1
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
2
|
Balderas-Villalobos J, Medina-Contreras JML, Lynch C, Kabadi R, Hayles J, Ramirez RJ, Tan AY, Kaszala K, Samsó M, Huizar JF, Eltit JM. Mechanisms of adaptive hypertrophic cardiac remodeling in a large animal model of premature ventricular contraction-induced cardiomyopathy. IUBMB Life 2023; 75:926-940. [PMID: 37427864 PMCID: PMC10592397 DOI: 10.1002/iub.2765] [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: 03/05/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023]
Abstract
Frequent premature ventricular contractions (PVCs) promoted eccentric cardiac hypertrophy and reduced ejection fraction (EF) in a large animal model of PVC-induced cardiomyopathy (PVC-CM), but the molecular mechanisms and markers of this hypertrophic remodeling remain unexplored. Healthy mongrel canines were implanted with pacemakers to deliver bigeminal PVCs (50% burden with 200-220 ms coupling interval). After 12 weeks, left ventricular (LV) free wall samples were studied from PVC-CM and Sham groups. In addition to reduced LV ejection fraction (LVEF), the PVC-CM group showed larger cardiac myocytes without evident ultrastructural alterations compared to the Sham group. Biochemical markers of pathological hypertrophy, such as store-operated Ca2+ entry, calcineurin/NFAT pathway, β-myosin heavy chain, and skeletal type α-actin were unaltered in the PVC-CM group. In contrast, pro-hypertrophic and antiapoptotic pathways including ERK1/2 and AKT/mTOR were activated and/or overexpressed in the PVC-CM group, which appeared counterbalanced by an overexpression of protein phosphatase 1 and a borderline elevation of the anti-hypertrophic factor atrial natriuretic peptide. Moreover, the potent angiogenic and pro-hypertrophic factor VEGF-A and its receptor VEGFR2 were significantly elevated in the PVC-CM group. In conclusion, a molecular program is in place to keep this structural remodeling associated with frequent PVCs as an adaptive pathological hypertrophy.
Collapse
Affiliation(s)
| | - JML Medina-Contreras
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Christopher Lynch
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Rajiv Kabadi
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Janée Hayles
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rafael J. Ramirez
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Alex Y. Tan
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Karoly Kaszala
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Montserrat Samsó
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Jose F. Huizar
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Jose M. Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| |
Collapse
|
3
|
Starkova T, Polyanichko A, Tomilin AN, Chikhirzhina E. Structure and Functions of HMGB2 Protein. Int J Mol Sci 2023; 24:ijms24098334. [PMID: 37176041 PMCID: PMC10179549 DOI: 10.3390/ijms24098334] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
High-Mobility Group (HMG) chromosomal proteins are the most numerous nuclear non-histone proteins. HMGB domain proteins are the most abundant and well-studied HMG proteins. They are involved in variety of biological processes. HMGB1 and HMGB2 were the first members of HMGB-family to be discovered and are found in all studied eukaryotes. Despite the high degree of homology, HMGB1 and HMGB2 proteins differ from each other both in structure and functions. In contrast to HMGB2, there is a large pool of works devoted to the HMGB1 protein whose structure-function properties have been described in detail in our previous review in 2020. In this review, we attempted to bring together diverse data about the structure and functions of the HMGB2 protein. The review also describes post-translational modifications of the HMGB2 protein and its role in the development of a number of diseases. Particular attention is paid to its interaction with various targets, including DNA and protein partners. The influence of the level of HMGB2 expression on various processes associated with cell differentiation and aging and its ability to mediate the differentiation of embryonic and adult stem cells are also discussed.
Collapse
Affiliation(s)
- Tatiana Starkova
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Alexander Polyanichko
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Alexey N Tomilin
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | - Elena Chikhirzhina
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| |
Collapse
|
4
|
Chen H, Chen C, Spanos M, Li G, Lu R, Bei Y, Xiao J. Exercise training maintains cardiovascular health: signaling pathways involved and potential therapeutics. Signal Transduct Target Ther 2022; 7:306. [PMID: 36050310 PMCID: PMC9437103 DOI: 10.1038/s41392-022-01153-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Exercise training has been widely recognized as a healthy lifestyle as well as an effective non-drug therapeutic strategy for cardiovascular diseases (CVD). Functional and mechanistic studies that employ animal exercise models as well as observational and interventional cohort studies with human participants, have contributed considerably in delineating the essential signaling pathways by which exercise promotes cardiovascular fitness and health. First, this review summarizes the beneficial impact of exercise on multiple aspects of cardiovascular health. We then discuss in detail the signaling pathways mediating exercise's benefits for cardiovascular health. The exercise-regulated signaling cascades have been shown to confer myocardial protection and drive systemic adaptations. The signaling molecules that are necessary for exercise-induced physiological cardiac hypertrophy have the potential to attenuate myocardial injury and reverse cardiac remodeling. Exercise-regulated noncoding RNAs and their associated signaling pathways are also discussed in detail for their roles and mechanisms in exercise-induced cardioprotective effects. Moreover, we address the exercise-mediated signaling pathways and molecules that can serve as potential therapeutic targets ranging from pharmacological approaches to gene therapies in CVD. We also discuss multiple factors that influence exercise's effect and highlight the importance and need for further investigations regarding the exercise-regulated molecules as therapeutic targets and biomarkers for CVD as well as the cross talk between the heart and other tissues or organs during exercise. We conclude that a deep understanding of the signaling pathways involved in exercise's benefits for cardiovascular health will undoubtedly contribute to the identification and development of novel therapeutic targets and strategies for CVD.
Collapse
Affiliation(s)
- Huihua Chen
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chen Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Rong Lu
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yihua Bei
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
5
|
Ahmadi F, Ghanbar Zadeh M, Habibi A, Karimi F. Effect of resistance training with Spirulina platensis on PI3K/Akt/mTOR/p70S6k signaling pathway in cardiac muscle. Sci Sports 2020. [DOI: 10.1016/j.scispo.2019.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
6
|
Ikeda S, Mizushima W, Sciarretta S, Abdellatif M, Zhai P, Mukai R, Fefelova N, Oka SI, Nakamura M, Del Re DP, Farrance I, Park JY, Tian B, Xie LH, Kumar M, Hsu CP, Sadayappan S, Shimokawa H, Lim DS, Sadoshima J. Hippo Deficiency Leads to Cardiac Dysfunction Accompanied by Cardiomyocyte Dedifferentiation During Pressure Overload. Circ Res 2019; 124:292-305. [PMID: 30582455 DOI: 10.1161/circresaha.118.314048] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE The Hippo pathway plays an important role in determining organ size through regulation of cell proliferation and apoptosis. Hippo inactivation and consequent activation of YAP (Yes-associated protein), a transcription cofactor, have been proposed as a strategy to promote myocardial regeneration after myocardial infarction. However, the long-term effects of Hippo deficiency on cardiac function under stress remain unknown. OBJECTIVE We investigated the long-term effect of Hippo deficiency on cardiac function in the presence of pressure overload (PO). METHODS AND RESULTS We used mice with cardiac-specific homozygous knockout of WW45 (WW45cKO), in which activation of Mst1 (Mammalian sterile 20-like 1) and Lats2 (large tumor suppressor kinase 2), the upstream kinases of the Hippo pathway, is effectively suppressed because of the absence of the scaffolding protein. We used male mice at 3 to 4 month of age in all animal experiments. We subjected WW45cKO mice to transverse aortic constriction for up to 12 weeks. WW45cKO mice exhibited higher levels of nuclear YAP in cardiomyocytes during PO. Unexpectedly, the progression of cardiac dysfunction induced by PO was exacerbated in WW45cKO mice, despite decreased apoptosis and activated cardiomyocyte cell cycle reentry. WW45cKO mice exhibited cardiomyocyte sarcomere disarray and upregulation of TEAD1 (transcriptional enhancer factor) target genes involved in cardiomyocyte dedifferentiation during PO. Genetic and pharmacological inactivation of the YAP-TEAD1 pathway reduced the PO-induced cardiac dysfunction in WW45cKO mice and attenuated cardiomyocyte dedifferentiation. Furthermore, the YAP-TEAD1 pathway upregulated OSM (oncostatin M) and OSM receptors, which played an essential role in mediating cardiomyocyte dedifferentiation. OSM also upregulated YAP and TEAD1 and promoted cardiomyocyte dedifferentiation, indicating the existence of a positive feedback mechanism consisting of YAP, TEAD1, and OSM. CONCLUSIONS Although activation of YAP promotes cardiomyocyte regeneration after cardiac injury, it induces cardiomyocyte dedifferentiation and heart failure in the long-term in the presence of PO through activation of the YAP-TEAD1-OSM positive feedback mechanism.
Collapse
Affiliation(s)
- Shohei Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Wataru Mizushima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S. Sciarretta).,Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S. Sciarretta)
| | - Maha Abdellatif
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Risa Mukai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Nadezhda Fefelova
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Shin-Ichi Oka
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Michinari Nakamura
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Dominic P Del Re
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | | | - Ji Yeon Park
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Lai-Hua Xie
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Dae-Sik Lim
- Department of Biological Science, National Creative Research Initiatives Center for Cell Division and Differentiation, Korea Advanced Institute of Science and Technology, Daejeon (D.-S.L.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| |
Collapse
|
7
|
Stewart RM, Rodriguez EC, King MC. Ablation of SUN2-containing LINC complexes drives cardiac hypertrophy without interstitial fibrosis. Mol Biol Cell 2019; 30:1664-1675. [PMID: 31091167 PMCID: PMC6727752 DOI: 10.1091/mbc.e18-07-0438] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cardiomyocyte cytoskeleton, including the sarcomeric contractile apparatus, forms a cohesive network with cellular adhesions at the plasma membrane and nuclear--cytoskeletal linkages (LINC complexes) at the nuclear envelope. Human cardiomyopathies are genetically linked to the LINC complex and A-type lamins, but a full understanding of disease etiology in these patients is lacking. Here we show that SUN2-null mice display cardiac hypertrophy coincident with enhanced AKT/MAPK signaling, as has been described previously for mice lacking A-type lamins. Surprisingly, in contrast to lamin A/C-null mice, SUN2-null mice fail to show coincident fibrosis or upregulation of pathological hypertrophy markers. Thus, cardiac hypertrophy is uncoupled from profibrotic signaling in this mouse model, which we tie to a requirement for the LINC complex in productive TGFβ signaling. In the absence of SUN2, we detect elevated levels of the integral inner nuclear membrane protein MAN1, an established negative regulator of TGFβ signaling, at the nuclear envelope. We suggest that A-type lamins and SUN2 play antagonistic roles in the modulation of profibrotic signaling through opposite effects on MAN1 levels at the nuclear lamina, suggesting a new perspective on disease etiology.
Collapse
Affiliation(s)
- Rachel M Stewart
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520-8002
| | - Elisa C Rodriguez
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520-8002
| | - Megan C King
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520-8002
| |
Collapse
|
8
|
Pathological cardiac hypertrophy: the synergy of adenylyl cyclases inhibition in cardiac and immune cells during chronic catecholamine stress. J Mol Med (Berl) 2019; 97:897-907. [PMID: 31062036 DOI: 10.1007/s00109-019-01790-0] [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: 01/19/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022]
Abstract
Response to stressors in our environment and daily lives is an adaptation conserved through evolution as it is beneficial in enhancing the survival and continuity of humans. Although stressors have evolved, the drastic physiological response they elicit still remains unchanged. The chronic secretion and circulation of catecholamines to produce physical responses when they are not required may result in pathological consequences which affect cardiac function drastically. This review seeks to point out the probable implication of chronic stress in inducing an inflammation disorder in the heart. We discussed the likely synergy of a G protein-independent stimuli signaling via β2-adrenergic receptors in both cardiomyocytes and immune cells during chronic catecholamine stress. To explain this synergy, we hypothesized the possibility of adenylyl cyclases having a regulatory effect on G protein-coupled receptor kinases. This was based on the negative correlations they exhibit during normal cardiac function and heart failures. As such, the downregulation of adenylyl cyclases in cardiomyocytes and immune cells during chronic catecholamine stress enhances the expressions of G protein-coupled receptor kinases. In addition, we explain the maladaptive roles played by G protein-coupled receptor kinase and extracellular signal-regulated kinase in the synergistic cascade that pathologically remodels the heart. Finally, we highlighted the therapeutic potentials of an adenylyl cyclases stimulator to attenuate pathological cardiac hypertrophy (PCH) and improve cardiac function in patients developing cardiac disorders due to chronic catecholamine stress.
Collapse
|
9
|
Sato M, Miyata K, Tian Z, Kadomatsu T, Ujihara Y, Morinaga J, Horiguchi H, Endo M, Zhao J, Zhu S, Sugizaki T, Igata K, Muramatsu M, Minami T, Ito T, Bianchi ME, Mohri S, Araki K, Node K, Oike Y. Loss of Endogenous HMGB2 Promotes Cardiac Dysfunction and Pressure Overload-Induced Heart Failure in Mice. Circ J 2019; 83:368-378. [PMID: 30487376 DOI: 10.1253/circj.cj-18-0925] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
BACKGROUND The rapid increase in the number of heart failure (HF) patients in parallel with the increase in the number of older people is receiving attention worldwide. HF not only increases mortality but decreases quality of life, creating medical and social problems. Thus, it is necessary to define molecular mechanisms underlying HF development and progression. HMGB2 is a member of the high-mobility group superfamily characterized as nuclear proteins that bind DNA to stabilize nucleosomes and promote transcription. A recent in vitro study revealed that HMGB2 loss in cardiomyocytes causes hypertrophy and increases HF-associated gene expression. However, it's in vivo function in the heart has not been assessed. METHODS AND RESULTS Western blotting analysis revealed increased HMGB2 expression in heart tissues undergoing pressure overload by transverse aorta constriction (TAC) in mice. Hmgb2 homozygous knockout (Hmgb2-/-) mice showed cardiac dysfunction due to AKT inactivation and decreased sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a activity. Compared to wild-type mice, Hmgb2-/- mice had worsened cardiac dysfunction after TAC surgery, predisposing mice to HF development and progression. CONCLUSIONS This study demonstrates that upregulation of cardiac HMGB2 is an adaptive response to cardiac stress, and that loss of this response could accelerate cardiac dysfunction, suggesting that HMGB2 plays a cardioprotective role.
Collapse
Affiliation(s)
- Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Department of Cardiovascular Medicine, Saga University
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Department of Immunology, Allergy and Vascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Zhe Tian
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | | | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Motoyoshi Endo
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Jiabin Zhao
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Shunshun Zhu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Taichi Sugizaki
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Kimihiro Igata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Masashi Muramatsu
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University
| | - Takashi Minami
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science
| | - Marco E Bianchi
- Chromatin Dynamics Unit, San Raffaele University and Scientific Institute
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging, Graduate School of Medical Sciences, Kumamoto University
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Center for Metabolic Regulation of Healthy Aging, Graduate School of Medical Sciences, Kumamoto University
| |
Collapse
|
10
|
Bozi LH, Takano AP, Campos JC, Rolim N, Dourado PM, Voltarelli VA, Wisløff U, Ferreira JC, Barreto-Chaves ML, Brum PC. Endoplasmic reticulum stress impairs cardiomyocyte contractility through JNK-dependent upregulation of BNIP3. Int J Cardiol 2018; 272:194-201. [DOI: 10.1016/j.ijcard.2018.08.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022]
|
11
|
Ramasamy S, Velmurugan G, Rekha B, Anusha S, Shanmugha Rajan K, Shanmugarajan S, Ramprasath T, Gopal P, Tomar D, Karthik KV, Verma SK, Garikipati VNS, Sudarsan R. Egr-1 mediated cardiac miR-99 family expression diverges physiological hypertrophy from pathological hypertrophy. Exp Cell Res 2018; 365:46-56. [DOI: 10.1016/j.yexcr.2018.02.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/24/2018] [Accepted: 02/16/2018] [Indexed: 01/08/2023]
|
12
|
Guo C, Cho KS, Li Y, Tchedre K, Antolik C, Ma J, Chew J, Utheim TP, Huang XA, Yu H, Malik MTA, Anzak N, Chen DF. IGFBPL1 Regulates Axon Growth through IGF-1-mediated Signaling Cascades. Sci Rep 2018; 8:2054. [PMID: 29391597 PMCID: PMC5794803 DOI: 10.1038/s41598-018-20463-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 01/19/2018] [Indexed: 11/29/2022] Open
Abstract
Activation of axonal growth program is a critical step in successful optic nerve regeneration following injury. Yet the molecular mechanisms that orchestrate this developmental transition are not fully understood. Here we identified a novel regulator, insulin-like growth factor binding protein-like 1 (IGFBPL1), for the growth of retinal ganglion cell (RGC) axons. Expression of IGFBPL1 correlates with RGC axon growth in development, and acute knockdown of IGFBPL1 with shRNA or IGFBPL1 knockout in vivo impaired RGC axon growth. In contrast, administration of IGFBPL1 promoted axon growth. Moreover, IGFBPL1 bound to insulin-like growth factor 1 (IGF-1) and subsequently induced calcium signaling and mammalian target of rapamycin (mTOR) phosphorylation to stimulate axon elongation. Blockage of IGF-1 signaling abolished IGFBPL1-mediated axon growth, and vice versa, IGF-1 required the presence of IGFBPL1 to promote RGC axon growth. These data reveal a novel element in the control of RGC axon growth and suggest an unknown signaling loop in the regulation of the pleiotropic functions of IGF-1. They suggest new therapeutic target for promoting optic nerve and axon regeneration and repair of the central nervous system.
Collapse
Affiliation(s)
- Chenying Guo
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yingqian Li
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Kissauo Tchedre
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Christian Antolik
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jie Ma
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Justin Chew
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
- Pritzker School of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Tor Paaske Utheim
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
- Department of Medical Biochemistry, Oslo University Hospital, Kirkeveien 166, 0407, Oslo, Norway
| | - Xizhong A Huang
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
- Oncology Translational Medicine, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, 02138, USA
| | - Honghua Yu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Muhammad Taimur A Malik
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Nada Anzak
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
- Guys, Kings & St Thomas' School of Medicine, Hodgkin Building, Guy's Campus, King's College London, London, UK
| | - Dong Feng Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
- Boston VA Healthcare System, 150 S. Huntington Ave, Boston, MA, 02130, USA.
| |
Collapse
|
13
|
Chemaly ER, Troncone L, Lebeche D. SERCA control of cell death and survival. Cell Calcium 2017; 69:46-61. [PMID: 28747251 DOI: 10.1016/j.ceca.2017.07.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 12/31/2022]
Abstract
Intracellular calcium (Ca2+) is a critical coordinator of various aspects of cellular physiology. It is increasingly apparent that changes in cellular Ca2+ dynamics contribute to the regulation of normal and pathological signal transduction that controls cell growth and survival. Aberrant perturbations in Ca2+ homeostasis have been implicated in a range of pathological conditions, such as cardiovascular diseases, diabetes, tumorigenesis and steatosis hepatitis. Intracellular Ca2+ concentrations are therefore tightly regulated by a number of Ca2+ handling enzymes, proteins, channels and transporters located in the plasma membrane and in Ca2+ storage organelles, which work in concert to fine tune a temporally and spatially precise Ca2+ signal. Chief amongst them is the sarco/endoplasmic reticulum (SR/ER) Ca2+ ATPase pump (SERCA) which actively re-accumulates released Ca2+ back into the SR/ER, therefore maintaining Ca2+ homeostasis. There are at least 14 different SERCA isoforms encoded by three ATP2A1-3 genes whose expressions are species- and tissue-specific. Altered SERCA expression and activity results in cellular malignancy and induction of ER stress and ER stress-associated apoptosis. The role of SERCA misregulation in the control of apoptosis in various cell types and disease setting with prospective therapeutic implications is the focus of this review. Ca2+ is a double edge sword for both life as well as death, and current experimental evidence supports a model in which Ca2+ homeostasis and SERCA activity represent a nodal point that controls cell survival. Pharmacological or genetic targeting of this axis constitutes an incredible therapeutic potential to treat different diseases sharing similar biological disorders.
Collapse
Affiliation(s)
- Elie R Chemaly
- Division of Nephrology and Hypertension, Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Luca Troncone
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
14
|
Matsuda T, Jeong JI, Ikeda S, Yamamoto T, Gao S, Babu GJ, Zhai P, Del Re DP. H-Ras Isoform Mediates Protection Against Pressure Overload-Induced Cardiac Dysfunction in Part Through Activation of AKT. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003658. [PMID: 28193718 DOI: 10.1161/circheartfailure.116.003658] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 01/11/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND In general, Ras proteins are thought to promote cardiac hypertrophy, an important risk factor for cardiovascular disease and heart failure. However, the contribution of different Ras isoforms has not been investigated. The objective of this study was to define the role of H- and K-Ras in modulating stress-induced myocardial hypertrophy and failure. METHODS AND RESULTS We used H- and K-Ras gene knockout mice and subjected them to pressure overload to induce cardiac hypertrophy and dysfunction. We observed a worsened cardiac phenotype in Hras-/- mice, while outcomes were improved in Kras+/- mice. We also used a neonatal rat cardiomyocyte culture system to elucidate the mechanisms underlying these observations. Our findings demonstrate that H-Ras, but not K-Ras, promotes cardiomyocyte hypertrophy both in vivo and in vitro. This response was mediated in part through the phosphoinositide 3-kinase-AKT signaling pathway. Adeno-associated virus-mediated increase in AKT activation improved the cardiac function in pressure overloaded Hras null hearts in vivo. These findings further support engagement of the phosphoinositide 3-kinase-AKT signaling axis by H-Ras. CONCLUSIONS Taken together, these findings indicate that H- and K-Ras have divergent effects on cardiac hypertrophy and heart failure in response to pressure overload stress.
Collapse
Affiliation(s)
- Takahisa Matsuda
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Jae Im Jeong
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Shohei Ikeda
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Takanobu Yamamoto
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Shumin Gao
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Gopal J Babu
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Peiyong Zhai
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ
| | - Dominic P Del Re
- From the Cardiovascular Research Institute and the Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ.
| |
Collapse
|
15
|
Okuhara Y, Yokoe S, Iwasaku T, Eguchi A, Nishimura K, Li W, Oboshi M, Naito Y, Mano T, Asahi M, Okamura H, Masuyama T, Hirotani S. Interleukin-18 gene deletion protects against sepsis-induced cardiac dysfunction by inhibiting PP2A activity. Int J Cardiol 2017; 243:396-403. [PMID: 28526544 DOI: 10.1016/j.ijcard.2017.04.082] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/20/2017] [Accepted: 04/24/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Interleukin-18 (IL-18) neutralization protects against lipopolysaccharide (LPS)-induced injuries, including myocardial dysfunction. However, the mechanism is yet to be fully elucidated. The aim of the present study was to determine whether IL-18 gene deletion prevents sepsis-induced cardiac dysfunction and to elucidate the potential mechanisms underlying IL-18-mediated cardiotoxicity by LPS. METHODS AND RESULTS Ten-week-old male wild-type (WT) and IL-18 knockout (IL-18 KO) mice were intraperitoneally administered LPS. Serial echocardiography showed better systolic pump function and less left ventricular (LV) dilatation in LPS-treated IL-18 KO mice compared with those in LPS-treated WT mice. LPS treatment significantly decreased the levels of phospholamban (PLN) and Akt phosphorylation in WT mice compared with those in saline-treated WT mice, while the LPS-induced decrease in the phosphorylation levels was attenuated in IL-18 KO mice compared with that in WT mice. IL-18 gene deletion also attenuated an LPS-induced increase of type 2 protein phosphatase 2A (PP2A) activity, a molecule that dephosphorylates PLN and Akt. There was no difference in type 1 protein phosphatase (PP1) activity. To address whether IL-18 affects PLN and Akt phosphorylation via PP2A activation in cardiomyocytes, rat neonatal cardiac myocytes were cultured and stimulated using 100ng/ml of recombinant rat IL-18. Exogenous IL-18 decreased the level of PLN and Akt phosphorylation in cardiomyocytes. PP2A activity but not PP1 activity was increased by IL-18 stimulation in cardiomyocytes. CONCLUSIONS IL-18 plays a pivotal role in advancing sepsis-induced cardiac dysfunction, and the mechanisms underlying IL-18-mediated cardiotoxicity potentially involve the regulation of PLN and Akt phosphorylation through PP2A activity.
Collapse
Affiliation(s)
- Yoshitaka Okuhara
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shunichi Yokoe
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Toshihiro Iwasaku
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Akiyo Eguchi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Koichi Nishimura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Makiko Oboshi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yoshiro Naito
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toshiaki Mano
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tohru Masuyama
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shinichi Hirotani
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.
| |
Collapse
|
16
|
Weeks KL, Bernardo BC, Ooi JYY, Patterson NL, McMullen JR. The IGF1-PI3K-Akt Signaling Pathway in Mediating Exercise-Induced Cardiac Hypertrophy and Protection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:187-210. [PMID: 29098623 DOI: 10.1007/978-981-10-4304-8_12] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Regular physical activity or exercise training can lead to heart enlargement known as cardiac hypertrophy. Cardiac hypertrophy is broadly defined as an increase in heart mass. In adults, cardiac hypertrophy is often considered a poor prognostic sign because it often progresses to heart failure. Heart enlargement in a setting of cardiac disease is referred to as pathological cardiac hypertrophy and is typically characterized by cell death and depressed cardiac function. By contrast, physiological cardiac hypertrophy, as occurs in response to chronic exercise training (i.e. the 'athlete's heart'), is associated with normal or enhanced cardiac function. The following chapter describes the morphologically distinct types of heart growth, and the key role of the insulin-like growth factor 1 (IGF1) - phosphoinositide 3-kinase (PI3K)-Akt signaling pathway in regulating exercise-induced physiological cardiac hypertrophy and cardiac protection. Finally we summarize therapeutic approaches that target the IGF1-PI3K-Akt signaling pathway which are showing promise in preclinical models of heart disease.
Collapse
Affiliation(s)
- Kate L Weeks
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia.
| | - Bianca C Bernardo
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Jenny Y Y Ooi
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Natalie L Patterson
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Julie R McMullen
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
17
|
ANGPTL2 activity in cardiac pathologies accelerates heart failure by perturbing cardiac function and energy metabolism. Nat Commun 2016; 7:13016. [PMID: 27677409 PMCID: PMC5052800 DOI: 10.1038/ncomms13016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/25/2016] [Indexed: 12/21/2022] Open
Abstract
A cardioprotective response that alters ventricular contractility or promotes cardiomyocyte enlargement occurs with increased workload in conditions such as hypertension. When that response is excessive, pathological cardiac remodelling occurs, which can progress to heart failure, a leading cause of death worldwide. Mechanisms underlying this response are not fully understood. Here, we report that expression of angiopoietin-like protein 2 (ANGPTL2) increases in pathologically-remodeled hearts of mice and humans, while decreased cardiac ANGPTL2 expression occurs in physiological cardiac remodelling induced by endurance training in mice. Mice overexpressing ANGPTL2 in heart show cardiac dysfunction caused by both inactivation of AKT and sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a signalling and decreased myocardial energy metabolism. Conversely, Angptl2 knockout mice exhibit increased left ventricular contractility and upregulated AKT-SERCA2a signalling and energy metabolism. Finally, ANGPTL2-knockdown in mice subjected to pressure overload ameliorates cardiac dysfunction. Overall, these studies suggest that therapeutic ANGPTL2 suppression could antagonize development of heart failure. Heart responds to increased workload by enlarging cardiomyocytes to preserve function, but in pathologies hypertrophy leads to heart failure. Here the authors show that ANGPTL2 activity in the heart is critical for determining beneficial vs. pathological hypertrophy via its effect on AKT-SERCA2a signaling and myocardial energy.
Collapse
|
18
|
Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol 2016; 97:245-62. [PMID: 27262674 DOI: 10.1016/j.yjmcc.2016.06.001] [Citation(s) in RCA: 637] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high energy consumption required by this role, the heart is equipped with multiple complex biological systems that allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and metabolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as physiological when it is associated with normal cardiac function or as pathological when associated with cardiac dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyocyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladaptive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses involved in physiological/pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
19
|
Manning JR, Withers CN, Levitan B, Smith JD, Andres DA, Satin J. Loss of Rad-GTPase produces a novel adaptive cardiac phenotype resistant to systolic decline with aging. Am J Physiol Heart Circ Physiol 2015; 309:H1336-45. [PMID: 26371164 DOI: 10.1152/ajpheart.00389.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/02/2015] [Indexed: 12/26/2022]
Abstract
Rad-GTPase is a regulator of L-type calcium current (LTCC), with increased calcium current observed in Rad knockout models. While mouse models that result in elevated LTCC have been associated with heart failure, our laboratory and others observe a hypercontractile phenotype with enhanced calcium homeostasis in Rad(-/-). It is currently unclear whether this observation represents an early time point in a decompensatory progression towards heart failure or whether Rad loss drives a novel phenotype with stable enhanced function. We test the hypothesis that Rad(-/-) drives a stable nonfailing hypercontractile phenotype in adult hearts, and we examine compensatory regulation of sarcoplasmic reticulum (SR) loading and protein changes. Heart function was measured in vivo with echocardiography. In vivo heart function was significantly improved in adult Rad(-/-) hearts compared with wild type. Heart wall dimensions were significantly increased, while heart size was decreased, and cardiac output was not changed. Cardiac function was maintained through 18 mo of age with no decompensation. SR releasable Ca(2+) was increased in isolated Rad(-/-) ventricular myocytes. Higher Ca(2+) load was accompanied by sarco/endoplasmic reticulum Ca(2+) ATPase 2a (SERCA2a) protein elevation as determined by immunoblotting and a rightward shift in the thapsigargan inhibitor-response curve. Rad(-/-) promotes morphological changes accompanied by a stable increase in contractility with aging and preserved cardiac output. The Rad(-/-) phenotype is marked by enhanced systolic and diastolic function with increased SR uptake, which is consistent with a model that does not progress into heart failure.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Physiology, University of Kentucky, Lexington, Kentucky; Department of Biochemistry, University of Kentucky, Lexington, Kentucky; and
| | - Catherine N Withers
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky; and
| | - Bryana Levitan
- Gill Heart Institute, University of Kentucky, Lexington, Kentucky
| | - Jeffrey D Smith
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky; and
| | - Douglas A Andres
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky; and
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, Kentucky;
| |
Collapse
|
20
|
Kana K, Song H, Laschinger C, Zandstra PW, Radisic M. PI3K Phosphorylation Is Linked to Improved Electrical Excitability in an In Vitro Engineered Heart Tissue Disease Model System. Tissue Eng Part A 2015; 21:2379-89. [PMID: 26120935 DOI: 10.1089/ten.tea.2014.0412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Myocardial infarction, a prevalent cardiovascular disease, is associated with cardiomyocyte cell death, and eventually heart failure. Cardiac tissue engineering has provided hopes for alternative treatment options, and high-fidelity tissue models for drug discovery. The signal transduction mechanisms relayed in response to mechanoelectrical (physical) stimulation or biochemical stimulation (hormones, cytokines, or drugs) in engineered heart tissues (EHTs) are poorly understood. In this study, an EHT model was used to elucidate the signaling mechanisms involved when insulin was applied in the presence of electrical stimulation, a stimulus that mimics functional heart tissue environment in vitro. EHTs were insulin treated, electrically stimulated, or applied in combination (insulin and electrical stimulation). Electrical excitability parameters (excitation threshold and maximum capture rate) were measured. Protein kinase B (AKT) and phosphatidylinositol-3-kinase (PI3K) phosphorylation revealed that insulin and electrical stimulation relayed electrical excitability through two separate signaling cascades, while there was a negative crosstalk between sustained activation of AKT and PI3K.
Collapse
Affiliation(s)
- Kujaany Kana
- 1 Institute of Biomaterials and Biomedical Engineering , Toronto, Canada
| | - Hannah Song
- 1 Institute of Biomaterials and Biomedical Engineering , Toronto, Canada
| | - Carol Laschinger
- 2 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Canada
| | - Peter W Zandstra
- 1 Institute of Biomaterials and Biomedical Engineering , Toronto, Canada .,3 Heart & Stroke/Richard Lewar Centre of Excellence , Toronto, Canada .,4 McEwen Centre for Regenerative Medicine, University of Health Network , Toronto, Canada
| | - Milica Radisic
- 1 Institute of Biomaterials and Biomedical Engineering , Toronto, Canada .,2 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Canada .,3 Heart & Stroke/Richard Lewar Centre of Excellence , Toronto, Canada
| |
Collapse
|
21
|
Chang P, Wang Q, Xu H, Yang M, Lin X, Li X, Zhang Z, Zhang X, Zhao F, Zhao X, Bai F, Yu J. Tetrahydrobiopterin reverse left ventricular hypertrophy and diastolic dysfunction through the PI3K/p-Akt pathway in spontaneously hypertensive rats. Biochem Biophys Res Commun 2015; 463:1012-20. [PMID: 26093301 DOI: 10.1016/j.bbrc.2015.06.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/08/2015] [Indexed: 11/23/2022]
Abstract
Hypertension induced hypertrophy and diastolic dysfunction and is associated with cardiac oxidation and reduced NO production. We hypothesized that tetrahydrobiopterin (BH4) can regulate the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway and reverse cardiac hypertrophy and diastolic dysfunction in spontaneously hypertensive rats. Ten-week-old male spontaneously hypertensive rats (SHR) and age-matched normotensive control Wistar-Kyoto (WKY) rats were divided into five groups, WKY, WKY + BH4, SHR, SHR + BH4 and SHR + VAL. In SHR, diastolic dysfunction was accompanied by concentric hypertrophy, cardiac oxidation, and reduced cardiac BH4 and NO production. Four-week BH4 and valsartan administration reversed hypertrophy and improved diastolic function. BH4 and valsartan blunted the expression of hypertrophy markers α-skeletal actin (α-SA) and β-myosin heavy chain (β-MHC). Only BH4 reduced hypertension and induced myocardial fibrosis and expression of transforming growth factor-β1 (TGF-β1). BH4 reduced cardiac oxidant stress and increased NO production. Exogenous BH4 increased phosphorylated Akt levels and increased Bcl-2 expression. In conclusion, less BH4 and reduced NO increases myocardial hypertrophy and cardiac oxidative stress, which exacerbates diastolic dysfunction. Exogenous BH4 ameliorates cardiac hypertrophy and diastolic dysfunction through the PI3K/p-Akt pathway. BH4 may be a potent therapy for hypertension with diastolic dysfunction.
Collapse
Affiliation(s)
- Peng Chang
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Qiongying Wang
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Han Xu
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Mina Yang
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Xin Lin
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Xiuli Li
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Zhengyi Zhang
- Department of General Medicine, Lanzhou University Second Hospital, Gansu, China
| | - Xiaowei Zhang
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Feng Zhao
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Xu Zhao
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Feng Bai
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China
| | - Jing Yu
- Department of Cardiology, Lanzhou University Second Hospital, Gansu, China.
| |
Collapse
|
22
|
Clay SA, Domeier TL, Hanft LM, McDonald KS, Krenz M. Elevated Ca2+ transients and increased myofibrillar power generation cause cardiac hypercontractility in a model of Noonan syndrome with multiple lentigines. Am J Physiol Heart Circ Physiol 2015; 308:H1086-95. [PMID: 25724491 PMCID: PMC4551123 DOI: 10.1152/ajpheart.00501.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 02/22/2015] [Indexed: 01/19/2023]
Abstract
Noonan syndrome with multiple lentigines (NSML) is primarily caused by mutations in the nonreceptor protein tyrosine phosphatase SHP2 and associated with congenital heart disease in the form of pulmonary valve stenosis and hypertrophic cardiomyopathy (HCM). Our goal was to elucidate the cellular mechanisms underlying the development of HCM caused by the Q510E mutation in SHP2. NSML patients carrying this mutation suffer from a particularly severe form of HCM. Drawing parallels to other, more common forms of HCM, we hypothesized that altered Ca(2+) homeostasis and/or sarcomeric mechanical properties play key roles in the pathomechanism. We used transgenic mice with cardiomyocyte-specific expression of Q510E-SHP2 starting before birth. Mice develop neonatal onset HCM with increased ejection fraction and fractional shortening at 4-6 wk of age. To assess Ca(2+) handling, isolated cardiomyocytes were loaded with fluo-4. Q510E-SHP2 expression increased Ca(2+) transient amplitudes during excitation-contraction coupling and increased sarcoplasmic reticulum Ca(2+) content concurrent with increased expression of sarco(endo)plasmic reticulum Ca(2+)-ATPase. In skinned cardiomyocyte preparations from Q510E-SHP2 mice, force-velocity relationships and power-load curves were shifted upward. The peak power-generating capacity was increased approximately twofold. Transmission electron microscopy revealed that the relative intracellular area occupied by sarcomeres was increased in Q510E-SHP2 cardiomyocytes. Triton X-100-based myofiber purification showed that Q510E-SHP2 increased the amount of sarcomeric proteins assembled into myofibers. In summary, Q510E-SHP2 expression leads to enhanced contractile performance early in disease progression by augmenting intracellular Ca(2+) cycling and increasing the number of power-generating sarcomeres. This gives important new insights into the cellular pathomechanisms of Q510E-SHP2-associated HCM.
Collapse
Affiliation(s)
- Sarah A Clay
- Department of Medical Pharmacology and Physiology/Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology/Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology/Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology/Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology/Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
23
|
Ballou LM, Lin RZ, Cohen IS. Control of cardiac repolarization by phosphoinositide 3-kinase signaling to ion channels. Circ Res 2015; 116:127-37. [PMID: 25552692 DOI: 10.1161/circresaha.116.303975] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Upregulation of phosphoinositide 3-kinase (PI3K) signaling is a common alteration in human cancer, and numerous drugs that target this pathway have been developed for cancer treatment. However, recent studies have implicated inhibition of the PI3K signaling pathway as the cause of a drug-induced long-QT syndrome in which alterations in several ion currents contribute to arrhythmogenic drug activity. Surprisingly, some drugs that were thought to induce long-QT syndrome by direct block of the rapid delayed rectifier (IKr) also seem to inhibit PI3K signaling, an effect that may contribute to their arrhythmogenicity. The importance of PI3K in regulating cardiac repolarization is underscored by evidence that QT interval prolongation in diabetes mellitus also may result from changes in multiple currents because of decreased insulin activation of PI3K in the heart. How PI3K signaling regulates ion channels to control the cardiac action potential is poorly understood. Hence, this review summarizes what is known about the effect of PI3K and its downstream effectors, including Akt, on sodium, potassium, and calcium currents in cardiac myocytes. We also refer to some studies in noncardiac cells that provide insight into potential mechanisms of ion channel regulation by this signaling pathway in the heart. Drug development and safety could be improved with a better understanding of the mechanisms by which PI3K regulates cardiac ion channels and the extent to which PI3K inhibition contributes to arrhythmogenic susceptibility.
Collapse
Affiliation(s)
- Lisa M Ballou
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.)
| | - Richard Z Lin
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| | - Ira S Cohen
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| |
Collapse
|
24
|
Henao-Martínez AF, Agler AH, Watson AM, Hennessy C, Davidson E, Demos-Davies K, McKinsey TA, Wilson M, Schwartz DA, Yang IV. AKT network of genes and impaired myocardial contractility during murine acute Chagasic myocarditis. Am J Trop Med Hyg 2015; 92:523-9. [PMID: 25582694 DOI: 10.4269/ajtmh.14-0433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Chagasic disease is associated with high morbidity in Latin America. Acute Chagasic myocarditis is consistently found in acute infections, but little is known about its contribution to chronic cardiomyopathy. The aim of the study was to phenotypically characterize two strains of mice with differential Chagas infection susceptibility and correlate strain myocarditis phenotypes with heart tissue gene expression. C57BL/6J and Balb/c mice were injected intraperitoneally with 0 or 150-200 tissue-derived trypomastigotes (Tulahuen strain). Echocardiograms, brain natriuretic peptide, and troponin were measured. Heart tissue was harvested for histopathological analysis and gene expression profiling on microarrays. Genes differently expressed between infected Balb/c and C57BL/6J mice were identified. Echocardiograms showed differences in Balb/c versus C57BL/6J infected mice in heart rate (413 versus 476 beats per minute; P = 0.0001), stroke volume (31.9 ± 9.3 versus 39.2 ± 5.5 μL; P = 0.03), and cardiac output (13.1 ± 3.5 versus 18.7 ± 3.2 μL/min; P = 0.002). Gene expression at 4 weeks analysis showed 32 statistically significant (q value < 0.05) differentially expressed genes between infected Balb/c and C57BL/6J mice that were enriched for genes related to the protein kinase B (AKT) pathway. These specific phenotypic features of cardiac response during acute Chagasic myocarditis may, in part, be related to host AKT network regulation.
Collapse
Affiliation(s)
- Andrés F Henao-Martínez
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Anne Hermetet Agler
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Alan M Watson
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Corinne Hennessy
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Elizabeth Davidson
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Kim Demos-Davies
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Timothy A McKinsey
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Michael Wilson
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - David A Schwartz
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| | - Ivana V Yang
- Division of Infectious Diseases and Departments of Medicine and Immunology, University of Colorado, Denver, Colorado; Department of Epidemiology, Colorado School of Public Health, Denver, Colorado; Department of Pathology, Denver Health, Denver, Colorado
| |
Collapse
|
25
|
Ghantous CM, Azrak Z, Hanache S, Abou-Kheir W, Zeidan A. Differential Role of Leptin and Adiponectin in Cardiovascular System. Int J Endocrinol 2015; 2015:534320. [PMID: 26064110 PMCID: PMC4433709 DOI: 10.1155/2015/534320] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/23/2015] [Indexed: 02/07/2023] Open
Abstract
Leptin and adiponectin are differentially expressed adipokines in obesity and cardiovascular diseases. Leptin levels are directly associated with adipose tissue mass, while adiponectin levels are downregulated in obesity. Although significantly produced by adipocytes, leptin is also produced by vascular smooth muscle cells and cardiomyocytes. Plasma leptin concentrations are elevated in cases of cardiovascular diseases, such as hypertension, congestive heart failure, and myocardial infarction. As for the event of left ventricular hypertrophy, researchers have been stirring controversy about the role of leptin in this form of cardiac remodeling. In this review, we discuss how leptin has been shown to play an antihypertrophic role in the development of left ventricular hypertrophy through in vitro experiments, population-based cross-sectional studies, and longitudinal cohort studies. Conversely, we also examine how leptin may actually promote left ventricular hypertrophy using in vitro analysis and human-based univariate and multiple linear stepwise regression analysis. On the other hand, as opposed to leptin's generally detrimental effects on the cardiovascular system, adiponectin is a cardioprotective hormone that reduces left ventricular and vascular hypertrophy, oxidative stress, and inflammation. In this review, we also highlight adiponectin signaling and its protective actions on the cardiovascular system.
Collapse
Affiliation(s)
- C. M. Ghantous
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - Z. Azrak
- Department of Pharmacology and Toxicology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - S. Hanache
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - W. Abou-Kheir
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - A. Zeidan
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
- *A. Zeidan:
| |
Collapse
|
26
|
Moc C, Taylor AE, Chesini GP, Zambrano CM, Barlow MS, Zhang X, Gustafsson ÅB, Purcell NH. Physiological activation of Akt by PHLPP1 deletion protects against pathological hypertrophy. Cardiovasc Res 2014; 105:160-70. [PMID: 25411382 DOI: 10.1093/cvr/cvu243] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIMS To examine the role of physiological Akt signalling in pathological hypertrophy through analysis of PHLPP1 (PH domain leucine-rich repeat protein phosphatase) knock-out (KO) mice. METHODS AND RESULTS To investigate the in vivo requirement for 'physiological' control of Akt activation in cardiac growth, we examined the effect of deleting the Akt phosphatase, PHLPP, on the induction of cardiac hypertrophy. Basal Akt phosphorylation increased nearly two-fold in the cardiomyocytes from PHLPP1 KO mice and physiological hypertrophy induced by swimming exercise was accentuated as assessed by increased heart size and myocyte cell area. In contrast, the development of pathophysiological hypertrophy induced by pressure overload and assessed by increases in heart size, myocyte cell area, and hypertrophic gene expression was attenuated. This attenuation coincided with decreased fibrosis and cell death in the KO mice. Cast moulding revealed increased capillary density basally in the KO hearts, which was further elevated relative to wild-type mouse hearts in response to pressure overload. In vitro studies with isolated myocytes in co-culture also demonstrated that PHLPP1 deletion in cardiomyocytes can enhance endothelial tube formation. Expression of the pro-angiogenic factor VEGF was also elevated basally and accentuated in response to transverse aortic constriction in hearts from KO mice. CONCLUSION Our data suggest that enhancing Akt activity by inhibiting its PHLPP1-mediated dephosphorylation promotes processes associated with physiological hypertrophy that may be beneficial in attenuating the development of pathological hypertrophy.
Collapse
Affiliation(s)
- Courtney Moc
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Amy E Taylor
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Gino P Chesini
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Cristina M Zambrano
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Melissa S Barlow
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Xiaoxue Zhang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Nicole H Purcell
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| |
Collapse
|
27
|
Hannoush H, Sachdev V, Brofferio A, Arai AE, LaRocca G, Sapp J, Sidenko S, Brenneman C, Biesecker LG, Keppler-Noreuil KM. Myocardial fat overgrowth in Proteus syndrome. Am J Med Genet A 2014; 167A:103-10. [PMID: 25377688 DOI: 10.1002/ajmg.a.36773] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 08/20/2014] [Indexed: 12/31/2022]
Abstract
Proteus syndrome (PS) is a rare, mosaic disorder with asymmetric and distorting overgrowth of the skeletal system, skin, and adipose tissues. Cardiac abnormalities are rare in this syndrome and only two prior cases have been reported. Many patients with PS followed at our institution underwent transthoracic echocardiograms for preoperative evaluation or as work-up for associated pulmonary disease. Some were noted to have prominent, focal echodense areas in the myocardium. We further investigated cardiac findings in a cohort of children and adult patients with PS. Patients with abnormal echocardiograms were referred for cardiac magnetic resonance imaging, Holter monitoring, and exercise treadmill testing. Twenty children and adults with PS, age 24 months to 50 years old, underwent transthoracic echocardiograms. Seven patients (35%) had focal bright echodense areas within the myocardium suggesting fatty infiltration. The majority of patients had significant involvement of the interventricular septum. The cardiac characteristics of all patients with fatty infiltration on transthoracic echocardiograms were compared to Proteus patients without these findings. There were no significant differences in chamber sizes, mass, systolic or diastolic function. No increased risk of conduction defects or arrhythmias was found. This study shows that abnormal fat overgrowth is a common finding in the myocardium in patients with Proteus syndrome; however, it is not associated with functional derangements or arrhythmias. Further evaluation of a larger number of Proteus patients is needed in order to determine the frequency and prognosis of cardiac involvement. Published 2014. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- H Hannoush
- Cardiovascular and Pulmonary Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lu J, Sun B, Huo R, Wang YC, Yang D, Xing Y, Xiao XL, Xie X, Dong DL. Bone morphogenetic protein-2 antagonizes bone morphogenetic protein-4 induced cardiomyocyte hypertrophy and apoptosis. J Cell Physiol 2014; 229:1503-10. [PMID: 24648278 DOI: 10.1002/jcp.24592] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 02/19/2014] [Indexed: 11/05/2022]
Abstract
Our previous work showed that the expression of bone morphogenetic protein-4 (BMP4) was up-regulated in pathological cardiac hypertrophy models and BMP4 induced cardiomyocyte hypertrophy and apoptosis. Bone morphogenetic protein-2 (BMP2) and BMP4 share greater than 80% amino acid homology and there exists an interaction between BMP2 and BMP4, so the aim of the present study was to elucidate the changes of BMP2 in the cardiac hypertrophy models and the effects of BMP2 on BMP4-induced cardiomyocyte hypertrophy and apoptosis. The in vivo cardiac hypertrophy models were induced by pressure-overload and swimming exercise in mice. BMP2 mRNA and protein expressions increased in pressure-overload and swimming-exercise induced cardiac hypertrophy. BMP2 itself did not elicit cardiomyocyte hypertrophy and apoptosis, but antagonized BMP4-induced cardiomyocyte hypertrophy and apoptosis. BMP2 stimulated Akt in cardiomyocytes and Akt inhibitor prevented the antagonism of BMP2 on BMP4-induced cardiomyocyte apoptosis. Furthermore, BMP2 inhibited BMP4-induced JNK activation in cardiomyocytes. In conclusion, BMP2 antagonizes BMP4-induced cardiomyocyte hypertrophy and apoptosis. The anti-apoptotic effects of BMP2 on BMP4-induced cardiomyocyte apoptosis might be through activating Akt and inhibiting JNK activation.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mourouzis I, Mantzouratou P, Galanopoulos G, Kostakou E, Dhalla AK, Belardinelli L, Pantos C. The beneficial effects of ranolazine on cardiac function after myocardial infarction are greater in diabetic than in nondiabetic rats. J Cardiovasc Pharmacol Ther 2014; 19:457-69. [PMID: 24651516 DOI: 10.1177/1074248414524481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ranolazine (RAN) is known to exert both anti-ischemic and antidiabetic actions. Thus, this study has explored the hypothesis that RAN would have greater effect on the recovery of cardiac function in diabetic mellitus (DM) rat hearts following myocardial infarction (MI). Myocardial infarction was induced in nondiabetic (MI, n = 14) and diabetic (streptozotocin induced; DM-MI, n = 13) Wistar rats by permanent ligation of the left coronary artery. Cardiac function was evaluated using echocardiography (left ventricular ejection fraction %) and in isolated heart preparations by measuring left ventricular developed pressure (LVDP), and the positive and negative first derivative of LVDP (± dp/dt). Ranolazine (20 mg/kg, ip once a day) was administered 24 hours after surgical procedure for 4 weeks to nondiabetic (MI + RAN, n = 17) and diabetic rats (DM-MI + RAN, n = 15). The RAN improved the recovery of function in both the nondiabetic and the diabetic postinfarcted hearts but this effect was greater and achieved statistical significance only in the diabetic group. The RAN resulted in increased levels of phosphorylated protein kinase B (Akt) and mammalian target of rapamycin (mTOR, a component of Akt signaling) in both nondiabetic and diabetic infarcted hearts without changes in the activation of mitogen-activated protein kinases (MAPKs; p38 MAPK, c-Jun N-terminal kinase, and extracellular signal-regulated kinase). In addition, in diabetic hearts, RAN resulted in a significant increase in the ratio of sarcoplasmic Ca(2+)-ATPase/phospholamban (a target of Akt signaling, 2.0-fold increase) and increased levels of phosphorylated calcium-regulated adenosine monophosphate-activated protein kinase (AMPK; 2.0-fold increase). In diabetic animals, RAN increased insulin and lowered glucose levels in serum. In conclusion, the beneficial effect of RAN on the recovery of cardiac function after MI was greater in DM rats. This response was associated with activation of Akt/mTOR and AMPK. These findings provide a plausible explanation for the results of the Type 2 Diabetes Evaluation of Ranolazine in Subjects With Chronic Stable Angina (TERISA) trial, which showed a greater antianginal effect of RAN in patients with coronary artery disease and diabetes.
Collapse
Affiliation(s)
| | | | | | - Erietta Kostakou
- Department of Pharmacology, University of Athens, Athens, Greece
| | | | | | | |
Collapse
|
30
|
Dostal DE, Feng H, Nizamutdinov D, Golden HB, Afroze SH, Dostal JD, Jacob JC, Foster DM, Tong C, Glaser S, Gerilechaogetu F. Mechanosensing and Regulation of Cardiac Function. ACTA ACUST UNITED AC 2014; 5:314. [PMID: 25485172 PMCID: PMC4255974 DOI: 10.4172/2155-9880.1000314] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The role of mechanical force as an important regulator of structure and function of mammalian cells, tissues, and organs has recently been recognized. However, mechanical overload is a pathogenesis or comorbidity existing in a variety of heart diseases, such as hypertension, aortic regurgitation and myocardial infarction. Physical stimuli sensed by cells are transmitted through intracellular signal transduction pathways resulting in altered physiological responses or pathological conditions. Emerging evidence from experimental studies indicate that β1-integrin and the angiotensin II type I (AT1) receptor play critical roles as mechanosensors in the regulation of heart contraction, growth and leading to heart failure. Integrin link the extracellular matrix and the intracellular cytoskeleton to initiate the mechanical signalling, whereas, the AT1 receptor could be activated by mechanical stress through an angiotensin-II-independent mechanism. Recent studies show that both Integrin and AT1 receptor and their downstream signalling factors including MAPKs, AKT, FAK, ILK and GTPase regulate heart function in cardiac myocytes. In this review we describe the role of mechanical sensors residing within the plasma membrane, mechanical sensor induced downstream signalling factors and its potential roles in cardiac contraction and growth.
Collapse
Affiliation(s)
- David E Dostal
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Hao Feng
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Damir Nizamutdinov
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Honey B Golden
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Syeda H Afroze
- Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Joseph D Dostal
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - John C Jacob
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Donald M Foster
- Central Texas Veterans Health Care System, Temple, Texas, USA
| | - Carl Tong
- Systems Biology and Translational Medicine, the Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Shannon Glaser
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Fnu Gerilechaogetu
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| |
Collapse
|
31
|
Prasad V, Lorenz JN, Lasko VM, Nieman ML, Al Moamen NJ, Shull GE. Loss of the AE3 Cl(-)/HCO(-) 3 exchanger in mice affects rate-dependent inotropy and stress-related AKT signaling in heart. Front Physiol 2013; 4:399. [PMID: 24427143 PMCID: PMC3875869 DOI: 10.3389/fphys.2013.00399] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/19/2013] [Indexed: 01/12/2023] Open
Abstract
Cl(-)/HCO(-) 3 exchangers are expressed abundantly in cardiac muscle, suggesting that HCO(-) 3 extrusion serves an important function in heart. Mice lacking Anion Exchanger Isoform 3 (AE3), a major cardiac Cl(-)/HCO(-) 3 exchanger, appear healthy, but loss of AE3 causes decompensation in a hypertrophic cardiomyopathy (HCM) model. Using intra-ventricular pressure analysis, in vivo pacing, and molecular studies we identified physiological and biochemical changes caused by loss of AE3 that may contribute to decompensation in HCM. AE3-null mice had normal cardiac contractility under basal conditions and after β-adrenergic stimulation, but pacing of hearts revealed that frequency-dependent inotropy was blunted, suggesting that AE3-mediated HCO(-) 3 extrusion is required for a robust force-frequency response (FFR) during acute biomechanical stress in vivo. Modest changes in expression of proteins that affect Ca(2+)-handling were observed, but Ca(2+)-transient analysis of AE3-null myocytes showed normal twitch-amplitude and Ca(2+)-clearance. Phosphorylation and expression of several proteins implicated in HCM and FFR, including phospholamban (PLN), myosin binding protein C, and troponin I were not altered in hearts of paced AE3-null mice; however, phosphorylation of Akt, which plays a central role in mechanosensory signaling, was significantly higher in paced AE3-null hearts than in wild-type controls and phosphorylation of AMPK, which is affected by Akt and is involved in energy metabolism and some cases of HCM, was reduced. These data show loss of AE3 leads to impaired rate-dependent inotropy, appears to affect mechanical stress-responsive signaling, and reduces activation of AMPK, which may contribute to decompensation in heart failure.
Collapse
Affiliation(s)
- Vikram Prasad
- Departments of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - John N Lorenz
- Departments of Cellular and Molecular Physiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Valerie M Lasko
- Departments of Cellular and Molecular Physiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Michelle L Nieman
- Departments of Cellular and Molecular Physiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Nabeel J Al Moamen
- Genetic Laboratory, Department of Pathology, Salmaniya Medical Complex Manama, Bahrain
| | - Gary E Shull
- Departments of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine Cincinnati, OH, USA
| |
Collapse
|
32
|
Nardosinone protects H9c2 cardiac cells from angiotensin II-induced hypertrophy. ACTA ACUST UNITED AC 2013; 33:822-826. [DOI: 10.1007/s11596-013-1205-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 09/12/2013] [Indexed: 10/25/2022]
|
33
|
Gloria-Bottini F, Banci M, Saccucci P, Nardi P, Scognamiglio M, Papetti F, Adanti S, Magrini A, Pellegrino A, Bottini E, Chiariello L. ACP1 Genetic Polymorphism and Coronary Artery Disease: Evidence of Effects on Clinical Parameters of Cardiac Function. Cardiol Res 2013; 4:101-108. [PMID: 28352429 PMCID: PMC5358247 DOI: 10.4021/cr277w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2013] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Kinases and phosphatases have an important role in the susceptibility and clinical variability of cardiac diseases. We have recently reported an association between a phosphoprotein phosphatase controlled by Acid Phosphatase locus 1 (ACP1), and Coronary artery disease (CAD) suggesting an effect on the susceptibility to this disease. In the present note we have investigated a possible role of ACP1 in the variability of clinical parameters of cardiac function. METHODS We have studied 345 subjects admitted to Valmontone Hospital for cardiovascular diseases: 202 subjects with CAD and 143 without CAD, 53 subjects admitted to Cardiac Surgery Division of Tor Vergata University were also considered. RESULTS In diabetic patients with CAD there is a significant negative association between Left ventricular ejection fraction (LVEF) and ACP1 S isoform concentration. Genotypes with high S isoform concentration show a lower value of LVEF as compared to genotypes with low S isoform concentration. We have also found a significant positive association between cNYHA class and ACP1 S isoform. After surgical intervention, in subjects with high S isoform concentration the decrease of LVEF is more marked as compared to subjects with low S isoform concentration. Overall these observations indicate that high S isoform activity has negative effects on cardiac function. The observation in patients undergoing cardiac surgery confirms the negative association between high S isoform activity and LVEF. CONCLUSIONS The present study suggests that ACP1 influences both susceptibility to CAD and clinical manifestations of the disease.
Collapse
Affiliation(s)
- Fulvia Gloria-Bottini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Maria Banci
- Department of Cardiology, Valmontone Hospital, Rome, Italy
| | - Patrizia Saccucci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Paolo Nardi
- Department of Cardiac Surgery, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Mattia Scognamiglio
- Department of Cardiac Surgery, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | | | - Sara Adanti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Andrea Magrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Antonio Pellegrino
- Department of Cardiac Surgery, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Egidio Bottini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| | - Luigi Chiariello
- Department of Cardiac Surgery, University of Rome Tor Vergata, School of Medicine, Rome, Italy
| |
Collapse
|
34
|
McLean BA, Zhabyeyev P, Pituskin E, Paterson I, Haykowsky MJ, Oudit GY. PI3K Inhibitors as Novel Cancer Therapies: Implications for Cardiovascular Medicine. J Card Fail 2013; 19:268-82. [DOI: 10.1016/j.cardfail.2013.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 01/09/2023]
|
35
|
Affiliation(s)
- Nina Mann
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Harvard/MIT Health Sciences and Technology Program, Boston, MA
| | - Anthony Rosenzweig
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
36
|
Katengua-Thamahane E, Engelbrecht AM, Esterhuyse AJ, Van Rooyen J. Inhibition of Akt Attenuates RPO-Induced Cardioprotection. Cardiol Res Pract 2012; 2012:392457. [PMID: 23082275 PMCID: PMC3469093 DOI: 10.1155/2012/392457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/11/2012] [Accepted: 08/20/2012] [Indexed: 12/23/2022] Open
Abstract
Previous studies have shown that red palm oil (RPO) supplementation protected rat hearts against ischaemia-reperfusion injury. Evidence from these studies suggested that Akt may be partly responsible for the observed protection. The aim of the current study was therefore to prove or refute the involvement of Akt in the RPO-induced cardioprotection by administration of a specific Akt inhibitor (A6730). Male Wistar rats were randomly divided into 2 groups: a control group receiving standard rat chow and an experimental group receiving standard rat chow plus 2 mL RPO for six weeks. Hearts were excised and mounted on the Langendorff perfusion system. Functional recovery was documented. A different set of hearts were freeze-clamped to assess total and phosphorylation status of Akt. Another set of hearts were subjected to the same perfusion conditions with addition of A6730. Hearts from this protocol were freeze-clamped and assessed for total and phospho-Akt. RPO improved functional recovery which was associated with increased phosphorylation of Akt on Ser473 and Thr308 residues. Blockade of Akt phosphorylation caused poor functional recovery. For the first time, these results prove that Akt plays an important role in the RPO-induced cardioprotection.
Collapse
Affiliation(s)
- Emma Katengua-Thamahane
- Experimental Anti-oxidant Research Division, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Symphony Road, Western Cape, Bellville 7535, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch 7600, South Africa
| | - Adriaan J. Esterhuyse
- Experimental Anti-oxidant Research Division, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Symphony Road, Western Cape, Bellville 7535, South Africa
| | - Jacques Van Rooyen
- Experimental Anti-oxidant Research Division, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Symphony Road, Western Cape, Bellville 7535, South Africa
| |
Collapse
|
37
|
Shiojima I, Schiekofer S, Schneider JG, Belisle K, Sato K, Andrassy M, Galasso G, Walsh K. Short-term akt activation in cardiac muscle cells improves contractile function in failing hearts. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1969-76. [PMID: 23031259 DOI: 10.1016/j.ajpath.2012.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 07/18/2012] [Accepted: 08/09/2012] [Indexed: 01/19/2023]
Abstract
Akt is a serine/threonine protein kinase that is activated by a variety of growth factors or cytokines in a phosphatidylinositol 3-kinase-dependent manner. By using a conditional transgenic system in which Akt signaling can be turned on or off in the adult heart, we previously showed that short-term Akt activation induces a physiological form of cardiac hypertrophy with enhanced coronary angiogenesis and maintained contractility. Here we tested the hypothesis that induction of physiological hypertrophy by short-term Akt activation might improve contractile function in failing hearts. When Akt signaling transiently was activated in murine hearts with impaired contractility, induced by pressure overload or doxorubicin treatment, contractile dysfunction was attenuated in both cases. Importantly, improvement of contractility was observed before the development of cardiac hypertrophy, indicating that Akt activation improves contractile function independently of its growth-promoting effects. To gain mechanistic insights into Akt-mediated positive inotropic effects, transcriptional profiles in the heart were determined in a pressure overload-induced heart failure model. Biological network analysis of differentially expressed transcripts revealed significant alterations in the expression of genes associated with cell death, and these alterations were reversed by short-term Akt activation. Thus, short-term Akt activation improves contractile function in failing hearts. This beneficial effect of Akt on contractility is hypertrophy-independent and may be mediated in part by inhibition of cell death associated with heart failure.
Collapse
Affiliation(s)
- Ichiro Shiojima
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Restoration of defective L-type Ca2+ current in cardiac myocytes of type 2 diabetic db/db mice by Akt and PKC-ι. J Cardiovasc Pharmacol 2012; 58:439-45. [PMID: 21753738 DOI: 10.1097/fjc.0b013e318228e68c] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetes is associated with an increased risk of heart failure and the development of a cardiomyopathy whose etiology is only partially understood. Ca entry through the voltage-dependent L-type Ca channel CaV1.2 initiates the contractile cycle in cardiac myocytes. Decreased cardiac contractility and depressed CaV1.2 function have been reported in obese type 2 diabetic db/db mice. Here, we demonstrate that a reduction in phosphoinositide 3-kinase (PI3K) signaling is a major contributor to the altered function of CaV1.2 in db/db cardiac myocytes. Using the whole-cell patch clamp technique, we determined that intracellular infusion of cardiac myocytes from db/db mice with phosphatidylinositol 3,4,5-trisphosphate (PIP3), the second messenger produced by PI3K, increased the L-type Ca current (ICa,L) density nearly to the level seen in wild-type cells. PIP3 also reversed the positive shift in the voltage dependence of the steady-state current activation observed in db/db myocytes. Infusion of protein kinases that act downstream of PI3K also affected ICa,L. Akt1 and Akt2 were as effective as PIP3 in restoring the ICa,L density in db/db myocytes but did not affect the voltage dependence of current activation. The infusion of atypical PKC-ι (the human homolog of mouse PKC-λ) caused a small but significant increase in the ICa,L density and completely reversed the shift in voltage dependence of steady-state current activation. These results indicate that a defect in PI3K/PIP3/Akt/PKC-λ signaling is mainly responsible for the depressed CaV1.2 function in the hearts of db/db mice with type 2 diabetes.
Collapse
|
39
|
Graves BM, Simerly T, Li C, Williams DL, Wondergem R. Phosphoinositide-3-kinase/akt - dependent signaling is required for maintenance of [Ca(2+)](i), I(Ca), and Ca(2+) transients in HL-1 cardiomyocytes. J Biomed Sci 2012; 19:59. [PMID: 22715995 PMCID: PMC3464604 DOI: 10.1186/1423-0127-19-59] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 06/20/2012] [Indexed: 01/27/2023] Open
Abstract
The phosphoinositide 3-kinases (PI3K/Akt) dependent signaling pathway plays an important role in cardiac function, specifically cardiac contractility. We have reported that sepsis decreases myocardial Akt activation, which correlates with cardiac dysfunction in sepsis. We also reported that preventing sepsis induced changes in myocardial Akt activation ameliorates cardiovascular dysfunction. In this study we investigated the role of PI3K/Akt on cardiomyocyte function by examining the role of PI3K/Akt-dependent signaling on [Ca2+]i, Ca2+ transients and membrane Ca2+ current, ICa, in cultured murine HL-1 cardiomyocytes. LY294002 (1–20 μM), a specific PI3K inhibitor, dramatically decreased HL-1 [Ca2+]i, Ca2+ transients and ICa. We also examined the effect of PI3K isoform specific inhibitors, i.e. α (PI3-kinase α inhibitor 2; 2–8 nM); β (TGX-221; 100 nM) and γ (AS-252424; 100 nM), to determine the contribution of specific isoforms to HL-1 [Ca2+]i regulation. Pharmacologic inhibition of each of the individual PI3K isoforms significantly decreased [Ca2+]i, and inhibited Ca2+ transients. Triciribine (1–20 μM), which inhibits AKT downstream of the PI3K pathway, also inhibited [Ca2+]i, and Ca2+ transients and ICa. We conclude that the PI3K/Akt pathway is required for normal maintenance of [Ca2+]i in HL-1 cardiomyocytes. Thus, myocardial PI3K/Akt-PKB signaling sustains [Ca2+]i required for excitation-contraction coupling in cardiomyoctyes.
Collapse
Affiliation(s)
- Bridget M Graves
- Departments of Surgery, James H. Quillen College of Medicine, East Tennessee State Universitycpr, Johnson City, TN 37614, USA
| | | | | | | | | |
Collapse
|
40
|
Fredersdorf S, Thumann C, Zimmermann WH, Vetter R, Graf T, Luchner A, Riegger GAJ, Schunkert H, Eschenhagen T, Weil J. Increased myocardial SERCA expression in early type 2 diabetes mellitus is insulin dependent: In vivo and in vitro data. Cardiovasc Diabetol 2012; 11:57. [PMID: 22621761 PMCID: PMC3447673 DOI: 10.1186/1475-2840-11-57] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 05/02/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Calcium (Ca2+) handling proteins are known to play a pivotal role in the pathophysiology of cardiomyopathy. However little is known about early changes in the diabetic heart and the impact of insulin treatment (Ins). METHODS Zucker Diabetic Fatty rats treated with or without insulin (ZDF ± Ins, n = 13) and lean littermates (controls, n = 7) were sacrificed at the age of 19 weeks. ZDF + Ins (n = 6) were treated with insulin for the last 6 weeks of life. Gene expression of Ca2+ ATPase in the cardiac sarcoplasmatic reticulum (SERCA2a, further abbreviated as SERCA) and phospholamban (PLB) were determined by northern blotting. Ca2+ transport of the sarcoplasmatic reticulum (SR) was assessed by oxalate-facilitated 45Ca-uptake in left ventricular homogenates. In addition, isolated neonatal cardiomyocytes were stimulated in cell culture with insulin, glucose or triiodthyronine (T3, positive control). mRNA expression of SERCA and PLB were measured by Taqman PCR. Furthermore, effects of insulin treatment on force of contraction and relaxation were evaluated by cardiomyocytes grown in a three-dimensional collagen matrix (engineered heart tissue, EHT) stimulated for 5 days by insulin. By western blot phosphorylations status of Akt was determed and the influence of wortmannin. RESULTS SERCA levels increased in both ZDF and ZDF + Ins compared to control (control 100 ± 6.2 vs. ZDF 152 ± 26.6* vs. ZDF + Ins 212 ± 18.5*# % of control, *p < 0.05 vs. control, #p < 0.05 vs. ZDF) whereas PLB was significantly decreased in ZDF and ZDF + Ins (control 100 ± 2.8 vs. ZDF 76.3 ± 13.5* vs. ZDF + Ins 79.4 ± 12.9* % of control, *p < 0.05 vs control). The increase in the SERCA/PLB ratio in ZDF and ZDF ± Ins was accompanied by enhanced Ca2+ uptake to the SR (control 1.58 ± 0.1 vs. ZDF 1.85 ± 0.06* vs. ZDF + Ins 2.03 ± 0.1* μg/mg/min, *p < 0.05 vs. control). Interestingly, there was a significant correlation between Ca2+ uptake and SERCA2a expression. As shown by in-vitro experiments, the effect of insulin on SERCA2a mRNA expression seemed to have a direct effect on cardiomyocytes. Furthermore, long-term treatment of engineered heart tissue with insulin increased the SERCA/PLB ratio and accelerated relaxation time. Akt was significantly phosphorylated by insulin. This effect could be abolished by wortmannin. CONCLUSION The current data demonstrate that early type 2 diabetes is associated with an increase in the SERCA/PLB ratio and that insulin directly stimulates SERCA expression and relaxation velocity. These results underline the important role of insulin and calcium handling proteins in the cardiac adaptation process of type 2 diabetes mellitus contributing to cardiac remodeling and show the important role of PI3-kinase-Akt-SERCA2a signaling cascade.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Northern
- Blotting, Western
- Calcium/metabolism
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Disease Models, Animal
- Gene Expression Regulation, Enzymologic/drug effects
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Male
- Myocardial Contraction/drug effects
- Myocardium/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Phosphorylation
- Polymerase Chain Reaction
- Proto-Oncogene Proteins c-akt
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Rats, Zucker
- Sarcoplasmic Reticulum/enzymology
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Time Factors
- Up-Regulation
Collapse
Affiliation(s)
- Sabine Fredersdorf
- Klinik und Poliklinik für Innere Medizin II, Universität Regensburg, Regensburg, Germany
- Klinik und Poliklinik für Innere Medizin II des Universitätsklinikums Regensburg, 93042, Regensburg, Germany
| | - Christian Thumann
- Klinik und Poliklinik für Innere Medizin II, Universität Regensburg, Regensburg, Germany
| | - Wolfram H Zimmermann
- Institut für Pharmakologie, Universitätsmedizin, Georg-August Universität Göttingen, Göttingen, Germany
| | - Roland Vetter
- Institut für Klinische Pharmakologie und Toxikologie, Universitätsmedizin - Berlin, Berlin, Germany
| | - Tobias Graf
- Medizinische Klinik II, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Andreas Luchner
- Klinik und Poliklinik für Innere Medizin II, Universität Regensburg, Regensburg, Germany
| | - Günter AJ Riegger
- Klinik und Poliklinik für Innere Medizin II, Universität Regensburg, Regensburg, Germany
| | - Heribert Schunkert
- Medizinische Klinik II, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Thomas Eschenhagen
- Institut für Klinische und Experimentelle Pharmakologie und Toxikologie, Universität Hamburg, Hamburg, Germany
| | - Joachim Weil
- Medizinische Klinik II, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
41
|
Nagoshi T, Yoshimura M, Rosano GMC, Lopaschuk GD, Mochizuki S. Optimization of cardiac metabolism in heart failure. Curr Pharm Des 2012; 17:3846-53. [PMID: 21933140 PMCID: PMC3271354 DOI: 10.2174/138161211798357773] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 09/05/2011] [Indexed: 02/06/2023]
Abstract
The derangement of the cardiac energy substrate metabolism plays a key role in the pathogenesis of heart failure. The utilization of non-carbohydrate substrates, such as fatty acids, is the predominant metabolic pathway in the normal heart, because this provides the highest energy yield per molecule of substrate metabolized. In contrast, glucose becomes an important preferential substrate for metabolism and ATP generation under specific pathological conditions, because it can provide greater efficiency in producing high energy products per oxygen consumed compared to fatty acids. Manipulations that shift energy substrate utilization away from fatty acids toward glucose can improve the cardiac function and slow the progression of heart failure. However, insulin resistance, which is highly prevalent in the heart failure population, impedes this adaptive metabolic shift. Therefore, the acceleration of the glucose metabolism, along with the restoration of insulin sensitivity, would be the ideal metabolic therapy for heart failure. This review discusses the therapeutic potential of modifying substrate utilization to optimize cardiac metabolism in heart failure.
Collapse
Affiliation(s)
- Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | | | | | | | | |
Collapse
|
42
|
Renninger JP, Murphy DJ, Morel DW. A selective Akt inhibitor produces hypotension and bradycardia in conscious rats due to inhibition of the autonomic nervous system. Toxicol Sci 2011; 125:578-85. [PMID: 22094455 DOI: 10.1093/toxsci/kfr316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Akt is a serine-threonine kinase that is amplified in a variety of human cancers, and as with other anticancer agents, some Akt inhibitors have produced functional cardiovascular effects such as marked hypotension that may limit their clinical benefit. Although identified in preclinical studies, the mechanism(s) responsible for these effects are often not fully characterized; potential targets include Akt signaling disruption in cardiac tissue, vascular smooth muscle, and/or autonomic system signaling. A selective Akt inhibitor was found to produce a rapid and marked hypotension and bradycardia in conscious rats. Isolated right atrial tissue and isolated thoracic aortic rings were used to examine direct effects of Akt inhibition on cardiac and vascular tissues, respectively. In addition, rats surgically prepared with telemetry units for monitoring blood pressure and heart rate were used to investigate potential effects on the autonomic nervous system (ANS). Whereas this Akt inhibitor did not produce any significant effect on atrial tissue, it did cause vasorelaxation of aortic rings. More significantly, in conscious rats, the Akt inhibitor inhibited the neural pressor response to the known nicotinic acetylcholine receptor (nAchR) agonist dimethylphenylpiperazinium (DMPP). In fact, the response observed was comparable to the response observed with the known ganglionic blocker hexamethonium. Thus, the hypotension and bradycardia produced by the Akt inhibitor is primarily due to blockade of nAchRs in autonomic ganglia. This finding highlights the importance of evaluating the ANS for cardiovascular effects associated with new chemical entities as well as suggesting a novel direct effect of an Akt inhibitor on nAchRs.
Collapse
Affiliation(s)
- Jonathan P Renninger
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
43
|
Li X, Mikhalkova D, Gao E, Zhang J, Myers V, Zincarelli C, Lei Y, Song J, Koch WJ, Peppel K, Cheung JY, Feldman AM, Chan TO. Myocardial injury after ischemia-reperfusion in mice deficient in Akt2 is associated with increased cardiac macrophage density. Am J Physiol Heart Circ Physiol 2011; 301:H1932-40. [PMID: 21890689 DOI: 10.1152/ajpheart.00755.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Akt2 protein kinase has been shown to promote cell migration and actin polymerization in several cell types, including macrophages. Because migrating macrophages constitute an important inflammatory response after myocardial ischemia, we determined cardiac macrophage expression after ischemia-reperfusion (I/R) injury and cryo-injury in mice lacking Akt2 (Akt2-KO). At 7 days post-I/R, Akt2-KO cardiac tissues showed an increase in immunohistochemical staining for macrophage markers (Galectin 3 and F4/80) compared with wild-type (WT) mice, indicating macrophage density was increased in the injured Akt2-KO myocardium. This change was time dependent because macrophage density was similar between WT and Akt2-KO myocardium at 3 days post-I/R, but by 7 and 14 days post-I/R, macrophage density was significantly increased in Akt2-KO myocardium. Concomitantly, infarct size was larger and cardiac function was reduced in Akt2-KO mice subjected to I/R. However, when cryo-infarction produced similar infarct sizes in the anterior wall in both WT and Akt2-KO mice, macrophage density remained higher in Akt2-KO mouse myocardium, suggesting Akt2 regulates myocardial macrophage density independent of infarct size. Consistently, bone marrow from Akt2-KO mice enhanced myocardial macrophage density in both C57/B6 WT and Akt2-KO recipient mice. Finally, reciprocal ex-vivo coculturing of macrophages and cardiac myocytes showed that activated Akt2-KO peritoneal macrophages had reduced mobility and adhesion when compared with WT littermate controls. Thus, although Akt-2 KO mice did not affect the initial inflammation response after injury and Akt2 deficiency has been shown to impair cell migration or motility in macrophages, our data suggested a novel mechanism in which increasing retention of Akt2-KO macrophages resulted in increasing cardiac Akt2-KO macrophage density in the myocardial space.
Collapse
Affiliation(s)
- Xue Li
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sussman MA, Völkers M, Fischer K, Bailey B, Cottage CT, Din S, Gude N, Avitabile D, Alvarez R, Sundararaman B, Quijada P, Mason M, Konstandin MH, Malhowski A, Cheng Z, Khan M, McGregor M. Myocardial AKT: the omnipresent nexus. Physiol Rev 2011; 91:1023-70. [PMID: 21742795 PMCID: PMC3674828 DOI: 10.1152/physrev.00024.2010] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.
Collapse
Affiliation(s)
- Mark A Sussman
- Department of Biology, San Diego State University, SDSU Heart Institute, San Diego, California 92182, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nelson MJ, Harris MB, Boluyt MO, Hwang HS, Starnes JW. Effect of N-2-mercaptopropionyl glycine on exercise-induced cardiac adaptations. Am J Physiol Regul Integr Comp Physiol 2011; 300:R993-R1000. [DOI: 10.1152/ajpregu.00405.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to test the hypothesis that exercise-induced cardiac adaptations would be attenuated by the free radical scavenger N-2-mercaptopropionyl glycine (MPG). Male Sprague-Dawley rats were divided into four groups ( n = 9–13 per group) for 3–4 wk: sedentary (S), S+MPG (100 mg/kg ip daily), exercised on a treadmill (E) (60 min/day, 5 days/wk, at a speed of 20 m/min up a 6° grade in a 6°C room), or E+MPG given 10 min prior to exercise. Additional rats ( n = 55) were used to determine acute exercise effects on myocardial redox state [nonprotein nonglutathione sulfhydryls (NPNGSH)] and PI3K/Akt signaling pathway activation. Compared with S, NPNGSH levels were 48% lower in E ( P < 0.05) and unchanged in E+MPG ( P > 0.05). MPG also attenuated exercise-induced activation of the signaling proteins Akt and S6. Hearts from the 4-wk groups were weighed, and cardiac function was evaluated using an isolated perfused working heart preparation. Similar increases ( P < 0.05) in both exercised groups were observed for heart weight and heart weight-to-body weight ratio. Cardiac function improved in E vs. S, as indicated by greater ( P < 0.05) external work performed (cardiac output × systolic pressure) and efficiency of external work (work/V̇o2). MPG prevented these exercise-induced functional improvements. Skeletal muscle mitochondria content increased to similar levels in E and E+MPG. This study provides evidence that free radicals do not play an essential role in the development of exercise-induced cardiac hypertrophy; however, they appear to be involved in functional cardiac adaptations, which may be mediated through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Matthew J. Nelson
- Department of Kinesiology and Health Education, University of Texas, Austin Texas
| | - M. Brennan Harris
- Department of Kinesiology and Health Education, University of Texas, Austin Texas
| | - Marvin O. Boluyt
- Center for Exercise Research, Division of Kinesiology, University of Michigan, Ann Arbor, Michigan; and
| | - Hyun Seok Hwang
- Center for Exercise Research, Division of Kinesiology, University of Michigan, Ann Arbor, Michigan; and
| | - Joseph W. Starnes
- Department of Kinesiology and Health Education, University of Texas, Austin Texas
- Department of Kinesiology, University of North Carolina, Greensboro, North Carolina
| |
Collapse
|
46
|
Fang F, Li D, Pan H, Chen D, Qi L, Zhang R, Sun H. Luteolin Inhibits Apoptosis and Improves Cardiomyocyte Contractile Function through the PI3K/Akt Pathway in Simulated Ischemia/Reperfusion. Pharmacology 2011; 88:149-58. [DOI: 10.1159/000330068] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/23/2011] [Indexed: 11/19/2022]
|
47
|
Hong HM, Song EJ, Oh E, Kabir MH, Lee C, Yoo YS. Endothelin-1- and isoproterenol-induced differential protein expression and signaling pathway in HL-1 cardiomyocytes. Proteomics 2010; 11:283-97. [PMID: 21204255 DOI: 10.1002/pmic.201000018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 08/13/2010] [Accepted: 10/20/2010] [Indexed: 11/07/2022]
Abstract
It is well known that the two chemical compounds endothelin-1 (ET-1) and isoproterenol (ISO) can individually induce cardiac hypertrophy through G protein-coupled receptors in cardiomyocytes. However, the cardiac hypertrophy signaling pathway activated by ET-1 and ISO is not well defined. Therefore, we investigated the protein expression profile and signaling transduction in HL-l cardiomyocyte cells treated with ET-1 and ISO. Following separation of the cell lysates by using 2-DE and silver staining, we identified 16 protein spots that were differentially expressed as compared to the controls. Of these 16 spots, three changed only after treatment with ET-1, whereas four changed only after treatment with ISO, suggesting that these two stimuli could induce different signaling pathways. In order to reveal the differences between ET-1- and ISO-induced signaling, we studied the different events that occur at each step of the signaling pathways, when selected biocomponents were blocked by inhibitors. Our results indicated that ET-1 and ISO used different pathways for phosphorylation of glycogen synthase kinase-3β (GSK3β). ET-1 mainly used the mitogen-activated protein kinase and phosphatidylinositol-3-kinase/AKT pathways to activate GSK3β, whereas under ISO stimulation, only the phosphatidylinositol-3-kinase/AKT pathway was required to trigger the GSK3β pathway. Furthermore, the strength of the GSK3β signal in ISO-induced cardiac hypertrophy was stronger than that in ET-1-induced cardiac hypertrophy. We found that these two agonists brought about different changes in the protein expression of HL-1 cardiomyocytes through distinct signaling pathways even though the destination of the two signaling pathways was the same.
Collapse
Affiliation(s)
- Hye-Min Hong
- Integrated Omics Center, Life/Health Division Korea Institute of Science and Technology, Cheongryang, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
48
|
Wan X, Dennis AT, Obejero-Paz C, Overholt JL, Heredia-Moya J, Kirk KL, Ficker E. Oxidative inactivation of the lipid phosphatase phosphatase and tensin homolog on chromosome ten (PTEN) as a novel mechanism of acquired long QT syndrome. J Biol Chem 2010; 286:2843-52. [PMID: 21097842 DOI: 10.1074/jbc.m110.125526] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The most common cause of cardiac side effects of pharmaco-therapy is acquired long QT syndrome, which is characterized by abnormal cardiac repolarization and most often caused by direct blockade of the cardiac potassium channel human ether a-go-go-related gene (hERG). However, little is known about therapeutic compounds that target ion channels other than hERG. We have discovered that arsenic trioxide (As(2)O(3)), a very potent antineoplastic compound for the treatment of acute promyelocytic leukemia, is proarrhythmic via two separate mechanisms: a well characterized inhibition of hERG/I(Kr) trafficking and a poorly understood increase of cardiac calcium currents. We have analyzed the latter mechanism in the present study using biochemical and electrophysiological methods. We find that oxidative inactivation of the lipid phosphatase PTEN by As(2)O(3) enhances cardiac calcium currents in the therapeutic concentration range via a PI3Kα-dependent increase in phosphatidylinositol 3,4,5-triphosphate (PIP(3)) production. In guinea pig ventricular myocytes, even a modest reduction in PTEN activity is sufficient to increase cellular PIP(3) levels. Under control conditions, PIP(3) levels are kept low by PTEN and do not affect calcium current amplitudes. Based on pharmacological experiments and intracellular infusion of PIP(3), we propose that in guinea pig ventricular myocytes, PIP(3) regulates calcium currents independently of the protein kinase Akt along a pathway that includes a secondary oxidation-sensitive target. Overall, our report describes a novel form of acquired long QT syndrome where the target modified by As(2)O(3) is an intracellular signaling cascade.
Collapse
Affiliation(s)
- Xiaoping Wan
- Rammelkamp Center for Education and Research, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio 44109, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Semple D, Smith K, Bhandari S, Seymour AML. Uremic cardiomyopathy and insulin resistance: a critical role for akt? J Am Soc Nephrol 2010; 22:207-15. [PMID: 20634295 DOI: 10.1681/asn.2009090900] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Uremic cardiomyopathy is a classic complication of chronic renal failure whose cause is unclear and treatment remains disappointing. Insulin resistance is an independent predictor of cardiovascular mortality in chronic renal failure. Underlying insulin resistance are defects in insulin signaling through the protein kinase, Akt. Akt acts as a nodal point in the control of both the metabolic and pleiotropic effects of insulin. Imbalance among these effects leads to cardiac hypertrophy, fibrosis, and apoptosis; less angiogenesis; metabolic remodeling; and altered calcium cycling, all key features of uremic cardiomyopathy. Here we consider the role of Akt in the development of uremic cardiomyopathy, drawing parallels from models of hypertrophic cardiac disease.
Collapse
Affiliation(s)
- David Semple
- Department of Biological Sciences, University of Hull, Kingston-upon-Hull, HU6 7RX, UK
| | | | | | | |
Collapse
|
50
|
Abstract
Signal transduction events are key modulators of cellular function and, in the cardiovascular system, an emerging role is played by phosphoinositide 3-kinases (PI3Ks), a family of enzymes containing a 3-phosphorylated phosphoinositide that produce lipid second messengers. In the heart, multiple PI3K isoforms are expressed, but play potentially distinct roles. Among cardiac PI3Ks, PI3Kalpha is triggered by tyrosine kinase receptors and plays a role in adaptive hypertrophy, while PI3Kgamma is triggered by G protein-coupled receptors and is involved in maladaptive heart remodeling. This view has been recently complicated by the finding that PI3Ks can also be involved in protein-protein interactions and affect signaling independently of their kinase activity. This review will thus focus on the effects of these multiple signaling events, with particular emphasis on their involvement in cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Federico Damilano
- Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy
| | | | | |
Collapse
|