protective role in CH-induced oxidative stress. Nevertheless, it is unclear how PHB1, a mitochondrial protein, has a protective role in CH. Therefore, we hypothesized that PHB1 maintains mitochondrial quality in CH. To test this hypothesis, we used Isoproterenol (ISO) to induce CH in H9C2 cells overexpressing PHB1 and elucidated mitochondrial quality control pathways. RESULTS We found that overexpressing PHB1 attenuates ISO-induced CH and restores mitochondrial morphology in H9C2 cells. In addition, PHB1 blocks the pro-hypertrophic IGF1R/AKT pathway and restores the mitochondrial membrane polarization in ISO-treated cells. We observed that overexpressing PHB1 promotes mitochondrial biogenesis, improves mitochondrial respiratory capacity, and triggers mitophagy. CONCLUSION We conclude that PHB1 maintains mitochondrial quality in ISO-induced CH in H9C2 cells. SIGNIFICANCE Based on our results, we suggest that small molecules that induce PHB1 in cardiac cells may prove beneficial in developing cardiac therapies.
Collapse
Affiliation(s)
- Moumita Chakrabarti
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ganesh Kumar Raut
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Nishant Jain
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manika Pal Bhadra
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
9
|
Liu X, Li D, Pi W, Wang B, Xu S, Yu L, Yao L, Sun Z, Jiang J, Mi Y. LCZ696 protects against doxorubicin-induced cardiotoxicity by inhibiting ferroptosis via AKT/SIRT3/SOD2 signaling pathway activation. Int Immunopharmacol 2022; 113:109379. [PMID: 36330913 DOI: 10.1016/j.intimp.2022.109379] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022]
Abstract
Doxorubicin (DOX) is an effective and widely used anticancer drug but has limited clinical applicability because of its cardiotoxicity. Ferroptosis plays a key role in DOX-induced cardiac damage and cardiomyocyte cell death. The inhibition of ferroptosis reverses DOX-induced cardiotoxicity (DIC). LCZ696, a first-in-class angiotensin receptor neprilysin inhibitor, protects against DIC. However, the mechanism of action of LCZ696, especially its effect on ferroptosis, is incompletely understood. This study investigates the cardioprotective effects of LCZ696 on DIC in vivo and in vitro.Cardiotoxicity was induced in Wistar rats by tail intravenous injection of 2.5 mg/kg DOX once a week for six weeks. Rats and H9c2 cells were treated with or without LCZ696 to determine the cardioprotective role and underlying mechanisms of LCZ696 against DIC. To assess the role of SIRT3 and correlated pathways in ferroptosis, SIRT3 knockout was performed using lentiviral vectors, and AKT was inhibited with LY294002. LCZ696 significantly attenuated DIC by decreasing the concentrations of lipid reactive oxygen species and malondialdehyde and increasing the levels of glutathione peroxidase-4 and reduced glutathione in cells and heart tissues. Moreover, LCZ696 remodeled myocardial structures and improved heart ventricular function in DOX-treated rats. LCZ696 treatment increased SIRT3 expression and deacetylated its target gene SOD2, and these changes were mediated by AKT activation. SIRT3 knockdown and AKT inhibition induced lipid peroxidation and reduced the protective effect of LCZ696 in H9c2 cells. Collectively,LCZ696 prevents DIC by inhibiting ferroptosis via AKT/SIRT3/SOD2 signaling pathway activation. Thus, LZC696 is a potential therapeutic strategy for DIC.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Danlei Li
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Bin Wang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Shasha Xu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Lei Yu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Lei Yao
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| | - Yafei Mi
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| |
Collapse
|
10
|
Balamurugan K, Chandra K, Sai Latha S, Swathi M, Joshi MB, Misra P, Parsa KVL. PHLPPs: Emerging players in metabolic disorders. Drug Discov Today 2022; 27:103317. [PMID: 35835313 DOI: 10.1016/j.drudis.2022.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 12/15/2022]
Abstract
That reversible protein phosphorylation by kinases and phosphatases occurs in metabolic disorders is well known. Various studies have revealed that a multi-faceted and tightly regulated phosphatase, pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP)-1/2 displays robust effects in cardioprotection, ischaemia/reperfusion (I/R), and vascular remodelling. PHLPP1 promotes foamy macrophage development through ChREBP/AMPK-dependent pathways. Adipocyte-specific loss of PHLPP2 reduces adiposity, improves glucose tolerance,and attenuates fatty liver via the PHLPP2-HSL-PPARα axis. Discoveries of PHLPP1-mediated insulin resistance and pancreatic β cell death via the PHLPP1/2-Mst1-mTORC1 triangular loop have shed light on its significance in diabetology. PHLPP1 downregulation attenuates diabetic cardiomyopathy (DCM) by restoring PI3K-Akt-mTOR signalling. In this review, we summarise the functional role of, and cellular signalling mediated by, PHLPPs in metabolic tissues and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keerthana Balamurugan
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Kanika Chandra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - S Sai Latha
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - M Swathi
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Kishore V L Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India.
| |
Collapse
|
11
|
Shen S, Sewanan LR, Shao S, Halder SS, Stankey P, Li X, Campbell SG. Physiological calcium combined with electrical pacing accelerates maturation of human engineered heart tissue. Stem Cell Reports 2022; 17:2037-2049. [PMID: 35931080 PMCID: PMC9481907 DOI: 10.1016/j.stemcr.2022.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have wide potential application in basic research, drug discovery, and regenerative medicine, but functional maturation remains challenging. Here, we present a method whereby maturation of hiPSC-CMs can be accelerated by simultaneous application of physiological Ca2+ and frequency-ramped electrical pacing in culture. This combination produces positive force-frequency behavior, physiological twitch kinetics, robust β-adrenergic response, improved Ca2+ handling, and cardiac troponin I expression within 25 days. This study provides insights into the role of Ca2+ in hiPSC-CM maturation and offers a scalable platform for translational and clinical research.
Collapse
Affiliation(s)
- Shi Shen
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Lorenzo R Sewanan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephanie Shao
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Saiti S Halder
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Paul Stankey
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Xia Li
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA.
| |
Collapse
|
12
|
Støle TP, Lunde M, Shen X, Martinsen M, Lunde PK, Li J, Lockwood F, Sjaastad I, Louch WE, Aronsen JM, Christensen G, Carlson CR. The female syndecan-4−/− heart has smaller cardiomyocytes, augmented insulin/pSer473-Akt/pSer9-GSK-3β signaling, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels. Front Cell Dev Biol 2022; 10:908126. [PMID: 36092718 PMCID: PMC9452846 DOI: 10.3389/fcell.2022.908126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In cardiac muscle, the ubiquitously expressed proteoglycan syndecan-4 is involved in the hypertrophic response to pressure overload. Protein kinase Akt signaling, which is known to regulate hypertrophy, has been found to be reduced in the cardiac muscle of exercised male syndecan-4−/− mice. In contrast, we have recently found that pSer473-Akt signaling is elevated in the skeletal muscle (tibialis anterior, TA) of female syndecan-4−/− mice. To determine if the differences seen in Akt signaling are sex specific, we have presently investigated Akt signaling in the cardiac muscle of sedentary and exercised female syndecan-4−/− mice. To get deeper insight into the female syndecan-4−/− heart, alterations in cardiomyocyte size, a wide variety of different extracellular matrix components, well-known syndecan-4 binding partners and associated signaling pathways have also been investigated.Methods: Left ventricles (LVs) from sedentary and exercise trained female syndecan-4−/− and WT mice were analyzed by immunoblotting and real-time PCR. Cardiomyocyte size and phosphorylated Ser473-Akt were analyzed in isolated adult cardiomyocytes from female syndecan-4−/− and WT mice by confocal imaging. LV and skeletal muscle (TA) from sedentary male syndecan-4−/− and WT mice were immunoblotted with Akt antibodies for comparison. Glucose levels were measured by a glucometer, and fasting blood serum insulin and C-peptide levels were measured by ELISA.Results: Compared to female WT hearts, sedentary female syndecan-4−/− LV cardiomyocytes were smaller and hearts had higher levels of pSer473-Akt and its downstream target pSer9-GSK-3β. The pSer473-Akt inhibitory phosphatase PHLPP1/SCOP was lowered, which may be in response to the elevated serum insulin levels found in the female syndecan-4−/− mice. We also observed lowered levels of pThr308-Akt/Akt and GLUT4 in the female syndecan-4−/− heart and an increased LRP6 level after exercise. Otherwise, few alterations were found. The pThr308-Akt and pSer473-Akt levels were unaltered in the cardiac and skeletal muscles of sedentary male syndecan-4−/− mice.Conclusion: Our data indicate smaller cardiomyocytes, an elevated insulin/pSer473-Akt/pSer9-GSK-3β signaling pathway, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels in the female syndecan-4−/− heart. In contrast, cardiomyocyte size, and Akt signaling were unaltered in both cardiac and skeletal muscles from male syndecan-4−/− mice, suggesting important sex differences.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- *Correspondence: Thea Parsberg Støle,
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Marita Martinsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Francesca Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - William Edward Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Charles S, Sreekumar J, Natarajan J. Transcriptomic meta-analysis reveals biomarker pairs and key pathways in Tetralogy of Fallot. J Bioinform Comput Biol 2022; 20:2240004. [DOI: 10.1142/s0219720022400042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
14
|
Mprah R, Ma Y, Adzika GK, Noah MLN, Adekunle AO, Duah M, Joseph A, Wowui PI, Okwuma JD, Weili Q, Cheng W. Metabotropic Glutamate Receptor 5 Blockade Attenuates Pathological Cardiac Remodeling in Pulmonary Arterial Hypertension. Clin Exp Pharmacol Physiol 2022; 49:558-566. [PMID: 35133684 DOI: 10.1111/1440-1681.13633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/02/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Richard Mprah
- Department of Physiology Xuzhou Medical University Xuzhou 221004 Jiangsu China
| | - Yanhong Ma
- Department of Physiology Xuzhou Medical University Xuzhou 221004 Jiangsu China
| | | | | | - Adebayo O. Adekunle
- Department of Physiology Xuzhou Medical University Xuzhou 221004 Jiangsu China
| | - Maxwell Duah
- Haematology Department Affiliated Hospital of Xuzhou Medical University Xuzhou 221006 Jiangsu China
| | | | | | | | - Qiao Weili
- Department of Physiology Xuzhou Medical University Xuzhou 221004 Jiangsu China
| | - Wang Cheng
- Department of Cardiology Affiliated Hospital of Xuzhou Medical University Xuzhou 221006 Jiangsu China
| |
Collapse
|
15
|
A mouse model of inherited choline kinase β-deficiency presents with specific cardiac abnormalities and a predisposition to arrhythmia. J Biol Chem 2022; 298:101716. [PMID: 35151687 PMCID: PMC8913350 DOI: 10.1016/j.jbc.2022.101716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
The CHKB gene encodes choline kinase β, which catalyzes the first step in the biosynthetic pathway for the major phospholipid phosphatidylcholine. Homozygous loss-of-function variants in human CHKB are associated with a congenital muscular dystrophy. Dilated cardiomyopathy is present in some CHKB patients and can cause heart failure and death. Mechanisms underlying a cardiac phenotype due to decreased CHKB levels are not well characterized. We determined that there is cardiac hypertrophy in Chkb−/− mice along with a decrease in left ventricle size, internal diameter, and stroke volume compared with wildtype and Chkb+/− mice. Unlike wildtype mice, 60% of the Chkb+/− and all Chkb−/− mice tested displayed arrhythmic events when challenged with isoproterenol. Lipidomic analysis revealed that the major change in lipid level in Chkb+/− and Chkb−/− hearts was an increase in the arrhythmogenic lipid acylcarnitine. An increase in acylcarnitine level is also associated with a defect in the ability of mitochondria to use fatty acids for energy and we observed that mitochondria from Chkb−/− hearts had abnormal cristae and inefficient electron transport chain activity. Atrial natriuretic peptide (ANP) is a hormone produced by the heart that protects against the development of heart failure including ventricular conduction defects. We determined that there was a decrease in expression of ANP, its receptor NPRA, as well as ventricular conduction system markers in Chkb+/− and Chkb−/− mice.
Collapse
|
16
|
Sen S, Hallee L, Lam CK. The Potential of Gamma Secretase as a Therapeutic Target for Cardiac Diseases. J Pers Med 2021; 11:jpm11121294. [PMID: 34945766 PMCID: PMC8703931 DOI: 10.3390/jpm11121294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Heart diseases are some of the most common and pressing threats to human health worldwide. The American Heart Association and the National Institute of Health jointly work to annually update data on cardiac diseases. In 2018, 126.9 million Americans were reported as having some form of cardiac disorder, with an estimated direct and indirect total cost of USD 363.4 billion. This necessitates developing therapeutic interventions for heart diseases to improve human life expectancy and economic relief. In this review, we look into gamma-secretase as a potential therapeutic target for cardiac diseases. Gamma-secretase, an aspartyl protease enzyme, is responsible for the cleavage and activation of a number of substrates that are relevant to normal cardiac development and function as found in mutation studies. Some of these substrates are involved in downstream signaling processes and crosstalk with pathways relevant to heart diseases. Most of the substrates and signaling events we explored were found to be potentially beneficial to maintain cardiac function in diseased conditions. This review presents an updated overview of the current knowledge on gamma-secretase processing of cardiac-relevant substrates and seeks to understand if the modulation of gamma-secretase activity would be beneficial to combat cardiac diseases.
Collapse
Affiliation(s)
- Sujoita Sen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Logan Hallee
- Department of Mathematical Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Correspondence: ; Tel.: +1-302-831-3165
| |
Collapse
|
17
|
Al-Shamasi AA, Elkaffash R, Mohamed M, Rayan M, Al-Khater D, Gadeau AP, Ahmed R, Hasan A, Eldassouki H, Yalcin HC, Abdul-Ghani M, Mraiche F. Crosstalk between Sodium-Glucose Cotransporter Inhibitors and Sodium-Hydrogen Exchanger 1 and 3 in Cardiometabolic Diseases. Int J Mol Sci 2021; 22:12677. [PMID: 34884494 PMCID: PMC8657861 DOI: 10.3390/ijms222312677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormality in glucose homeostasis due to hyperglycemia or insulin resistance is the hallmark of type 2 diabetes mellitus (T2DM). These metabolic abnormalities in T2DM lead to cellular dysfunction and the development of diabetic cardiomyopathy leading to heart failure. New antihyperglycemic agents including glucagon-like peptide-1 receptor agonists and the sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been shown to attenuate endothelial dysfunction at the cellular level. In addition, they improved cardiovascular safety by exhibiting cardioprotective effects. The mechanism by which these drugs exert their cardioprotective effects is unknown, although recent studies have shown that cardiovascular homeostasis occurs through the interplay of the sodium-hydrogen exchangers (NHE), specifically NHE1 and NHE3, with SGLT2i. Another theoretical explanation for the cardioprotective effects of SGLT2i is through natriuresis by the kidney. This theory highlights the possible involvement of renal NHE transporters in the management of heart failure. This review outlines the possible mechanisms responsible for causing diabetic cardiomyopathy and discusses the interaction between NHE and SGLT2i in cardiovascular diseases.
Collapse
Affiliation(s)
- Al-Anood Al-Shamasi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Rozina Elkaffash
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Meram Mohamed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Dhabya Al-Khater
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Alain-Pierre Gadeau
- INSERM, Biology of Cardiovascular Disease, University of Bordeaux, U1034 Pessac, France;
| | - Rashid Ahmed
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Anwarul Hasan
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussein Eldassouki
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | | | - Muhammad Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center at San Antonio, Floyd Curl Drive, San Antonio, TX 7703, USA;
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
18
|
Berg PC, Hansson ÅML, Røsand Ø, Marwarha G, Høydal MA. Overexpression of Neuron-Derived Orphan Receptor 1 (NOR-1) Rescues Cardiomyocytes from Cell Death and Improves Viability after Doxorubicin Induced Stress. Biomedicines 2021; 9:1233. [PMID: 34572418 PMCID: PMC8471245 DOI: 10.3390/biomedicines9091233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022] Open
Abstract
Following myocardial infarction, reperfusion injury (RI) is commonly observed due to the excessive formation of, e.g., reactive oxygen species (ROS). Doxorubicin (DOX), a widely used anti-cancer drug, is also known to cause cardiotoxicity due to excessive ROS production. Exercise training has been shown to protect the heart against both RI- and DOX-induced cardiotoxicity, but the exact mechanism is still unknown. Neuron-derived orphan receptor 1 (NOR-1) is an important exercise-responsive protein in the skeletal muscle which has also been reported to facilitate cellular survival during hypoxia. Therefore, we hypothesized that NOR-1 could protect cardiomyocytes (CMs) against cellular stress induced by DOX. We also hypothesized that NOR-1 is involved in preparing the CMs against a stress situation during nonstimulated conditions by increasing cell viability. To determine the protective effect of NOR-1 in CMs stressed with DOX challenge, we overexpressed NOR-1 in AC16 human CMs treated with 5 µM DOX for 12 h or the respective vehicle control, followed by performing Lactate dehydrogenase (LDH) activity, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and caspase-3 activity assays to measure cell death, cell viability, and apoptosis, respectively. In addition, Western blotting analysis was performed to determine the expression of key proteins involved in cardioprotection. We demonstrated that NOR-1 overexpression decreased cell death (p < 0.105) and apoptosis (p < 0.01) while increasing cell viability (p < 0.05) in DOX-treated CMs. We also observed that NOR-1 overexpression increased phosphorylation of extracellular signal-regulated kinase (ERK) (p < 0.01) and protein expression levels of B cell lymphoma extra-large (Bcl-xL) (p < 0.01). We did not detect any significant changes in phosphorylation of protein kinase B (Akt), glycogen synthase kinase-3β (GSK-3β) and signal transducer and activator of transcription 3 (STAT3) or expression levels of superoxide dismutase 2 (SOD2) and cyclin D1. Furthermore, we demonstrated that NOR-1 overexpression increased the cell viability (p < 0.0001) of CMs during nonstimulated conditions without affecting cell death or apoptosis. Our findings indicate that NOR-1 could serve as a potential cardioprotective protein in response to Doxorubicin-induced cellular stress.
Collapse
Affiliation(s)
| | | | | | | | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Technology and Science (NTNU), 7030 Trondheim, Norway; (P.-C.B.); (Å.M.L.H.); (Ø.R.); (G.M.)
| |
Collapse
|
19
|
Guo G, Watterson S, Zhang SD, Bjourson A, McGilligan V, Peace A, Rai TS. The role of senescence in the pathogenesis of atrial fibrillation: A target process for health improvement and drug development. Ageing Res Rev 2021; 69:101363. [PMID: 34023420 DOI: 10.1016/j.arr.2021.101363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a state of growth arrest that occurs after cells encounter various stresses. Senescence contributes to tumour suppression, embryonic development, and wound healing. It impacts on the pathology of various diseases by secreting inflammatory chemokines, immune modulators and other bioactive factors. These secretory biosignatures ultimately cause inflammation, tissue fibrosis, immunosenescence and many ageing-related diseases such as atrial fibrillation (AF). Because the molecular mechanisms underpinning AF development remain unclear, current treatments are suboptimal and have serious side effects. In this review, we summarize recent results describing the role of senescence in AF. We propose that senescence factors induce AF and have a causative role. Hence, targeting senescence and its secretory phenotype may attenuate AF.
Collapse
|
20
|
Wingard MC, Dalal S, Shook PL, Myers R, Connelly BA, Thewke DP, Singh M, Singh K. Deficiency of ataxia-telangiectasia mutated kinase modulates functional and biochemical parameters of the heart in response to Western-type diet. Am J Physiol Heart Circ Physiol 2021; 320:H2324-H2338. [PMID: 33929897 PMCID: PMC8289354 DOI: 10.1152/ajpheart.00990.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 02/08/2023]
Abstract
Ataxia-telangiectasia mutated (ATM) kinase deficiency exacerbates heart dysfunction late after myocardial infarction. Here, we hypothesized that ATM deficiency modulates Western-type diet (WD)-induced cardiac remodeling with an emphasis on functional and biochemical parameters of the heart. Weight gain was assessed in male wild-type (WT) and ATM heterozygous knockout (hKO) mice on weekly basis, whereas cardiac functional and biochemical parameters were measured 14 wk post-WD. hKO-WD mice exhibited rapid body weight gain at weeks 5, 6, 7, 8, and 10 versus WT-WD. WD decreased percent fractional shortening and ejection fraction, and increased end-systolic volumes and diameters to a similar extent in both genotypes. However, WD decreased stroke volume, cardiac output, peak velocity of early ventricular filling, and aortic ejection time and increased isovolumetric relaxation time (IVRT) and Tei index versus WT-NC (normal chow). Conversely, IVRT, isovolumetric contraction time, and Tei index were lower in hKO-WD versus hKO-NC and WT-WD. Myocyte apoptosis and hypertrophy were higher in hKO-WD versus WT-WD. WD increased fibrosis and expression of collagen-1α1, matrix metalloproteinase (MMP)-2, and MMP-9 in WT. WD enhanced AMPK activation, while decreasing mTOR activation in hKO. Akt and IKK-α/β activation, and Bax, PARP-1, and Glut-4 expression were higher in WT-WD versus WT-NC, whereas NF-κB activation and Glut-4 expression were lower in hKO-WD versus hKO-NC. Circulating concentrations of IL-12(p70), eotaxin, IFN-γ, macrophage inflammatory protein (MIP)-1α, and MIP-1β were higher in hKO-WD versus WT-WD. Thus, ATM deficiency accelerates weight gain, induces systolic dysfunction with increased preload, and associates with increased apoptosis, hypertrophy, and inflammation in response to WD.NEW & NOTEWORTHY Ataxia-telangiectasia mutated (ATM) kinase deficiency in humans associates with enhanced susceptibility to ischemic heart disease. Here, we provide evidence that ATM deficiency accelerates body weight gain and associates with increased cardiac preload, hypertrophy, and apoptosis in mice fed with Western-type diet (WD). Further investigations of the role of ATM deficiency in WD-induced alterations in function and biochemical parameters of the heart may provide clinically applicable information on treatment and/or nutritional counseling for patients with ATM deficiency.
Collapse
Affiliation(s)
- Mary C Wingard
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Suman Dalal
- Department of Health Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Paige L Shook
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Rachel Myers
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Barbara A Connelly
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| | - Douglas P Thewke
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
21
|
Jozefczuk E, Szczepaniak P, Guzik TJ, Siedlinski M. Silencing of Sphingosine kinase 1 Affects Maturation Pathways in Mouse Neonatal Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22073616. [PMID: 33807180 PMCID: PMC8037404 DOI: 10.3390/ijms22073616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Sphingosine kinase-1 (Sphk1) and its product, sphingosine-1-phosphate (S1P) are important regulators of cardiac growth and function. Numerous studies have reported that Sphk1/S1P signaling is essential for embryonic cardiac development and promotes pathological cardiac hypertrophy in adulthood. However, no studies have addressed the role of Sphk1 in postnatal cardiomyocyte (CM) development so far. The present study aimed to assess the molecular mechanism(s) by which Sphk1 silencing might influence CMs development and hypertrophy in vitro. Neonatal mouse CMs were transfected with siRNA against Sphk1 or negative control, and subsequently treated with 1 µM angiotensin II (AngII) or a control buffer for 24 h. The results of RNASeq analysis revealed that diminished expression of Sphk1 significantly accelerated neonatal CM maturation by inhibiting cell proliferation and inducing developmental pathways in the stress (AngII-induced) conditions. Importantly, similar effects were observed in the control conditions. Enhanced maturation of Sphk1-lacking CMs was further confirmed by the upregulation of the physiological hypertrophy-related signaling pathway involving Akt and downstream glycogen synthase kinase 3 beta (Gsk3β) downregulation. In summary, we demonstrated that the Sphk1 silencing in neonatal mouse CMs facilitated their postnatal maturation in both physiological and stress conditions.
Collapse
Affiliation(s)
- Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Tomasz Jan Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
- Correspondence:
| |
Collapse
|
22
|
Wang J, Bai J, Duan P, Wang H, Li Y, Zhu Q. Kir6.1 improves cardiac dysfunction in diabetic cardiomyopathy via the AKT-FoxO1 signalling pathway. J Cell Mol Med 2021; 25:3935-3949. [PMID: 33547878 PMCID: PMC8051713 DOI: 10.1111/jcmm.16346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that the expression of inwardly rectifying potassium channel 6.1 (Kir6.1) in heart mitochondria is significantly reduced in type 1 diabetes. However, whether its expression and function are changed and what role it plays in type 2 diabetic cardiomyopathy (DCM) have not been reported. This study investigated the role and mechanism of Kir6.1 in DCM. We found that the cardiac function and the Kir6.1 expression in DCM mice were decreased. We generated mice overexpressing or lacking Kir6.1 gene specifically in the heart. Kir6.1 overexpression improved cardiac dysfunction in DCM. Cardiac‐specific Kir6.1 knockout aggravated cardiac dysfunction. Kir6.1 regulated the phosphorylation of AKT and Foxo1 in DCM. We further found that Kir6.1 overexpression also improved cardiomyocyte dysfunction and up‐regulated the phosphorylation of AKT and FoxO1 in neonatal rat ventricular cardiomyocytes with insulin resistance. Furthermore, FoxO1 activation down‐regulated the expression of Kir6.1 and decreased the mitochondrial membrane potential (ΔΨm) in cardiomyocytes. FoxO1 inactivation up‐regulated the expression of Kir6.1 and increased the ΔΨm in cardiomyocytes. Chromatin immunoprecipitation assay demonstrated that the Kir6.1 promoter region contains a functional FoxO1‐binding site. In conclusion, Kir6.1 improves cardiac dysfunction in DCM, probably through the AKT‐FoxO1 signalling pathway.
Collapse
Affiliation(s)
- Jinxin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.,Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jing Bai
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Peng Duan
- Department of Cardiology, Chinese PLA No. 371 Hospital, Henan, China
| | - Hao Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinglei Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Gu L, Xie C, Peng Q, Zhang J, Li J, Tang Z. Arecoline suppresses epithelial cell viability through the Akt/mTOR signaling pathway via upregulation of PHLPP2. Toxicology 2019; 419:32-39. [PMID: 30910432 DOI: 10.1016/j.tox.2019.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/19/2019] [Accepted: 03/20/2019] [Indexed: 12/16/2022]
Abstract
Arecoline, the major active ingredient of the betel nut, is involved in the pathogenesis of oral submucous fibrosis. However, the underlying mechanism of this pathogenesis remains unclear. In this study, we found that arecoline suppresses the cell proliferation of the HaCaT epithelial cell and induces cell cycle arrest at the G1/S phase with an IC50 of 50 μg/mL. Furthermore, we found that arecoline reduces the protein level of cyclin D1, but it has no effect on its mRNA level and protein stability, implying that arecoline may modulate the translation of cyclin D1. We also observed the downregulation of the Akt/mTOR signaling pathway after treatment with arecoline, which may be related to the translation of cyclin D1. RNA-seq analysis identified that PHLPP2, the direct upstream target of Akt, is significantly upregulated after arecoline treatment. siRNA-mediated knockdown of PHLPP2 recovered the phosphorylation state of Akt, as well as attenuated the effect of arecoline on cell viability. Thus, our study revealed the crucial role of PHLPP2 in arecoline-induced cell viability suppression.
Collapse
Affiliation(s)
- Liqun Gu
- Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China; Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Changqing Xie
- Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Qian Peng
- Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Jiaming Zhang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Jiada Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Zhangui Tang
- Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China; Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
24
|
Ackermann MA, King B, Lieberman NAP, Bobbili PJ, Rudloff M, Berndsen CE, Wright NT, Hecker PA, Kontrogianni-Konstantopoulos A. Novel obscurins mediate cardiomyocyte adhesion and size via the PI3K/AKT/mTOR signaling pathway. J Mol Cell Cardiol 2017; 111:27-39. [PMID: 28826662 PMCID: PMC5694667 DOI: 10.1016/j.yjmcc.2017.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 12/29/2022]
Abstract
The intercalated disc of cardiac muscle embodies a highly-ordered, multifunctional network, essential for the synchronous contraction of the heart. Over 200 known proteins localize to the intercalated disc. The challenge now lies in their characterization as it relates to the coupling of neighboring cells and whole heart function. Using molecular, biochemical and imaging techniques, we characterized for the first time two small obscurin isoforms, obscurin-40 and obscurin-80, which are enriched at distinct locations of the intercalated disc. Both proteins bind specifically and directly to select phospholipids via their pleckstrin homology (PH) domain. Overexpression of either isoform or the PH-domain in cardiomyocytes results in decreased cell adhesion and size via reduced activation of the PI3K/AKT/mTOR pathway that is intimately linked to cardiac hypertrophy. In addition, obscurin-80 and obscurin-40 are significantly reduced in acute (myocardial infarction) and chronic (pressure overload) murine cardiac-stress models underscoring their key role in maintaining cardiac homeostasis. Our novel findings implicate small obscurins in the maintenance of cardiomyocyte size and coupling, and the development of heart failure by antagonizing the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Maegen A Ackermann
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States; Department of Physiology and Cell Biology, Wexner College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States.
| | - Brendan King
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States
| | - Nicole A P Lieberman
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States
| | - Prameela J Bobbili
- Department of Physiology and Cell Biology, Wexner College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States
| | - Michael Rudloff
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Christopher E Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Peter A Hecker
- Division of Cardiology and Department of Medicine, University of Maryland, Baltimore, MD 20201, United States
| | | |
Collapse
|
25
|
Ma Y, Yang L, Ma J, Lu L, Wang X, Ren J, Yang J. Rutin attenuates doxorubicin-induced cardiotoxicity via regulating autophagy and apoptosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1904-1911. [PMID: 28069395 DOI: 10.1016/j.bbadis.2016.12.021] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/01/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
Abstract
Doxorubicin as anticancer agent can cause dose-dependent cardiotoxicity and heart failure in the long term. Rutin as a polyphenolic flavonoid has been illustrated to protect hearts from diverse cardiovascular diseases. Its function is known to be related to its antioxidant and antiinflammatory activity which may regulate multiple cellular signal pathways. However, the role of rutin on doxorubicin-induced cardiotoxicity has yet to be discovered. In this study, we explored the protective role of rutin on doxorubicin-induced heart failure and elucidated the potential mechanisms of protective effects of rutin against cardiomyocyte death. We analyzed cardiac tissues at the time point of 8weeks after doxorubicin treatment. The results by echocardiography, TUNEL staining, Masson's trichrome staining as well as Western blot analysis revealed that doxorubicin induced remarkable cardiac dysfunction and cardiotoxicity in mice hearts and cardiomyocytes, which were alleviated by rutin treatment. Western blot analysis indicated that the underlying mechanisms included inhibition excessive autophagy and apoptosis mediated by Akt activation. Collectively, our findings suggest that suppression of autophagy and apoptosis by administration of rutin could attenuate doxorubicin-induced cardiotoxicity, which enhances our knowledge to explore new drugs and strategies for combating this devastating side effect induced by doxorubicin. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Yanyan Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lifang Yang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jipeng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jun Ren
- College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA.
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
26
|
Gao J, Chen T, Zhao D, Zheng J, Liu Z. Ginkgolide B Exerts Cardioprotective Properties against Doxorubicin-Induced Cardiotoxicity by Regulating Reactive Oxygen Species, Akt and Calcium Signaling Pathways In Vitro and In Vivo. PLoS One 2016; 11:e0168219. [PMID: 27973574 PMCID: PMC5156426 DOI: 10.1371/journal.pone.0168219] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/08/2016] [Indexed: 12/30/2022] Open
Abstract
The aim of this study was to evaluate the effect of Ginkgolide B (GB) on doxorubicin (DOX) induced cardiotoxicity in vitro and in vivo. Rat cardiomyocyte cell line H9c2 was pretreated with GB and subsequently subjected to doxorubicin treatment. Cell viability and cell apoptosis were assessed by MTT assay and Hoechst staining, respectively. Reactive oxygen species (ROS), Akt phosphorylation and intracellular calcium were equally determined in order to explore the underlying molecular mechanism. To verify the in vivo therapeutic effect of GB, we established a mouse model of cardiotoxicity and determined left ventricle ejection fraction (LVEF) and left ventricular mass (LVM). The in vitro experimental results indicated that pretreatment with GB significantly decreases the viability and apoptosis of H9c2 cells by decreasing ROS and intracellular calcium levels and activating Akt phosphorylation. In the in vivo study, we recorded an improved LVEF and a decreased LVM in the group of cardiotoxic rats treated with GB. Altogether, our findings anticipate that GB exerts a cardioprotective effect through possible regulation of the ROS, Akt and calcium pathways. The findings suggest that combination of GB with DOX in chemotherapy could help avoid the cardiotoxic side effects of GB.
Collapse
Affiliation(s)
- Junqing Gao
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Tao Chen
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Deqiang Zhao
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jianpu Zheng
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zongjun Liu
- Department of Cardiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
27
|
Nishiga M, Horie T, Kuwabara Y, Nagao K, Baba O, Nakao T, Nishino T, Hakuno D, Nakashima Y, Nishi H, Nakazeki F, Ide Y, Koyama S, Kimura M, Hanada R, Nakamura T, Inada T, Hasegawa K, Conway SJ, Kita T, Kimura T, Ono K. MicroRNA-33 Controls Adaptive Fibrotic Response in the Remodeling Heart by Preserving Lipid Raft Cholesterol. Circ Res 2016; 120:835-847. [PMID: 27920122 DOI: 10.1161/circresaha.116.309528] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/27/2016] [Accepted: 12/05/2016] [Indexed: 12/13/2022]
Abstract
RATIONALE Heart failure and atherosclerosis share the underlying mechanisms of chronic inflammation followed by fibrosis. A highly conserved microRNA (miR), miR-33, is considered as a potential therapeutic target for atherosclerosis because it regulates lipid metabolism and inflammation. However, the role of miR-33 in heart failure remains to be elucidated. OBJECTIVE To clarify the role of miR-33 involved in heart failure. METHODS AND RESULTS We first investigated the expression levels of miR-33a/b in human cardiac tissue samples with dilated cardiomyopathy. Increased expression of miR-33a was associated with improving hemodynamic parameters. To clarify the role of miR-33 in remodeling hearts, we investigated the responses to pressure overload by transverse aortic constriction in miR-33-deficient (knockout [KO]) mice. When mice were subjected to transverse aortic constriction, miR-33 expression levels were significantly upregulated in wild-type left ventricles. There was no difference in hypertrophic responses between wild-type and miR-33KO hearts, whereas cardiac fibrosis was ameliorated in miR-33KO hearts compared with wild-type hearts. Despite the ameliorated cardiac fibrosis, miR-33KO mice showed impaired systolic function after transverse aortic constriction. We also found that cardiac fibroblasts were mainly responsible for miR-33 expression in the heart. Deficiency of miR-33 impaired cardiac fibroblast proliferation, which was considered to be caused by altered lipid raft cholesterol content. Moreover, cardiac fibroblast-specific miR-33-deficient mice also showed decreased cardiac fibrosis induced by transverse aortic constriction as systemic miR-33KO mice. CONCLUSION Our results demonstrate that miR-33 is involved in cardiac remodeling, and it preserves lipid raft cholesterol content in fibroblasts and maintains adaptive fibrotic responses in the remodeling heart.
Collapse
Affiliation(s)
- Masataka Nishiga
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Takahiro Horie
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Yasuhide Kuwabara
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Kazuya Nagao
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Osamu Baba
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Tetsushi Nakao
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Tomohiro Nishino
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Daihiko Hakuno
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Yasuhiro Nakashima
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Hitoo Nishi
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Fumiko Nakazeki
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Yuya Ide
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Satoshi Koyama
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Masahiro Kimura
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Ritsuko Hanada
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Tomoyuki Nakamura
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Tsukasa Inada
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Koji Hasegawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Simon J Conway
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Toru Kita
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Takeshi Kimura
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita)
| | - Koh Ono
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan (M.N., T.H., Y.K., O.B., T.Nakao, T.Nishino, D.H., Y.N., H.N., F.N., Y.I., S.K., M.K., R.H., T.Kimura, K.O.); Department of Cardiovascular Center, Osaka Red Cross Hospital, Japan (K.N., T.I.); Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan (T.Nakamura); Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Japan (K.H.); Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University of Medicine, Indianapolis (S.J.C.); and Kobe City Medical Center General Hospital, Japan (T.Kita).
| |
Collapse
|
28
|
Fang X, Stroud MJ, Ouyang K, Fang L, Zhang J, Dalton ND, Gu Y, Wu T, Peterson KL, Huang HD, Chen J, Wang N. Adipocyte-specific loss of PPAR γ attenuates cardiac hypertrophy. JCI Insight 2016; 1:e89908. [PMID: 27734035 DOI: 10.1172/jci.insight.89908] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a key endocrine organ that governs systemic homeostasis. PPARγ is a master regulator of adipose tissue signaling that plays an essential role in insulin sensitivity, making it an important therapeutic target. The selective PPARγ agonist rosiglitazone (RSG) has been used to treat diabetes. However, adverse cardiovascular effects have seriously hindered its clinical application. Experimental models have revealed that PPARγ activation increases cardiac hypertrophy. RSG stimulates cardiac hypertrophy and oxidative stress in cardiomyocyte-specific PPARγ knockout mice, implying that RSG might stimulate cardiac hypertrophy independently of cardiomyocyte PPARγ. However, candidate cell types responsible for RSG-induced cardiomyocyte hypertrophy remain unexplored. Utilizing cocultures of adipocytes and cardiomyocytes, we found that stimulation of PPARγ signaling in adipocytes increased miR-200a expression and secretion. Delivery of miR-200a in adipocyte-derived exosomes to cardiomyocytes resulted in decreased TSC1 and subsequent mTOR activation, leading to cardiomyocyte hypertrophy. Treatment with an antagomir to miR-200a blunted this hypertrophic response in cardiomyocytes. In vivo, specific ablation of PPARγ in adipocytes was sufficient to blunt hypertrophy induced by RSG treatment. By delineating mechanisms by which RSG elicits cardiac hypertrophy, we have identified pathways that mediate the crosstalk between adipocytes and cardiomyocytes to regulate cardiac remodeling.
Collapse
Affiliation(s)
- Xi Fang
- Institute of Cardiovascular Science, Peking University Health Science Center, Beijing, China.,Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Matthew J Stroud
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Kunfu Ouyang
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Li Fang
- Institute of Cardiovascular Science, Peking University Health Science Center, Beijing, China
| | - Jianlin Zhang
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Nancy D Dalton
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Yusu Gu
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Tongbin Wu
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Kirk L Peterson
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Hsien-Da Huang
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan
| | - Ju Chen
- Department of Medicine, School of Medicine, UCSD, La Jolla, California, USA
| | - Nanping Wang
- Institute of Cardiovascular Science, Peking University Health Science Center, Beijing, China.,The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
29
|
Stability and function of adult vasculature is sustained by Akt/Jagged1 signalling axis in endothelium. Nat Commun 2016; 7:10960. [PMID: 26971877 PMCID: PMC4793084 DOI: 10.1038/ncomms10960] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/04/2016] [Indexed: 12/12/2022] Open
Abstract
The signalling pathways operational in quiescent, post-development vasculature remain enigmatic. Here we show that unlike neovascularization, endothelial Akt signalling in established vasculature is crucial not for endothelial cell (EC) survival, but for sustained interactions with pericytes and vascular smooth muscle cells (VSMCs) regulating vascular stability and function. Inducible endothelial-specific Akt1 deletion in adult global Akt2KO mice triggers progressive VSMC apoptosis. In hearts, this causes a loss of arteries and arterioles and, despite a high capillary density, diminished vascular patency and severe cardiac dysfunction. Similarly, endothelial Akt deletion induces retinal VSMC loss and basement membrane deterioration resulting in vascular regression and retinal atrophy. Mechanistically, the Akt/mTOR axis controls endothelial Jagged1 expression and, thereby, Notch signalling regulating VSMC maintenance. Jagged1 peptide treatment of Akt1ΔEC;Akt2KO mice and Jagged1 re-expression in Akt-deficient endothelium restores VSMC coverage. Thus, sustained endothelial Akt1/2 signalling is critical in maintaining vascular stability and homeostasis, thereby preserving tissue and organ function. The Akt pathway integrates multiple signals, but whether it affects vasculature function is debatable. Here the authors show that Akt pathway shutdown in adult mouse endothelium causes destabilization of vasculature leading to cardiac and retinal dysfunction, due to decreased levels of Jagged1 and impaired Notch signaling.
Collapse
|
30
|
Sundararaj K, Pleasant DL, Moschella PC, Panneerselvam K, Balasubramanian S, Kuppuswamy D. mTOR Complexes Repress Hypertrophic Agonist-Stimulated Expression of Connective Tissue Growth Factor in Adult Cardiac Muscle Cells. J Cardiovasc Pharmacol 2016; 67:110-20. [PMID: 26371948 PMCID: PMC7334753 DOI: 10.1097/fjc.0000000000000322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Connective tissue growth factor (CTGF) is a fibrogenic cytokine that promotes fibrosis in various organs. In the heart, both cardiomyocytes (CM) and cardiac fibroblasts have been reported as a source of CTGF expression, aiding cardiac fibrosis. Although the mammalian target of rapamycin (mTOR) forms 2 distinct complexes, mTORC1 and mTORC2, and plays a central role in integrating biochemical signals for protein synthesis and cellular homeostasis, we explored its role in CTGF expression in adult feline CM. CM were stimulated with 10 μM phenylephrine (PE), 200 nM angiotensin (Ang), or 100 nM insulin for 24 hours. PE and Ang, but not insulin, caused an increase in CTGF mRNA expression with the highest expression observed with PE. Inhibition of mTOR with torin1 but not rapamycin significantly enhanced PE-stimulated CTGF expression. Furthermore, silencing of raptor and rictor using shRNA adenoviral vectors to suppress mTORC1 and mTORC2, respectively, or blocking phosphatidylinositol 3-kinase (PI3K) signaling with LY294002 (LY) or Akt signaling by dominant-negative Akt expression caused a substantial increase in PE-stimulated CTGF expression as measured by both mRNA and secreted protein levels. However, studies with dominant-negative delta isoform of protein kinase C demonstrate that delta isoform of protein kinase C is required for both agonist-induced CTGF expression and mTORC2/Akt-mediated CTGF suppression. Finally, PE-stimulated CTGF expression was accompanied with a corresponding increase in Smad3 phosphorylation and pretreatment of cells with SIS3, a Smad3 specific inhibitor, partially blocked the PE-stimulated CTGF expression. Therefore, a PI3K/mTOR/Akt axis plays a suppressive role on agonist-stimulated CTGF expression where the loss of this mechanism could be a contributing factor for the onset of cardiac fibrosis in the hypertrophying myocardium.
Collapse
Affiliation(s)
- Kamala Sundararaj
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Dorea L. Pleasant
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Phillip C. Moschella
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Kavin Panneerselvam
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Sundaravadivel Balasubramanian
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Dhandapani Kuppuswamy
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
- Corresponding author at: Gazes Cardiac Research Institute, Medical University of South Carolina, 114 Doughty Street, Charleston, South Carolina 29425-2221 Telephone: 843-876 5067; Fax: 843-876 5068;
| |
Collapse
|
31
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
32
|
Saraf R, Huang T, Mahmood F, Owais K, Bardia A, Khabbaz KR, Liu D, Senthilnathan V, Lassaletta AD, Sellke F, Matyal R. Early Cellular Changes in the Ascending Aorta and Myocardium in a Swine Model of Metabolic Syndrome. PLoS One 2016; 11:e0146481. [PMID: 26766185 PMCID: PMC4713205 DOI: 10.1371/journal.pone.0146481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 12/17/2015] [Indexed: 01/10/2023] Open
Abstract
Background Metabolic syndrome is associated with pathological remodeling of the heart and adjacent vessels. The early biochemical and cellular changes underlying the vascular damage are not fully understood. In this study, we sought to establish the nature, extent, and initial timeline of cytochemical derangements underlying reduced ventriculo-arterial compliance in a swine model of metabolic syndrome. Methods Yorkshire swine (n = 8 per group) were fed a normal diet (ND) or a high-cholesterol (HCD) for 12 weeks. Myocardial function and blood flow was assessed before harvesting the heart. Immuno-blotting and immuno-histochemical staining were used to assess the cellular changes in the myocardium, ascending aorta and left anterior descending artery (LAD). Results There was significant increase in body mass index, blood glucose and mean arterial pressures (p = 0.002, p = 0.001 and p = 0.024 respectively) in HCD group. At the cellular level there was significant increase in anti-apoptotic factors p-Akt (p = 0.007 and p = 0.002) and Bcl-xL (p = 0.05 and p = 0.01) in the HCD aorta and myocardium, respectively. Pro-fibrotic markers TGF-β (p = 0.01), pSmad1/5 (p = 0.03) and MMP-9 (p = 0.005) were significantly increased in the HCD aorta. The levels of pro-apoptotic p38MAPK, Apaf-1 and cleaved Caspase3 were significantly increased in aorta of HCD (p = 0.03, p = 0.04 and p = 0.007 respectively). Similar changes in coronary arteries were not observed in either group. Functionally, the high cholesterol diet resulted in significant increase in ventricular end systolic pressure and–dp/dt (p = 0.05 and p = 0.007 respectively) in the HCD group. Conclusion Preclinical metabolic syndrome initiates pro-apoptosis and pro-fibrosis pathways in the heart and ascending aorta, while sparing coronary arteries at this early stage of dietary modification.
Collapse
Affiliation(s)
- Rabya Saraf
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thomas Huang
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Khurram Owais
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amit Bardia
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kamal R. Khabbaz
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Liu
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Venkatachalam Senthilnathan
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Antonio D. Lassaletta
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Surgery, Rhode Island Hospital, Brown Alpert School of Medicine, Providence, Rhode Island, United States of America
| | - Frank Sellke
- Department of Surgery, Rhode Island Hospital, Brown Alpert School of Medicine, Providence, Rhode Island, United States of America
| | - Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
33
|
Mohamed BA, Schnelle M, Khadjeh S, Lbik D, Herwig M, Linke WA, Hasenfuss G, Toischer K. Molecular and structural transition mechanisms in long-term volume overload. Eur J Heart Fail 2015; 18:362-71. [PMID: 26694078 PMCID: PMC5064674 DOI: 10.1002/ejhf.465] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/08/2015] [Accepted: 11/14/2015] [Indexed: 01/09/2023] Open
Abstract
Aim We have previously reported that early phase (1 week) of experimental volume overload (VO) has an adaptive phenotype while wall stress‐matched pressure overload (PO) is maladaptive. Here we investigate the transition from adaptation to heart failure (HF) in long‐term VO. Methods and results FVB/N wild‐type mice were subjected to VO induced by aortocaval shunt, and were followed by serial echocardiography until in vivo left ventricular ejection fraction was below <50% (135 ± 35 days). Heart failure was evident from increased lung and liver weight and increased mortality compared with sham. Maladaptive remodelling resulted in significantly reduced sarcomeric titin phosphorylation (causing increased sarcomeric stiffness), whereas interstitial fibrosis was not increased. This was paralleled by re‐expression of the fetal gene program, activation of calcium/calmodulin‐dependent protein kinase II (CaMKII), decreased protein kinase B (Akt) phosphorylation, high oxidative stress, and increased apoptosis. Consistently, development of HF and mortality were significantly aggravated in Akt‐deficient mice. Conclusion Transition to HF in VO is associated with decreased Akt and increased CaMKII signalling pathways together with increased oxidative stress and apoptosis. Lack of interstitial fibrosis together with sarcomeric titin hypophosphorylation indicates an increased stiffness at the sarcomeric but not matrix level in VO‐induced HF (in contrast to PO). Transition to HF may result from myocyte loss and myocyte dysfunction owing to increased stiffness.
Collapse
Affiliation(s)
- Belal A Mohamed
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany.,Faculty of Medicine, Mansoura University, Egypt
| | - Moritz Schnelle
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany
| | - Sara Khadjeh
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Dawid Lbik
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Melissa Herwig
- Department of Cardiovascular Physiology, Ruhr University Bochum, Germany
| | - Wolfgang A Linke
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany.,Department of Cardiovascular Physiology, Ruhr University Bochum, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| |
Collapse
|
34
|
Gong X, Wang P, Wu Q, Wang S, Yu L, Wang G. Human umbilical cord blood derived mesenchymal stem cells improve cardiac function in cTnT(R141W) transgenic mouse of dilated cardiomyopathy. Eur J Cell Biol 2015; 95:57-67. [PMID: 26655348 DOI: 10.1016/j.ejcb.2015.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/15/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Cell transplantation is a promising strategy in regenerative medicine. Beneficial effects of bone marrow mesenchymal stem cells (BM-MSCs) on heart disease have been widely reported. However, the MSCs in these studies have been mainly derived from autologous animals, and data on MSCs from human umbilical cord blood (UCB-MSCs) are still scarce. We investigated whether intramyocardial xenogeneic administration of UCB-MSCs is beneficial for preserving heart function in a cTnT(R141W) transgenic mouse of dilated cardiomyopathy (DCM). Cultured UCB-MSCs, which were identified by there morphology, differentiation and cell surface markers, were transplanted into cTnT(R141W) transgenic mice to examine apoptosis, fibrosis, vasculogenesis and the associated Akt pathway. Moreover, we measured the expression levels of VEGF and IGF-1, which are growth factors required for differentiation into cardiomyocytes, and are also involved in cardiac regeneration and improving heart function. One month after transplantation, MSCs significantly decreased chamber dilation and contractile dysfunction in the cTnT(R141W) mice. MSCs transplanted hearts showed a significant decrease in cardiac apoptosis and its regulation by the Akt pathway. Cardiac fibrosis and cytoplasmic vacuolisation were significantly attenuated in the MSCs group. Importantly, the levels of VEGF and IGF-1 were increased in the MSCs transplanted hearts. In vitro, the MSC-conditioned medium displayed anti-apoptotic activity in h9c2 cardiomyocytes subjected to hypoxia. These results further confirm the paracrine effects of MSCs. In conclusion, UCB-MSCs preserve cardiac function after intramyocardial transplantation in a DCM mouse, and this effect may be associated with reductions in cellular apoptosis, inflammation, hypertrophy and myocardial fibrosis; in addition to; up-regulation of Akt, VEGF and IGF-1; and enhanced angiogenesis.
Collapse
Affiliation(s)
- Xuhe Gong
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Pengbo Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qingqing Wu
- Departments of Obstetrics and Gynaecology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Sijia Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Litian Yu
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Guogan Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Departments of Obstetrics and Gynaecology, Fuxing Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
35
|
ZÁLEŠÁK M, BLAŽÍČEK P, GABLOVSKÝ I, LEDVÉNYIOVÁ V, BARTEKOVÁ M, ZIEGELHÖFFER A, RAVINGEROVÁ T. Impaired PI3K/Akt Signaling as a Potential Cause of Failure to Precondition Rat Hearts Under Conditions of Simulated Hyperglycemia. Physiol Res 2015; 64:633-41. [DOI: 10.33549/physiolres.932883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The aim of the study was to evaluate the impact of simulated acute hyperglycemia (HG) on PI3K/Akt signaling in preconditioned and non-preconditioned isolated rat hearts perfused with Krebs-Henseleit solution containing normal (11 mmol/l) or elevated (22 mmol/l) glucose subjected to ischemia-reperfusion. Ischemic preconditioning (IP) was induced by two 5-min cycles of coronary occlusion followed by 5-min reperfusion. Protein levels of Akt, phosphorylated (activated) Akt (P-Akt), as well as contents of BAX protein were assayed (Western blotting) in cytosolic fraction of myocardial tissue samples taken prior to and after 30-min global ischemia and 40-min reperfusion. In “normoglycemic” conditions (NG), IP significantly increased P-Akt at the end of long-term ischemia, while reperfusion led to its decrease together with the decline of BAX levels as compared to non-preconditioned hearts. On the contrary, under HG conditions, P-Akt tended to decline in IP-hearts after long-term ischemia, and it was significantly higher after reperfusion than in non-preconditioned controls. No significant influence of IP on BAX levels at the end of I/R was observed under HG conditions. It seems that high glucose may influence IP-induced activation of Akt and its downstream targets, as well as maintain persistent Akt activity that may be detrimental for the heart under above conditions.
Collapse
Affiliation(s)
- M. ZÁLEŠÁK
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG Bratislava, Slovak Republic
| | | | | | | | | | | | | |
Collapse
|
36
|
Xie C, Zhang Y, Tran TDN, Wang H, Li S, George EV, Zhuang H, Zhang P, Kandel A, Lai Y, Tang D, Reeves WH, Cheng H, Ding Y, Yang LJ. Irisin Controls Growth, Intracellular Ca2+ Signals, and Mitochondrial Thermogenesis in Cardiomyoblasts. PLoS One 2015; 10:e0136816. [PMID: 26305684 PMCID: PMC4549318 DOI: 10.1371/journal.pone.0136816] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/07/2015] [Indexed: 12/31/2022] Open
Abstract
Exercise offers short-term and long-term health benefits, including an increased metabolic rate and energy expenditure in myocardium. The newly-discovered exercise-induced myokine, irisin, stimulates conversion of white into brown adipocytes as well as increased mitochondrial biogenesis and energy expenditure. Remarkably, irisin is highly expressed in myocardium, but its physiological effects in the heart are unknown. The objective of this work is to investigate irisin’s potential multifaceted effects on cardiomyoblasts and myocardium. For this purpose, H9C2 cells were treated with recombinant irisin produced in yeast cells (r-irisin) and in HEK293 cells (hr-irisin) for examining its effects on cell proliferation by MTT [3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and on gene transcription profiles by qRT-PCR. R-irisin and hr-irisin both inhibited cell proliferation and activated genes related to cardiomyocyte metabolic function and differentiation, including myocardin, follistatin, smooth muscle actin, and nuclear respiratory factor-1. Signal transduction pathways affected by r-irisin in H9C2 cells and C57BL/6 mice were examined by detecting phosphorylation of PI3K-AKT, p38, ERK or STAT3. We also measured intracellular Ca2+ signaling and mitochondrial thermogenesis and energy expenditure in r-irisin-treated H9C2 cells. The results showed that r-irisin, in a certain concentration rage, could activate PI3K-AKT and intracellular Ca2+ signaling and increase cellular oxygen consumption in H9C2 cells. Our study also suggests the existence of irisin-specific receptor on the membrane of H9C2 cells. In conclusion, irisin in a certain concentration rage increased myocardial cell metabolism, inhibited cell proliferation and promoted cell differentiation. These effects might be mediated through PI3K-AKT and Ca2+ signaling, which are known to activate expression of exercise-related genes such as follistatin and myocardin. This work supports the value of exercise, which promotes irisin release.
Collapse
Affiliation(s)
- Chao Xie
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Yuan Zhang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P. R. China
| | - Tran D. N. Tran
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, United States of America
| | - Hai Wang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Shiwu Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Eva Vertes George
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Haoyang Zhuang
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Peilan Zhang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States of America
| | - Avi Kandel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Yimu Lai
- Department of Cell Biology and Anatomy, University of South Carolina of Medicine, Columbia, SC, 29209, United States of America
| | - Dongqi Tang
- Center for Stem Cell & Regenerative Medicine, The Second Hospital of Shandong University, Jinan, 250012, P. R. China
| | - Westley H. Reeves
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
| | - Henrique Cheng
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, United States of America
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States of America
- * E-mail: (L-JY); (YD)
| | - Li-Jun Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, United States of America
- * E-mail: (L-JY); (YD)
| |
Collapse
|
37
|
Holzem KM, Marmerstein JT, Madden EJ, Efimov IR. Diet-induced obesity promotes altered remodeling and exacerbated cardiac hypertrophy following pressure overload. Physiol Rep 2015; 3:3/8/e12489. [PMID: 26290533 PMCID: PMC4562575 DOI: 10.14814/phy2.12489] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Heart failure (HF) is the end stage of cardiovascular disease, in which hypertrophic remodeling no longer meets cardiac output demand. Established animal models of HF have provided insights into disease pathogenesis. However, these models are developed on dissimilar metabolic backgrounds from humans – patients with HF are frequently overweight or obese, whereas animal models of HF are typically lean. Thus, we aimed to develop and investigate model for cardiac hypertrophy and failure that also recapitulates the cardiometabolic state of HF in humans. We subjected mice with established diet-induced obesity (DIO) to cardiac pressure overload provoked by transverse aortic constriction (TAC). Briefly, we fed WT male mice a normal chow or high-fat diet for 10 weeks prior to sham/TAC procedures and until surgical follow-up. We then analyzed cardiac hypertrophy, mechanical function, and electrophysiology at 5–6 weeks after surgery. In DIO mice with TAC, hypertrophy and systolic dysfunction were exacerbated relative to chow TAC animals, which showed minimal remodeling with our moderate constriction intensity. Normalized heart weight was 55.8% greater and fractional shortening was 30.9% less in DIO TAC compared with chow TAC hearts. However, electrophysiologic properties were surprisingly similar between DIO sham and TAC animals. To examine molecular pathways activated by DIO and TAC, we screened prohypertrophic signaling cascades, and the exacerbated remodeling was associated with early activation of the c-Jun-N-terminal kinase (JNK1/2) signaling pathway. Thus, DIO aggravates the progression of hypertrophy and HF caused by pressure overload, which is associated with JNK1/2 signaling, and cardiometabolic state can significantly modify HF pathogenesis.
Collapse
Affiliation(s)
- Katherine M Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Joseph T Marmerstein
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eli J Madden
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
38
|
Lin CY, Hsu YJ, Hsu SC, Chen Y, Lee HS, Lin SH, Huang SM, Tsai CS, Shih CC. CB1 cannabinoid receptor antagonist attenuates left ventricular hypertrophy and Akt-mediated cardiac fibrosis in experimental uremia. J Mol Cell Cardiol 2015; 85:249-61. [PMID: 26093151 DOI: 10.1016/j.yjmcc.2015.06.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 06/12/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023]
Abstract
Cannabinoid receptor type 1 (CB1R) plays an important role in the development of myocardial hypertrophy and fibrosis-2 pathological features of uremic cardiomyopathy. However, it remains unknown whether CB1R is involved in the pathogenesis of uremic cardiomyopathy. Here, we aimed to elucidate the role of CB1R in the development of uremic cardiomyopathy via modulation of Akt signalling. The heart size and myocardial fibrosis were evaluated by echocardiography and immunohistochemical staining, respectively, in 5/6 nephrectomy chronic kidney disease (CKD) mice treated with a CB1R antagonist. CB1R and fibrosis marker expression levels were determined by immunoblotting in H9c2 cells exposed to the uremic toxin indoxyl sulfate (IS), with an organic anion transporter 1 inhibitor or a CB1R antagonist or agonist. Akt phosphorylation was also assessed to examine the signaling pathways downstream of CB1R activation induced by IS in H9c2 cells. CKD mice exhibited marked left ventricular hypertrophy and myocardial fibrosis, which were reversed by treatment with the CB1R antagonist. CB1R, collagen I, transforming growth factor (TGF)-β, and α-smooth muscle actin (SMA) expression showed time- and dose-dependent upregulation in H9c2 cells treated with IS. The inhibition of CB1R by either CB1R antagonist or small interfering RNA-mediated knockdown attenuated the expression of collagen I, TGF-β, and α-SMA in IS-treated H9c2 cells, while Akt phosphorylation was enhanced by CB1R agonist and abrogated by CB1R antagonist in these cells. In summary, we conclude that CB1R blockade attenuates LVH and Akt-mediated cardiac fibrosis in a CKD mouse model. Uremic toxin IS stimulates the expression of CB1R and fibrotic markers and CB1R inhibition exerts anti-fibrotic effects via modulation of Akt signaling in H9c2 myofibroblasts. Therefore, the development of drugs targeting CB1R may have therapeutic potential in the treatment of uremic cardiomyopathy.
Collapse
Affiliation(s)
- Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Che Hsu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ying Chen
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Che Shih
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
39
|
de Almeida PWM, Melo MB, Lima RDF, Gavioli M, Santiago NM, Greco L, Jesus ICG, Nocchi E, Parreira A, Alves MNM, Mitraud L, Resende RR, Campagnole-Santos MJ, Dos Santos RAS, Guatimosim S. Beneficial effects of angiotensin-(1-7) against deoxycorticosterone acetate-induced diastolic dysfunction occur independently of changes in blood pressure. Hypertension 2015; 66:389-95. [PMID: 26077567 DOI: 10.1161/hypertensionaha.114.04893] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 04/13/2015] [Indexed: 01/26/2023]
Abstract
Mineralocorticoids have been implicated in the pathogenesis of diastolic heart failure. On the contrary, angiotensin (Ang)-(1-7) has emerged as a potential strategy for treatment of cardiac dysfunction induced by excessive mineralocorticoid receptor activation. A critical question about the cardioprotective effect of Ang-(1-7) in hypertensive models is its dependence on blood pressure (BP) reduction. Here, we addressed this question by investigating the mechanisms involved in Ang-(1-7) cardioprotection against mineralocorticoid receptor activation. Sprague-Dawley (SD) and transgenic (TG) rats that overexpress an Ang-(1-7) producing fusion protein (TG(A1-7)3292) were treated with deoxycorticosterone acetate (DOCA) for 6 weeks. After treatment, SD rats became hypertensive and developed ventricular hypertrophy. These parameters were attenuated in TG-DOCA. SD-DOCA rats developed diastolic dysfunction which was associated at the cellular level with reduced Ca(2+) transient. Oppositely, TG-DOCA myocytes presented enhanced Ca(2+) transient. Moreover, higher extracellular signal-regulated kinase phosphorylation, type 1 phosphatase, and protein kinase Cα levels were found in SD-DOCA cells. In vivo, pressor effects of DOCA can contribute to the diastolic dysfunction, raising the question of whether protection in TG was a consequence of reduced BP. To address this issue, BP in SD-DOCA was kept at TG-DOCA level by giving hydralazine or by reducing the DOCA amount given to rats (Low-DOCA). Under similar BP, diastolic dysfunction and molecular changes were still evident in DOCA-hydralazine and SD-low-DOCA, but not in TG-DOCA. In conclusion, Ang-(1-7) protective signaling against DOCA-induced diastolic dysfunction occurs independently of BP attenuation and is mediated by the activation of pathways involved in Ca(2+) handling, hypertrophy, and survival.
Collapse
Affiliation(s)
- Pedro W Machado de Almeida
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcos Barrouin Melo
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo de Freitas Lima
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana Gavioli
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nivia M Santiago
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Greco
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar C G Jesus
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Nocchi
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Amanda Parreira
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcia N M Alves
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana Mitraud
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Ribeiro Resende
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria José Campagnole-Santos
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Robson Augusto Souza Dos Santos
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- From the Department of Physiology and Biophysics, Institute of Biological Sciences (P.W.M.d.A., M.B.M., R.d.F.L., M.G., N.M.S., L.G., I.C.G.J., E.N., A.P., M.N.M.A., L.M., M.J.C.-S., R.A.S.d.S., S.G.), Department of Biochemistry and Immunology, Institute of Biological Sciences (R.R.R.), and National Institute of Science and Technology in Nanobiopharmaceutics (M.B.M., M.J.C.-S., R.A.S.d.S., S.G.), Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
40
|
Luo W, Zhao X, Jin H, Tao L, Zhu J, Wang H, Hemmings BA, Yang Z. Akt1 signaling coordinates BMP signaling and β-catenin activity to regulate second heart field progenitor development. Development 2015; 142:732-42. [DOI: 10.1242/dev.119016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Second heart field (SHF) progenitors exhibit continued proliferation and delayed differentiation, which are modulated by FGF4/8/10, BMP and canonical Wnt/β-catenin signaling. PTEN-Akt signaling regulates the stem cell/progenitor cell homeostasis in several systems, such as hematopoietic stem cells, intestinal stem cells and neural progenitor cells. To address whether PTEN-Akt signaling is involved in regulating cardiac progenitors, we deleted Pten in SHF progenitors. Deletion of Pten caused SHF expansion and increased the size of the SHF derivatives, the right ventricle and the outflow tract. Cell proliferation of cardiac progenitors was enhanced, whereas cardiac differentiation was unaffected by Pten deletion. Removal of Akt1 rescued the phenotype and early lethality of Pten deletion mice, suggesting that Akt1 was the key downstream target that was negatively regulated by PTEN in cardiac progenitors. Furthermore, we found that inhibition of FOXO by Akt1 suppressed the expression of the gene encoding the BMP ligand (BMP7), leading to dampened BMP signaling in the hearts of Pten deletion mice. Cardiac activation of Akt also increased the Ser552 phosphorylation of β-catenin, thus enhancing its activity. Reducing β-catenin levels could partially rescue heart defects of Pten deletion mice. We conclude that Akt signaling regulates the cell proliferation of SHF progenitors through coordination of BMP signaling and β-catenin activity.
Collapse
Affiliation(s)
- Wen Luo
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Xia Zhao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Hengwei Jin
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Lichan Tao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Jingai Zhu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Huijuan Wang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Brian A. Hemmings
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
- Collaborative Innovation Center for Genetics and Development, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
41
|
Astragalus polysaccharide suppresses doxorubicin-induced cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:674219. [PMID: 25386226 PMCID: PMC4216718 DOI: 10.1155/2014/674219] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/11/2014] [Indexed: 01/02/2023]
Abstract
Background. Doxorubicin, a potent chemotherapeutic agent, is associated with acute and chronic cardiotoxicity, which is cumulatively dose-dependent. Astragalus polysaccharide (APS), the extract of Astragalus membranaceus with strong antitumor and antiglomerulonephritis activity, can effectively alleviate inflammation. However, whether APS could ameliorate chemotherapy-induced cardiotoxicity is not understood. Here, we investigated the protective effects of APS on doxorubicin-induced cardiotoxicity and elucidated the underlying mechanisms of the protective effects of APS. Methods. We analyzed myocardial injury in cancer patients who underwent doxorubicin chemotherapy and generated a doxorubicin-induced neonatal rat cardiomyocyte injury model and a mouse heart failure model. Echocardiography, reactive oxygen species (ROS) production, TUNEL, DNA laddering, and Western blotting were performed to observe cell survival, oxidative stress, and inflammatory signal pathways in cardiomyocytes. Results. Treatment of patients with the chemotherapeutic drug doxorubicin led to heart dysfunction. Doxorubicin reduced cardiomyocyte viability and induced C57BL/6J mouse heart failure with concurrent elevated ROS generation and apoptosis, which, however, was attenuated by APS treatment. In addition, there was profound inhibition of p38MAPK and activation of Akt after APS treatment. Conclusions. These results demonstrate that APS could suppress oxidative stress and apoptosis, ameliorating doxorubicin-mediated cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways.
Collapse
|
42
|
Zhou Y, Wang D, Gao X, Lew K, Richards AM, Wang P. mTORC2 phosphorylation of Akt1: a possible mechanism for hydrogen sulfide-induced cardioprotection. PLoS One 2014; 9:e99665. [PMID: 24949720 PMCID: PMC4064967 DOI: 10.1371/journal.pone.0099665] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 05/16/2014] [Indexed: 01/05/2023] Open
Abstract
Hydrogen sulfide (H2S) is known to have cardiac protective effects through Akt activation. Akt acts as a 'central sensor' for myocyte survival or death; its activity is regulated by multiple kinases including PI3K, mTORC2, PDK1 and phosphatases including PTEN, PP2A and PHLPPL. Based on the previous finding that PI3K inhibitor LY294002 abolishes H2S-induced Akt phosphorylation and cardioprotection, it is accepted that PI3K is the mediator of H2S-induced Akt phosphorylation. However, LY294002 inhibits both PI3K and mTOR, and PI3K only recruits Akt to the membrane where Akt is phosphorylated by Akt kinases. We undertook a series of experiments to further evaluate the role of mTORC2, PDK1, PTEN, PP2A and PHLPPL in H2S-induced Akt phosphorylation and cardioprotection, which, we believe, has not been investigated before. Hearts from adult Sprague-Dawley rats were isolated and subjected to (i) normoxia, (ii) global ischemia and (iii) ischemia/reperfusion in the presence or absence of 50 µM of H2S donor NaHS. Cardiac mechanical function and lactate dehydrogenase (LDH) release were assessed. All hearts also were Western analyzed at the end of perfusion for Akt and a panel of appropriate Akt regulators and targets. Hearts pretreated with 50 µM NaHS had improved function at the end of reperfusion (Rate pressure product; 19±4×10(3) vs. 10±3×10(3) mmHg/min, p<0.05) and reduced cell injury (LDH release 19±10 vs. 170±87 mU/ml p<0.05) compared to untreated hearts. NaHS significantly increased phospho-Akt, phospho-mTOR, phospho-Bim and Bcl-2 in reperfused hearts (P<0.05). Furthermore using H9c2 cells we demonstrate that NaHS pretreatment reduces apoptosis following hypoxia/re-oxygenation. Importantly, PP242, a specific mTOR inhibitor, abolished both cardioprotection and protein phosphorylation in isolated heart and reduced apoptotic effects in H9c2 cells. Treating hearts with NaHS only during reperfusion produced less cardioprotection through a similar mechanism. These data suggest mTORC2 phosphorylation of Akt is a key mediator of H2S-induced cardioprotection in I/R.
Collapse
Affiliation(s)
- Yue Zhou
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Daying Wang
- Department of Cardiology, Putuo Hospital, Shanghai, China
| | - Xiufang Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Karsheng Lew
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Peipei Wang
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
43
|
Johnson AM, Kartha CC. Proliferation of murine c-kit(pos) cardiac stem cells stimulated with IGF-1 is associated with Akt-1 mediated phosphorylation and nuclear export of FoxO3a and its effect on downstream cell cycle regulators. Growth Factors 2014; 32:53-62. [PMID: 24580070 DOI: 10.3109/08977194.2014.889694] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) is known to promote proliferation in many cell types including c-kit(pos) cardiac stem cells (CSCs). Downstream signaling pathways of IGF-1 induced CSC proliferation have not been investigated. An important downstream target of IGF-1/Akt-1 signaling is FoxO3a, a key negative regulator of cell-cycle progression. We studied the effect of IGF-1 on proliferation of c-kit(pos) murine CSCs and found that IGF-1-mediated cell proliferation is associated with FoxO3a phosphorylation and inactivation of its transcriptional activity. PI3 inhibitors LY294002 and Wortmannin abolished the effect of IGF-1 on FoxO3a phosphorylation indicating that FoxO3a phosphorylation is mediated by PI3/Akt-1 pathway. In cells with FoxO3a translocation to the cytoplasm, there is decreased expression of cell-cycle inhibitors such as p27(kip1) and p57(kip2) and increased expression of CyclinD1. Our study provides evidence that IGF-1 induced CSC proliferation could be the result of FoxO3a inactivation and its downstream effect on cell-cycle regulators.
Collapse
Affiliation(s)
- Ann Mary Johnson
- Cardiovascular Disease Biology Division, Rajiv Gandhi Centre for Biotechnology , Trivandrum, Kerala , India
| | | |
Collapse
|
44
|
Ulm S, Liu W, Zi M, Tsui H, Chowdhury SK, Endo S, Satoh Y, Prehar S, Wang R, Cartwright EJ, Wang X. Targeted deletion of ERK2 in cardiomyocytes attenuates hypertrophic response but provokes pathological stress induced cardiac dysfunction. J Mol Cell Cardiol 2014; 72:104-16. [PMID: 24631771 PMCID: PMC4046245 DOI: 10.1016/j.yjmcc.2014.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/28/2014] [Accepted: 03/03/2014] [Indexed: 11/16/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) are involved in the regulation of cardiac hypertrophy and myocyte survival. Extracellular signal regulated protein kinase 1 and 2 (ERK1/2) are key components in the MAPK signaling pathways. Dysfunction of ERK1/2 in congenital heart diseases (Noonan syndrome and LEOPARD syndrome) leads to cardiac hypertrophy. ERK2 contributes 70% of protein content to total ERK1/2 content in myocardium; however, the specific role of ERK2 in regulating cardiac hypertrophy is yet to be further defined. To investigate the specific role of ERK2 played in the cardiomyocytes, we generated and examined mice with cardiomyocyte-specific deletion of the erk2 gene (ERK2cko mice). Following short-term pathological hypertrophic stresses, the mutant mice showed attenuated hypertrophic remodeling characterized by a blunted increase in the cross-sectional area of individual myocytes, downregulation of hypertrophic foetal gene markers (ANP and BNP), and less interstitial fibrosis. However, increased cardiomyocyte apoptosis was observed. Upon prolonged stimulation, ERK2cko mice developed deterioration in cardiac function. However, absence of ERK2 did not affect physiological hypertrophy induced by 4 weeks of swimming exercise. These results revealed an essential role for ERK2 in cardiomyocytes in the development of pathological hypertrophic remodeling and resistance to cell death.
ERK2 is required for pathological cardiac hypertrophy. ERK2 is essential for cardiomyocyte survival through preventing apoptosis. ERK2 deficiency in cardiomyocytes accelerates heart failure progress. ERK2 is unlikely to be involved in the regulation of physiological hypertrophy.
Collapse
Affiliation(s)
- Susanne Ulm
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Wei Liu
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK.
| | - Min Zi
- Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Hoyee Tsui
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Sanjoy K Chowdhury
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Yasushi Satoh
- Department of Anesthesiology, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan
| | - Sukhpal Prehar
- Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Ruoxi Wang
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Elizabeth J Cartwright
- Faculty of Medical and Human Sciences, The University of Manchester, Manchester M13 9NT, UK
| | - Xin Wang
- Faculty of Life Sciences, The University of Manchester, Manchester M13 9NT, UK
| |
Collapse
|
45
|
Saks V, Schlattner U, Tokarska-Schlattner M, Wallimann T, Bagur R, Zorman S, Pelosse M, Santos PD, Boucher F, Kaambre T, Guzun R. Systems Level Regulation of Cardiac Energy Fluxes Via Metabolic Cycles: Role of Creatine, Phosphotransfer Pathways, and AMPK Signaling. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Mori J, Zhang L, Oudit GY, Lopaschuk GD. Impact of the renin–angiotensin system on cardiac energy metabolism in heart failure. J Mol Cell Cardiol 2013; 63:98-106. [DOI: 10.1016/j.yjmcc.2013.07.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/12/2013] [Accepted: 07/14/2013] [Indexed: 01/12/2023]
|
47
|
Moschella PC, McKillop J, Pleasant DL, Harston RK, Balasubramanian S, Kuppuswamy D. mTOR complex 2 mediates Akt phosphorylation that requires PKCε in adult cardiac muscle cells. Cell Signal 2013; 25:1904-12. [PMID: 23673367 PMCID: PMC3704180 DOI: 10.1016/j.cellsig.2013.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 05/06/2013] [Indexed: 01/28/2023]
Abstract
Our earlier work showed that mammalian target of rapamycin (mTOR) is essential to the development of various hypertrophic responses, including cardiomyocyte survival. mTOR forms two independent complexes, mTORC1 and mTORC2, by associating with common and distinct cellular proteins. Both complexes are sensitive to a pharmacological inhibitor, torin1, although only mTORC1 is inhibited by rapamycin. Since mTORC2 is known to mediate the activation of a prosurvival kinase, Akt, we analyzed whether mTORC2 directly mediates Akt activation or whether it requires the participation of another prosurvival kinase, PKCε (epsilon isoform of protein kinase-C). Our studies reveal that treatment of adult feline cardiomyocytes in vitro with insulin results in Akt phosphorylation at S473 for its activation which could be augmented with rapamycin but blocked by torin1. Silencing the expression of Rictor (rapamycin-insensitive companion of mTOR), an mTORC2 component, with a sh-RNA in cardiomyocytes lowers both insulin-stimulated Akt and PKCε phosphorylation. Furthermore, phosphorylation of PKCε and Akt at the critical S729 and S473 sites respectively was blocked by torin1 or Rictor knockdown but not by rapamycin, indicating that the phosphorylation at these specific sites occurs downstream of mTORC2. Additionally, expression of DN-PKCε significantly lowered the insulin-stimulated Akt S473 phosphorylation, indicating an upstream role for PKCε in the Akt activation. Biochemical analyses also revealed that PKCε was part of Rictor but not Raptor (a binding partner and component of mTORC1). Together, these studies demonstrate that mTORC2 mediates prosurvival signaling in adult cardiomyocytes where PKCε functions downstream of mTORC2 leading to Akt activation.
Collapse
Affiliation(s)
- Phillip C. Moschella
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - John McKillop
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Dorea L. Pleasant
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Rebecca K. Harston
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Sundaravadivel Balasubramanian
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| | - Dhandapani Kuppuswamy
- Cardiology Division of the Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC 29425-2221
| |
Collapse
|
48
|
Dhingra R, Gang H, Wang Y, Biala AK, Aviv Y, Margulets V, Tee A, Kirshenbaum LA. Bidirectional regulation of nuclear factor-κB and mammalian target of rapamycin signaling functionally links Bnip3 gene repression and cell survival of ventricular myocytes. Circ Heart Fail 2013; 6:335-43. [PMID: 23395931 DOI: 10.1161/circheartfailure.112.000061] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tumor necrosis factor-α and other proinflammatory cytokines activate the canonical nuclear factor (NF)-κB pathway through the kinase IKKβ. Previously, we established that IKKβ is also critical for Akt-mediated NF-κB activation in ventricular myocytes. Akt activates the kinase mammalian target of rapamycin (mTOR), which mediates important processes such as cardiac hypertrophy. However, whether mTOR regulates cardiac myocyte cell survival is unknown. METHODS AND RESULTS Herein, we demonstrate bidirectional regulation between NF-κB signaling and mTOR, the balance which determines ventricular myocyte survival. Overexpression of IKKβ resulted in mTOR activation and conversely overexpression of mTOR lead to NF-κB activation. Loss of function approaches demonstrated that endogenous levels of IKKβ and mTOR also signal through this pathway. NF-κB activation by mTOR was mediated by phosphorylation of the NF-κB p65 subunit increasing p65 nuclear translocation and activation of gene transcription. This circuit was also important for NF-κB activation by the canonical tumor necrosis factor-α pathway. Our previous work has shown that NF-κB signaling suppresses transcription of the death gene Bnip3 resulting in ventricular myocyte survival. Inhibition of mTOR with rapamycin decreased NF-κB activation resulting in increased Bnip3 expression and cell death. Conversely, mTOR overexpression suppressed Bnip3 levels and cell death of ventricular myocytes in response to hypoxia. CONCLUSIONS To our knowledge, these data provide the first evidence for a bidirectional link between NF-κB signaling and mTOR that is critical in the regulation of Bnip3 expression and cardiac myocyte death. Hence, modulation of this axis may be cardioprotective during ischemia.
Collapse
Affiliation(s)
- Rimpy Dhingra
- Departments of Physiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Masayuki Shimano
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, Boston, MA 02118, USA
| | | | | |
Collapse
|
50
|
Koga K, Kenessey A, Ojamaa K. Macrophage migration inhibitory factor antagonizes pressure overload-induced cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2012; 304:H282-93. [PMID: 23144312 DOI: 10.1152/ajpheart.00595.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a proinflammatory cytokine when secreted from the cell, but it also exhibits antioxidant properties by virtue of its intrinsic oxidoreductase activity. Since increased production of ROS is implicated in the development of left ventricular hypertrophy, we hypothesized that the redox activity of MIF protects the myocardium when exposed to hemodynamic stress. In a mouse model of myocardial hypertrophy induced by transverse aortic coarctation (TAC) for 10 days, we showed that growth of the MIF-deficient heart was significantly greater by 32% compared with wild-type (WT) TAC hearts and that fibrosis was increased by fourfold (2.62 ± 0.2% vs. 0.6 ± 0.1%). Circulating MIF was increased in TAC animals, and expression of MIF receptor, CD74, was increased in the hypertrophic myocardium. Gene expression analysis showed a 10-fold increase (P < 0.01) in ROS-generating mitochondrial NADPH oxidase and 2- to 3-fold reductions (P < 0.01) in mitochondrial SOD2 and mitochondrial aconitase activities, indicating enhanced oxidative injury in the hypertrophied MIF-deficient ventricle. Hypertrophic signaling pathways showed that phosphorylation of cytosolic glycogen synthase kinase-3α was greater (P < 0.05) at baseline in MIF-deficient hearts than in WT hearts and remained elevated after 10-day TAC. In the hemodynamically stressed MIF-deficient heart, nuclear p21(CIP1) increased sevenfold (P < 0.01), and the cytosolic increase of phospho-p21(CIP1) was significantly greater than in WT TAC hearts. We conclude that MIF antagonizes myocardial hypertrophy and fibrosis in response to hemodynamic stress by maintaining a redox homeostatic phenotype and attenuating stress-induced activation of hypertrophic signaling pathways.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|