1
|
Devilée LA, Miller JM, Reid JD, Salama ABM, Ou Q, Jamal M, Nong Y, Andres D, Satin J, Mohamed TMA, Hudson JE, Abouleisa RRE. Pharmacological or genetic inhibition of LTCC promotes cardiomyocyte proliferation through inhibition of calcineurin activity. RESEARCH SQUARE 2023:rs.3.rs-3552794. [PMID: 38076903 PMCID: PMC10705701 DOI: 10.21203/rs.3.rs-3552794/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Cardiomyocytes (CMs) lost during ischemic cardiac injury cannot be replaced due to their limited proliferative capacity, which leads to progressive heart failure. Calcium (Ca2+) is an important signal transducer that regulates key cellular processes, but its role in regulating CM proliferation is incompletely understood. A drug screen targeting proteins involved in CM calcium cycling in human embryonic stem cell-derived cardiac organoids (hCOs) revealed that only the inhibition of L-Type Calcium Channel (LTCC), but not other Ca2+ regulatory proteins (SERCA or RYR), induced the CM cell cycle. Furthermore, overexpression of Ras-related associated with Diabetes (RRAD), an endogenous inhibitor of LTCC, induced CM cell cycle activity in vitro, in human cardiac slices, and in vivo. Mechanistically, LTCC inhibition by RRAD induces the cell cycle in CMs by modulating calcineurin activity and translocating Hoxb13 to the CM nucleus. Together, this represents a robust pathway for regenerative strategies.
Collapse
Affiliation(s)
- Lynn A.C. Devilée
- QIMR Berghofer Medical Research Institute, Cardiac Bioengineering Laboratory, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jessica M. Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Surgery Department, Baylor College of Medicine, Houston, TX, U.S.A
| | - Janice D. Reid
- QIMR Berghofer Medical Research Institute, Cardiac Bioengineering Laboratory, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Abou Bakr M. Salama
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Surgery Department, Baylor College of Medicine, Houston, TX, U.S.A
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Madiha Jamal
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Yibing Nong
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, U.S.A
| | - Douglas Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Jonathan Satin
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Tamer M. A. Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Surgery Department, Baylor College of Medicine, Houston, TX, U.S.A
| | - James E. Hudson
- QIMR Berghofer Medical Research Institute, Cardiac Bioengineering Laboratory, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Riham R. E. Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Surgery Department, Baylor College of Medicine, Houston, TX, U.S.A
| |
Collapse
|
2
|
Liao X, Han Y, He Y, Liu J, Wang Y. Natural compounds targeting mitochondrial dysfunction: emerging therapeutics for target organ damage in hypertension. Front Pharmacol 2023; 14:1209890. [PMID: 37397478 PMCID: PMC10311420 DOI: 10.3389/fphar.2023.1209890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/08/2023] [Indexed: 07/04/2023] Open
Abstract
Hypertension generally causes target organ damage (TOD) in the heart, brain, kidney, and blood vessels. This can result in atherosclerosis, plaque formation, cardiovascular and cerebrovascular events, and renal failure. Recent studies have indicated that mitochondrial dysfunction is crucial in hypertensive target organ damage. Consequently, mitochondria-targeted therapies attract increasing attention. Natural compounds are valuable resources for drug discovery and development. Many studies have demonstrated that natural compounds can ameliorate mitochondrial dysfunction in hypertensive target organ damage. This review examines the contribution of mitochondrial dysfunction to the development of target organ damage in hypertension. Moreover, it summarizes therapeutic strategies based on natural compounds that target mitochondrial dysfunction, which may be beneficial for preventing and treating hypertensive target organ damage.
Collapse
Affiliation(s)
- Xiaolin Liao
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuanshan Han
- Scientific Research Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying He
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jianjun Liu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Maccari S, Buoncervello M, Ascione B, Stati T, Macchia D, Fidanza S, Catalano L, Matarrese P, Gabriele L, Marano G. α-adrenoceptor stimulation attenuates melanoma growth in mice. Br J Pharmacol 2021; 179:1371-1383. [PMID: 34766341 DOI: 10.1111/bph.15731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 09/27/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Recently, β-adrenoceptor blockade has emerged as a potential strategy to inhibit melanoma growth. However, it remains to be ascertained whether β-adrenoceptor stimulation by circulating catecholamines increases melanoma growth in mice. EXPERIMENTAL APPROACH B16F10 melanoma-bearing mice were used to evaluate effects of adrenaline and specific adrenoceptor (AR) ligands on tumor volume. AR expression as well as effects of AR ligands on cell viability, production of mitochondrial reactive oxygen species (mROS) and proliferation activity in B16F10 cells were determined by biochemical analyses. KEY RESULTS qPCR analyses revealed that B16F10 cells express both α- (α1B-, α2A- and α2B-AR) and β-ARs (β2 -AR). We found that treatment with the α- and β-AR agonist adrenaline or with the synthetic catecholamine isoprenaline, that selectively stimulates β-ARs, did not affect melanoma growth. Conversely, adrenaline reduced tumor growth in mice co-treated with propranolol, a β1β2-AR antagonist. Adrenaline had no effect in tumor-bearing β1β2-AR knockout mice, in which β1- and β2-ARs are lacking, but it reduced tumor growth when co-administered with propranolol suggesting that tumor β2-ARs negatively regulate adrenaline antitumor activity. Additionally, we found that α1-AR stimulation with cirazoline yielded a decrease in B16F10 melanoma size. These effects on melanoma growth were paralleled by reduced cell viability and proliferation activity as well as increased mROS production in α1-AR-stimulated B16F10 cells. Decreased viability, proliferation and mitochondrial function in B16F10 cells also occurred after α2-AR stimulation by α2-AR agonist ST-91. CONCLUSIONS AND IMPLICATIONS In B16F10 melanoma model, stimulation of α-AR subtypes yields in vivo and in vitro anticancer activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Stefano Fidanza
- Center for animal experimentation and well-being, National Institute of Health, Rome, Italy
| | | | | | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine
| | | |
Collapse
|
4
|
Fang T, Shang W, Liu C, Liu Y, Ye A. Single-Cell Multimodal Analytical Approach by Integrating Raman Optical Tweezers and RNA Sequencing. Anal Chem 2020; 92:10433-10441. [PMID: 32643364 DOI: 10.1021/acs.analchem.0c00912] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Single-cell analysis has become a state-of-art approach to heterogeneity profiling in tumor cells. Herein, we realize a kind of single-cell multimodal analytical approach by combining single-cell RNA sequencing (scRNA-seq) with Raman optical tweezers (ROT), a label-free single-cell identification and isolation technique, and apply it to investigate drug sensitivity. The drug sensitivity of human BGC823 gastric cancer cells toward different drugs, paclitaxel and sodium dichloroacetate, was distinguished in the conjoint analytical way including morphology monitoring, Raman identification, and transcriptomic profiling. Each individual BGC823 cancer cell was measured by Raman spectroscopy, then nondestructively isolated out by ROT, and finally RNA-sequenced. Our results demonstrate each analytical mode can reflect cell response to the drugs from different perspectives and is consistent and complementary with each other. Therefore, we believe the multimodal analytical approach offers an access to comprehensive characterizations of the unicellular complexity, which especially makes sense for studying tumor heterogeneity or a desired special cell from a mixture cell sample such as whole blood.
Collapse
|
5
|
Wang M, Shi Y, Yao L, Li Q, Wang Y, Fu D. Potential Molecular Mechanisms and Drugs for Aconitine-Induced Cardiotoxicity in Zebrafish through RNA Sequencing and Bioinformatics Analysis. Med Sci Monit 2020; 26:e924092. [PMID: 32598336 PMCID: PMC7341694 DOI: 10.12659/msm.924092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Accumulating evidence suggests that cardiotoxicity is one of the main manifestations of aconitine (AC) poisoning. However, the molecular mechanism of AC-induced cardiotoxicity remains unclear, there is little direct evidence for therapeutic targets and drugs of AC-induced cardiotoxicity. Material/Methods Zebrafish were exposed to AC to evaluate cardiotoxicity by calculating the heart rates and observing the changes of cardiac and vascular structure. RNA-seq (RNA sequencing) and bioinformatics analysis were used to obtain differentially expressed genes (DEGs). The anti-AC cardiotoxicity compound was identified via connectivity map (CMAP) analysis and molecular docking. Results AC-induced cardiotoxicity in zebrafish predominantly included arrhythmias, extended sinus venous and bulbus arteriosus (SV-BA) distance, and larger pericardial edema aera. A total of 1380 DEGs were identified by RNA-seq and bioinformatics analysis. cyclin-dependent kinase-1 (CDK1) was screened as the hub gene and the most potential therapeutic target due to its significant downregulation in cardiotoxicity based on protein-protein interaction (PPI) and drug-gene interaction (DGIdb) network analysis. Cell cycle signal pathway was the most significant pathways identified in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Furthermore, the expression of CDK1 was validated in the Gene Expression Omnibus (GEO) database GSE71906, GSE65705, and GSE95140. Finally, heptaminol was identified as a novel anti-AC cardiotoxicity compound via CMAP analysis and molecular docking. Conclusions Totally, hub genes and key pathways identified in this study can aid in the understanding of the molecular changes in AC-induced cardiotoxicity. Meanwhile, we provide a systematic method to explore drug toxicity prevention and treatment.
Collapse
Affiliation(s)
- Mingzhu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Yanan Shi
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Lei Yao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Qiang Li
- Children's Hospital of Fudan University, Shanghai, China (mainland)
| | - Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| | - Deyu Fu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China (mainland)
| |
Collapse
|
6
|
Maccari S, Pace V, Barbagallo F, Stati T, Ambrosio C, Grò MC, Molinari P, Vezzi V, Catalano L, Matarrese P, Patrizio M, Rizzi R, Marano G. Intermittent β-adrenergic blockade downregulates the gene expression of β-myosin heavy chain in the mouse heart. Eur J Pharmacol 2020; 882:173287. [PMID: 32585157 DOI: 10.1016/j.ejphar.2020.173287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/14/2023]
Abstract
Expression of the β-myosin heavy chain (β-MHC), a major component of the cardiac contractile apparatus, is tightly regulated as even modest increases can be detrimental to heart under stress. In healthy hearts, continuous inhibition of β-adrenergic tone upregulates β-MHC expression. However, it is unknown whether the duration of the β-adrenergic inhibition and β-MHC expression are related. Here, we evaluated the effects of intermittent β-blockade on cardiac β-MHC expression. To this end, the β-blocker propranolol, at the dose of 15mg/kg, was administered once a day in mice for 14 days. This dosing schedule caused daily drug-free periods of at least 6 h as evidenced by propranolol plasma concentrations and cardiac β-adrenergic responsiveness. Under these conditions, β-MHC expression decreased by about 75% compared to controls. This effect was abolished in mice lacking β1- but not β2-adrenergic receptors (β-AR) indicating that β-MHC expression is regulated in a β1-AR-dependent manner. In β1-AR knockout mice, the baseline β-MHC expression was fourfold higher than in wild-type mice. Also, we evaluated the impact of intermittent β-blockade on β-MHC expression in mice with systolic dysfunction, in which an increased β-MHC expression occurs. At 3 weeks after myocardial infarction, mice showed systolic dysfunction and upregulation of β-MHC expression. Intermittent β-blockade decreased β-MHC expression while attenuating cardiac dysfunction. In vitro studies showed that propranolol does not affect β-MHC expression on its own but antagonizes catecholamine effects on β-MHC expression. In conclusion, a direct relationship occurs between the duration of the β-adrenergic inhibition and β-MHC expression through the β1-AR.
Collapse
Affiliation(s)
- Sonia Maccari
- Center for Gender-Specific Medicine, National Institute of Health, Rome, Italy
| | - Valentina Pace
- Institute of Biochemistry and Cellular Biology, National Council of Research, Monterotondo (RM), Italy
| | | | - Tonino Stati
- Center for Gender-Specific Medicine, National Institute of Health, Rome, Italy
| | - Caterina Ambrosio
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | - Maria Cristina Grò
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | - Paola Molinari
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | - Vanessa Vezzi
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, Rome, Italy
| | - Mario Patrizio
- Center for Gender-Specific Medicine, National Institute of Health, Rome, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Institute for Biomedical Technologies, National Council of Research, Milan, Italy
| | - Giuseppe Marano
- Center for Gender-Specific Medicine, National Institute of Health, Rome, Italy.
| |
Collapse
|
7
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
8
|
Finan A, Richard S. Stimulating endogenous cardiac repair. Front Cell Dev Biol 2015; 3:57. [PMID: 26484341 PMCID: PMC4586501 DOI: 10.3389/fcell.2015.00057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 09/08/2015] [Indexed: 01/10/2023] Open
Abstract
The healthy adult heart has a low turnover of cardiac myocytes. The renewal capacity, however, is augmented after cardiac injury. Participants in cardiac regeneration include cardiac myocytes themselves, cardiac progenitor cells, and peripheral stem cells, particularly from the bone marrow compartment. Cardiac progenitor cells and bone marrow stem cells are augmented after cardiac injury, migrate to the myocardium, and support regeneration. Depletion studies of these populations have demonstrated their necessary role in cardiac repair. However, the potential of these cells to completely regenerate the heart is limited. Efforts are now being focused on ways to augment these natural pathways to improve cardiac healing, primarily after ischemic injury but in other cardiac pathologies as well. Cell and gene therapy or pharmacological interventions are proposed mechanisms. Cell therapy has demonstrated modest results and has passed into clinical trials. However, the beneficial effects of cell therapy have primarily been their ability to produce paracrine effects on the cardiac tissue and recruit endogenous stem cell populations as opposed to direct cardiac regeneration. Gene therapy efforts have focused on prolonging or reactivating natural signaling pathways. Positive results have been demonstrated to activate the endogenous stem cell populations and are currently being tested in clinical trials. A potential new avenue may be to refine pharmacological treatments that are currently in place in the clinic. Evidence is mounting that drugs such as statins or beta blockers may alter endogenous stem cell activity. Understanding the effects of these drugs on stem cell repair while keeping in mind their primary function may strike a balance in myocardial healing. To maximize endogenous cardiac regeneration, a combination of these approaches could ameliorate the overall repair process to incorporate the participation of multiple cellular players.
Collapse
Affiliation(s)
- Amanda Finan
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| | - Sylvain Richard
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| |
Collapse
|
9
|
Patrizio M, Musumeci M, Piccone A, Raggi C, Mattei E, Marano G. Hormonal regulation of β-myosin heavy chain expression in the mouse left ventricle. J Endocrinol 2013. [PMID: 23179080 DOI: 10.1530/joe-12-0201] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We investigated the influence of sex hormones on the expression of α- and β-cardiac myosin heavy chain isoforms (α-MHC and β-MHC) in C57bl/6 mice of both sexes under physiological and pathological conditions. In the left ventricles (LVs) of fertile female mice, β-MHC expression was tenfold higher compared with the age-matched males, whereas no difference was found in α-MHC expression. These differences disappeared after ovariectomy or in immature mice. We also found a sex-related difference in expression of β-adrenoceptors (β1-AR), as mRNA levels of this gene were 40% lower in fertile females compared with males of the same age but did not differ in prepubertal or ovariectomized animals. Interestingly, the deletion of both β1- and β2-ARs abolished sex difference of β-MHC expression, as mRNA levels in the LVs of knockout males were increased and reached values comparable to those of knockout females. Moreover, the β1-AR antagonist metoprolol induced about a threefold increase in β-MHC expression in adult male mice. The capability of gender to regulate β-MHC expression was also evaluated in the presence of hemodynamic overload. Thoracic aortic coarctation (TAC) produced cardiac hypertrophy along with a 12-fold increase in β-MHC and a 50% decrease in β1-AR expression in males but not in females, thus abolishing the gender difference observed in sham animals for such genes. By contrast, TAC did not change β2-AR expression. In conclusion, our results show that the expression of β-MHC and β1-AR in the LVs undergo gender-related and correlated changes under both physiological and pathological conditions and suggest a role of β1-AR-mediated signaling.
Collapse
Affiliation(s)
- Mario Patrizio
- Department of Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
10
|
Khan M, Mohsin S, Avitabile D, Siddiqi S, Nguyen J, Wallach K, Quijada P, McGregor M, Gude N, Alvarez R, Tilley DG, Koch WJ, Sussman MA. β-Adrenergic regulation of cardiac progenitor cell death versus survival and proliferation. Circ Res 2012; 112:476-86. [PMID: 23243208 DOI: 10.1161/circresaha.112.280735] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE Short-term β-adrenergic stimulation promotes contractility in response to stress but is ultimately detrimental in the failing heart because of accrual of cardiomyocyte death. Endogenous cardiac progenitor cell (CPC) activation may partially offset cardiomyocyte losses, but consequences of long-term β-adrenergic drive on CPC survival and proliferation are unknown. OBJECTIVE We sought to determine the relationship between β-adrenergic activity and regulation of CPC function. METHODS AND RESULTS Mouse and human CPCs express only β2 adrenergic receptor (β2-AR) in conjunction with stem cell marker c-kit. Activation of β2-AR signaling promotes proliferation associated with increased AKT, extracellular signal-regulated kinase 1/2, and endothelial NO synthase phosphorylation, upregulation of cyclin D1, and decreased levels of G protein-coupled receptor kinase 2. Conversely, silencing of β2-AR expression or treatment with β2-antagonist ICI 118, 551 impairs CPC proliferation and survival. β1-AR expression in CPC is induced by differentiation stimuli, sensitizing CPC to isoproterenol-induced cell death that is abrogated by metoprolol. Efficacy of β1-AR blockade by metoprolol to increase CPC survival and proliferation was confirmed in vivo by adoptive transfer of CPC into failing mouse myocardium. CONCLUSIONS β-adrenergic stimulation promotes expansion and survival of CPCs through β2-AR, but acquisition of β1-AR on commitment to the myocyte lineage results in loss of CPCs and early myocyte precursors.
Collapse
Affiliation(s)
- Mohsin Khan
- San Diego Heart Research Institute, San Diego State University, San Diego, CA 92182, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Stiles J, Amaya C, Pham R, Rowntree RK, Lacaze M, Mulne A, Bischoff J, Kokta V, Boucheron LE, Mitchell DC, Bryan BA. Propranolol treatment of infantile hemangioma endothelial cells: A molecular analysis. Exp Ther Med 2012; 4:594-604. [PMID: 23170111 PMCID: PMC3501380 DOI: 10.3892/etm.2012.654] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/27/2012] [Indexed: 12/20/2022] Open
Abstract
Infantile hemangiomas (IHs) are non-malignant, largely cutaneous vascular tumors affecting approximately 5-10% of children to varying degrees. During the first year of life, these tumors are strongly proliferative, reaching an average size ranging from 2 to 20 cm. These lesions subsequently stabilize, undergo a spontaneous slow involution and are fully regressed by 5 to 10 years of age. Systemic treatment of infants with the non-selective β-adrenergic receptor blocker, propranolol, has demonstrated remarkable efficacy in reducing the size and appearance of IHs. However, the mechanism by which this occurs is largely unknown. In this study, we sought to understand the molecular mechanisms underlying the effectiveness of β blocker treatment in IHs. Our data reveal that propranolol treatment of IH endothelial cells, as well as a panel of normal primary endothelial cells, blocks endothelial cell proliferation, migration, and formation of the actin cytoskeleton coincident with alterations in vascular endothelial growth factor receptor-2 (VEGFR-2), p38 and cofilin signaling. Moreover, propranolol induces major alterations in the protein levels of key cyclins and cyclin-dependent kinase inhibitors, and modulates global gene expression patterns with a particular affect on genes involved in lipid/sterol metabolism, cell cycle regulation, angiogenesis and ubiquitination. Interestingly, the effects of propranolol were endothelial cell-type independent, affecting the properties of IH endothelial cells at similar levels to that observed in neonatal dermal microvascular and coronary artery endothelial cells. This data suggests that while propranolol markedly inhibits hemangioma and normal endothelial cell function, its lack of endothelial cell specificity hints that the efficacy of this drug in the treatment of IHs may be more complex than simply blockage of endothelial function as previously believed.
Collapse
Affiliation(s)
- Jessica Stiles
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|