1
|
Adepu KK, Anishkin A, Adams SH, Chintapalli SV. A versatile delivery vehicle for cellular oxygen and fuels or metabolic sensor? A review and perspective on the functions of myoglobin. Physiol Rev 2024; 104:1611-1642. [PMID: 38696337 PMCID: PMC11495214 DOI: 10.1152/physrev.00031.2023] [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: 07/28/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/04/2024] Open
Abstract
A canonical view of the primary physiological function of myoglobin (Mb) is that it is an oxygen (O2) storage protein supporting mitochondrial oxidative phosphorylation, especially as the tissue O2 partial pressure (Po2) drops and Mb off-loads O2. Besides O2 storage/transport, recent findings support functions for Mb in lipid trafficking and sequestration, interacting with cellular glycolytic metabolites such as lactate (LAC) and pyruvate (PYR), and "ectopic" expression in some types of cancer cells and in brown adipose tissue (BAT). Data from Mb knockout (Mb-/-) mice and biochemical models suggest additional metabolic roles for Mb, especially regulation of nitric oxide (NO) pools, modulation of BAT bioenergetics, thermogenesis, and lipid storage phenotypes. From these and other findings in the literature over many decades, Mb's function is not confined to delivering O2 in support of oxidative phosphorylation but may serve as an O2 sensor that modulates intracellular Po2- and NO-responsive molecular signaling pathways. This paradigm reflects a fundamental change in how oxidative metabolism and cell regulation are viewed in Mb-expressing cells such as skeletal muscle, heart, brown adipocytes, and select cancer cells. Here, we review historic and emerging views related to the physiological roles for Mb and present working models illustrating the possible importance of interactions between Mb, gases, and small-molecule metabolites in regulation of cell signaling and bioenergetics.
Collapse
Affiliation(s)
- Kiran Kumar Adepu
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, Maryland, United States
| | - Sean H Adams
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, California, United States
- Center for Alimentary and Metabolic Science, School of Medicine, University of California Davis, Sacramento, California, United States
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| |
Collapse
|
2
|
Perez-Bonilla P, LaViolette B, Bhandary B, Ullas S, Chen X, Hirenallur-Shanthappa D. Isoproterenol induced cardiac hypertrophy: A comparison of three doses and two delivery methods in C57BL/6J mice. PLoS One 2024; 19:e0307467. [PMID: 39038017 PMCID: PMC11262646 DOI: 10.1371/journal.pone.0307467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Heart Failure (HF) continues to be a complex public health issue with increasing world population prevalence. Although overall mortality has decreased for HF and hypertrophic cardiomyopathy (HCM), a precursor for HF, their prevalence continues to increase annually. Because the etiology of HF and HCM is heterogeneous, it has been difficult to identify novel therapies to combat these diseases. Isoproterenol (ISP), a non-selective β-adrenoreceptor agonist, is commonly used to induce cardiotoxicity and cause acute and chronic HCM and HF in mice. However, the variability in dose and duration of ISP treatment used in studies has made it difficult to determine the optimal combination of ISP dose and delivery method to develop a reliable ISP-induced mouse model for disease. Here we examined cardiac effects induced by ISP via subcutaneous (SQ) and SQ-minipump (SMP) infusions across 3 doses (2, 4, and 10mg/kg/day) over 2 weeks to determine whether SQ and SMP ISP delivery induced comparable disease severity in C57BL/6J mice. To assess disease, we measured body and heart weight, surface electrocardiogram (ECG), and echocardiography recordings. We found all 3 ISP doses comparably increase heart weight, but these increases are more pronounced when ISP was administered via SMP. We also found that the combination of ISP treatment and delivery method induces contrasting heart rate, RR interval, and R and S amplitudes that may place SMP treated mice at higher risk for sustained disease burden. Mice treated via SMP also had increased heart wall thickness and LV Mass, but mice treated via SQ showed greater increase in gene markers for hypertrophy and fibrosis. Overall, these data suggest that at 2 weeks, mice treated with 2, 4, or 10mg/kg/day ISP via SQ and SMP routes cause similar pathological heart phenotypes but highlight the importance of drug delivery method to induce differing disease pathways.
Collapse
Affiliation(s)
- Patricia Perez-Bonilla
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Brianna LaViolette
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Bidur Bhandary
- Rare Diseases Research Unit, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Soumya Ullas
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Xian Chen
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Dinesh Hirenallur-Shanthappa
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| |
Collapse
|
3
|
Rao K, Rochon E, Singh A, Jagannathan R, Peng Z, Mansoor H, Wang B, Moulik M, Zhang M, Saraf A, Corti P, Shiva S. Myoglobin modulates the Hippo pathway to promote cardiomyocyte differentiation. iScience 2024; 27:109146. [PMID: 38414852 PMCID: PMC10897895 DOI: 10.1016/j.isci.2024.109146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 09/30/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
The endogenous mechanisms that propagate cardiomyocyte differentiation and prevent de-differentiation remain unclear. While the expression of the heme protein myoglobin increases by over 50% during cardiomyocyte differentiation, a role for myoglobin in regulating cardiomyocyte differentiation has not been tested. Here, we show that deletion of myoglobin in cardiomyocyte models decreases the gene expression of differentiation markers and stimulates cellular proliferation, consistent with cardiomyocyte de-differentiation. Mechanistically, the heme prosthetic group of myoglobin catalyzes the oxidation of the Hippo pathway kinase LATS1, resulting in phosphorylation and inactivation of yes-associated protein (YAP). In vivo, myoglobin-deficient zebrafish hearts show YAP dephosphorylation and accelerated cardiac regeneration after apical injury. Similarly, myoglobin knockdown in neonatal murine hearts shows increased YAP dephosphorylation and cardiomyocyte cycling. These data demonstrate a novel role for myoglobin as an endogenous driver of cardiomyocyte differentiation and highlight myoglobin as a potential target to enhance cardiac development and improve cardiac repair and regeneration.
Collapse
Affiliation(s)
- Krithika Rao
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Elizabeth Rochon
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anuradha Singh
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rajaganapathi Jagannathan
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zishan Peng
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Haris Mansoor
- Heart and Vascular Institute Division of Cardiology, Department of Medicine and Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bing Wang
- Molecular Therapy Lab, Stem Cell Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mousumi Moulik
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Manling Zhang
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Veteran Affair Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anita Saraf
- Heart and Vascular Institute Division of Cardiology, Department of Medicine and Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Paola Corti
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sruti Shiva
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
4
|
Simonides W, Tijsma A, Boelen A, Jongejan R, de Rijke Y, Peeters R, Dentice M, Salvatore D, Muller A. Divergent Thyroid Hormone Levels in Plasma and Left Ventricle of the Heart in Compensated and Decompensated Cardiac Hypertrophy Induced by Chronic Adrenergic Stimulation in Mice. Metabolites 2023; 13:metabo13020308. [PMID: 36837927 PMCID: PMC9960204 DOI: 10.3390/metabo13020308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/05/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Chronic hemodynamic overload of the heart induces ventricular hypertrophy that may be either compensatory or progress to decompensation and heart failure. The gradual impairment of ventricular function is, at least in part, the result of a reduction of cardiac thyroid-hormone (TH) action. Here, we examined the proposed roles of increased cardiac expression of the TH-inactivating enzyme deiodinase type 3 (D3) and reduced plasma TH levels in diminishing cardiac TH levels. Using minipumps, mice were infused for one and two weeks with isoproterenol (ISO) alone or in combination with phenylephrine (PE). Remodeling of the heart induced by these adrenergic agonists was assessed by echocardiography. Left ventricular (LV) tissue and plasma TH levels (T4 and T3) were determined using liquid chromatography-tandem mass spectrometry. LV D3 activity was determined by conversion of radiolabeled substrate and quantification following HPLC. The results show that ISO induced compensated LV hypertrophy with maintained cardiac output. Plasma levels of T4 and T3 remained normal, but LV hormone levels were reduced by approximately 30% after two weeks, while LV D3 activity was not significantly increased. ISO + PE induced decompensated LV hypertrophy with diminished cardiac output. Plasma levels of T4 and T3 were substantially reduced after one and two weeks, together with a more than 50% reduction of hormone levels in the LV. D3 activity was increased after one week and returned to control levels after two weeks. These data show for the first time that relative to controls, decompensated LV hypertrophy with diminished cardiac output is associated with a greater reduction of cardiac TH levels than compensated hypertrophy with maintained cardiac output. LV D3 activity is unlikely to account for these reductions after two weeks in either condition. Whereas the mechanism of the mild reduction in compensated hypertrophy is unclear, changes in systemic TH homeostasis appear to determine the marked drop in LV TH levels and associated impairment of ventricular function in decompensated hypertrophy.
Collapse
Affiliation(s)
- Warner Simonides
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
- Correspondence: (W.S.); (A.M.)
| | - Alice Tijsma
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Rutchanna Jongejan
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Yolanda de Rijke
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Robin Peeters
- Department of Internal Medicine, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA Rotterdam, The Netherlands
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Alice Muller
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, 1081 HZ Amsterdam, The Netherlands
- Correspondence: (W.S.); (A.M.)
| |
Collapse
|
5
|
Burman P, Jaiswal R, Devi K, Moharana B. Aurintricarboxylic acid protects isoproterenol induced left ventricular hypertrophy by modulating TWEAK signaling. Cardiovasc Pathol 2022; 61:107468. [PMID: 35977688 DOI: 10.1016/j.carpath.2022.107468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cardiac hypertrophy is regarded as a compensation mechanism to overcome the increased workload. Aurintricarboxylic acid (ATA) is a derivative of quinomethanes and a selective inhibitor of TWEAK/Fn14 pathway. In this study, we investigated the effect of ATA on isoproterenol (ISO)-induced pathological cardiac hypertrophy. METHODS Cardiac hypertrophy in H9C2 cells was induced using ISO 20 μM dissolved in PBS. H9C2 cells were treated with ATA (5 µM, 10 µM, 20 µM) followed by ISO stimulation for 24 h. Male SD rats were injected ISO (5 mg/kg/day, s.c) for 21 days and followed by treatment with ATA (10 mg/kg, i.p.) for 14 days. Cardiac functions were assessed. After sacrifice, hearts were subjected to histopathological and western blot analysis. RESULTS In in-vitro results, upon ATA treatment, ICC results showed significant decrease in TWEAK and ANP expression. In in-vivo results, echocardiography showed significant restoration of cardiac function in ATA treated rats. Histopathological analysis showed a significant decrease in left ventricular wall thickness, cardiomyocytes width and reduced fibrosis in ATA treated rats. Western blotting showed decreased expression of the cardiac hypertrophy maker ANP, inflammatory markers including TWEAK and apoptotic markers after ATA treatment. CONCLUSION These findings suggested that the TWEAK/Fn14 pathway could be a potential target for therapeutic exploration in ISO induced cardiac hypertrophy. ATA, as an inhibitor of this pathway, exerted significant cardioprotective effect against ISO-induced cardiac hypertrophy in rats.
Collapse
Affiliation(s)
- Prabha Burman
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rahul Jaiswal
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kusum Devi
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, UP, 201002, India
| | - Baisakhi Moharana
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, UP, 201002, India.
| |
Collapse
|
6
|
Sui X, Li N, Shi X, Li X, Han D, Qiu Z, Deng Y, Sun G. Pinocembrin Protects Cardiomyocytes Against Isoproterenol-Induced Hypertrophy. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211033216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiac hypertrophy is characterized by an increase in myocardial cell volume and extracellular matrix production. Persistent cardiac hypertrophy can cause dilated cardiomyopathy, heart failure, and even death. Pinocembrin (5,7-dihydroxyflavanone) is a type of flavonoid, extracted from propolis, that has antimicrobial, antioxidant, antiinflammatory, and anticancer properties. The results of the present study showed that pretreatment of isoproterenol (ISO)-treated H9c2 cardiomyocytes with pinocembrin reduced the messenger RNA levels of hypertrophic markers, including atrial natriuretic factor and βeta-myosin heavy chain, and inflammatory cytokines, such as tumor necrosis factor-α, interleukin-6, interleukin-1β, and interferon-γ, and also inhibited p65 phosphorylation and nuclear factor-kappa B (NF-κB) translocation. In addition, the activity of IκBα, an inhibitor of NF-κB, was increased while that of caspase-3 was reduced under these conditions. These results indicate that pinocembrin may inhibit ISO-induced myocardial hypertrophy by attenuating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xin Sui
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Na Li
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Xiaozheng Shi
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Xiaohua Li
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Dong Han
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Zhidong Qiu
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Yue Deng
- Changchun University of Chinese Medicine, Changchun, Jilin, P.R. China
| | - Guangwei Sun
- First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
- Chinese Traditional Medicine Institute of Jilin Province, Changchun, Jilin, P.R. China
| |
Collapse
|
7
|
CD47 Deficiency Attenuates Isoproterenol-Induced Cardiac Remodeling in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7121763. [PMID: 31827695 PMCID: PMC6885801 DOI: 10.1155/2019/7121763] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
In this study, we investigated whether CD47 deficiency attenuates isoproterenol- (ISO-) induced cardiac remodeling in mice. Cardiac remodeling was induced by intraperitoneal (i.p.) injection of ISO (60 mg·kg−1·d−1 in 100 μl of sterile normal saline) daily for 14 days and was confirmed by increased levels of lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB), increased heart weight to body weight (HW/BW) ratios, and visible cardiac fibrosis. Apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Levels of malondialdehyde (MDA) and reactive oxygen species (ROS) were found to be significantly higher in the ISO group than in the control group, while superoxide dismutase (SOD) levels were suppressed in the ISO group. However, CD47 knockout significantly limited ISO-induced increases in LDH, CK-MB, and HW/BW ratios, cardiac fibrosis, oxidative stress, and apoptosis in the heart. In addition, CD47 deficiency also increased p-AMPK and LAMP2 expression and decreased HDAC3, cleaved Caspase-3, cleaved Caspase-9, LC3II, and p62 expression in cardiac tissues. In conclusion, CD47 deficiency reduced i.p. ISO-induced cardiac remodeling probably by inhibiting the HDAC3 pathway, improving AMPK signaling and autophagy flux, and rescuing autophagic clearance.
Collapse
|
8
|
Gao J, Li Y, Wang T, Shi Z, Zhang Y, Liu S, Wen P, Ma C. Analyzing gene expression profiles with preliminary validations in cardiac hypertrophy induced by pressure overload. Can J Physiol Pharmacol 2018; 96:701-709. [PMID: 29510080 DOI: 10.1139/cjpp-2017-0585] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The aim of this study was to identify the key genes involved in the cardiac hypertrophy (CH) induced by pressure overload. mRNA microarray data sets GSE5500 and GSE18801 were downloaded from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) were screened using the Limma package; then, functional and pathway enrichment analysis were performed for common DEGs using the Database for Annotation, Visualization and Integrated Discovery database. Furthermore, the top DEGs were further validated using quantitative PCR in the hypertrophic heart tissue induced by isoprenaline. A total of 113 common DEGs with absolute fold change > 0.5, including 60 significantly upregulated DEGs and 53 downregulated DEGs, were obtained. Gene ontology term enrichment analysis suggested that common upregulated DEG were mainly enriched in neutrophil chemotaxis, extracellular fibril organization, and cell proliferation; and the common downregulated genes were significantly enriched in ion transport, endoplasmic reticulum, and dendritic spine. Kyoto Encyclopedia of Genes and Genomes pathway analysis found that the common DEGs were mainly enriched in extracellular matrix receptor interaction, phagosome, and focal adhesion. Additionally, the expression of Mfap4, Ltbp2, Aspn, Serpina3n, and Cnksr1 were upregulated in the model of CH, while the expression of Anp32a was downregulated. The current study identified the key deregulated genes and pathways involved in the CH, which could shed new light to understand the mechanism of CH.
Collapse
Affiliation(s)
- Jing Gao
- a Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang 110001, China.,b Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Yuhong Li
- b Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Tongmei Wang
- c Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Zhuo Shi
- d Department of Anatomy, Jinzhou Medical University, Jinzhou 121001, China
| | - Yiqi Zhang
- c Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Shuang Liu
- a Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang 110001, China
| | - Pushuai Wen
- c Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Chunyan Ma
- a Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
9
|
Maayah ZH, Levasseur J, Siva Piragasam R, Abdelhamid G, Dyck JRB, Fahlman RP, Siraki AG, El-Kadi AOS. 2-Methoxyestradiol protects against pressure overload-induced left ventricular hypertrophy. Sci Rep 2018; 8:2780. [PMID: 29426916 PMCID: PMC5807528 DOI: 10.1038/s41598-018-20613-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Numerous experimental studies have supported the evidence that 2-methoxyestradiol (2 ME) is a biologically active metabolite that mediates multiple effects on the cardiovascular system, largely independent of the estrogen receptor. 2 ME is a major cytochrome P450 1B1 (CYP1B1) metabolite and has been reported to have vasoprotective and anti-inflammatory actions. However, whether 2 ME would prevent cardiac hypertrophy induced by abdominal aortic constriction (AAC) has not been investigated yet. Therefore, the overall objectives of the present study were to elucidate the potential antihypertrophic effect of 2 ME and explore the mechanism(s) involved. Our results showed that 2 ME significantly inhibited AAC-induced left ventricular hypertrophy using echocardiography. The antihypertrophic effect of 2 ME was associated with a significant inhibition of CYP1B1 and mid-chain hydroxyeicosatetraenoic acids. Based on proteomics data, the protective effect of 2 ME is linked to the induction of antioxidant and anti-inflammatory proteins in addition to the modulation of proteins involved in myocardial energy metabolism. In vitro, 2 ME has shown a direct antihypertrophic effect through mitogen-activated protein kinases- and nuclear factor-κB-dependent mechanisms. The present work shows a strong evidence that 2 ME protects against left ventricular hypertrophy. Our data suggest the potential of repurposing 2 ME as a selective CYP1B1 inhibitor for the treatment of heart failure.
Collapse
Affiliation(s)
- Zaid H Maayah
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Jody Levasseur
- Cardiovascular Research Centre, Department of Pediatrics, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ramanaguru Siva Piragasam
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Ghada Abdelhamid
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Richard P Fahlman
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada.,Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Arno G Siraki
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy & Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, T6G 2E1, Canada.
| |
Collapse
|
10
|
Nour MS, Sarhan NR, Mazroa SA, Gawish SA. Histological and immunohistochemical study of cardiac telocytes in a rat model of isoproterenol-induced myocardial infarction with a reference to the effect of grape seed extract. Acta Histochem 2017; 119:747-758. [PMID: 28943000 DOI: 10.1016/j.acthis.2017.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cardiac telocytes (TCs) represent a unique type of cells that make a supportive network for stem cells that contribute in cardiac renewal, but their role during myocardial infarction (MI) is not clear. Grape seed extract (GSE) is a powerful natural antioxidant. AIM OF THE WORK Quantitative study of cardiac TCs in a rat model of Isoproterenol (ISO)-induced MI, and to evaluate the effect of GSE on TCs and MI progression. MATERIALS AND METHODS Seventy adult male albino rats were assigned into 4 groups; group I; control rats, group II received GSE (100mg/kg/day) dissolved in distilled water orally, group III received 2 intra-peritoneal injections of 85mg/kg ISO dissolved in saline on 14th and 15th day to induce MI, and group IV received GSE and ISO. Myocardium was obtained 1 and 14days after ISO i.e. on day 16 and day 30 respectively. Tissue was prepared for histological and immunohistochemical study of CD117 and CD34 as two markers for TCs. CD34 was used also as a marker for angiogenesis. RESULTS Group III showed focal areas of myocardial infarction 1day and 14days after ISO. Degenerated cardiomyocytes showed loss of striation and hypereosinophilic vacuolated cytoplasm with condensed nuclei. Mononuclear cell infiltration and a significantly increased percentage area of fibrosis 14days after ISO were observed. CD117 and CD34 positive TCs were hardly detected 1day after ISO. Their number slightly increased 14days after ISO with insignificant difference to control. There was also a significant increase in the number of CD34 positive blood vessels 14days after ISO. Group IV showed much better histological picture with a significant decrease in the percentage area of fibrosis and a significant increase in the number of CD117 and CD34 positive TCs and the number of CD34 positive blood vessels as compared to group III. CONCLUSION Telocytes were significantly decreased in MI. GSE reduced ISO-induced histological changes and increased the number of TCs that improved angiogenesis.
Collapse
Affiliation(s)
- Mai Salah Nour
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, 35516, Egypt.
| | - Nahla Reda Sarhan
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, 35516, Egypt.
| | - Shireen A Mazroa
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, 35516, Egypt.
| | - Salwa A Gawish
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, 35516, Egypt.
| |
Collapse
|
11
|
Ankyrin Repeat Domain 1 Protein: A Functionally Pleiotropic Protein with Cardiac Biomarker Potential. Int J Mol Sci 2017; 18:ijms18071362. [PMID: 28672880 PMCID: PMC5535855 DOI: 10.3390/ijms18071362] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022] Open
Abstract
The ankyrin repeat domain 1 (ANKRD1) protein is a cardiac-specific stress-response protein that is part of the muscle ankyrin repeat protein family. ANKRD1 is functionally pleiotropic, playing pivotal roles in transcriptional regulation, sarcomere assembly and mechano-sensing in the heart. Importantly, cardiac ANKRD1 has been shown to be highly induced in various cardiomyopathies and in heart failure, although it is still unclear what impact this may have on the pathophysiology of heart failure. This review aims at highlighting the known properties, functions and regulation of ANKRD1, with focus on the underlying mechanisms that may be involved. The current views on the actions of ANKRD1 in cardiovascular disease and its utility as a candidate cardiac biomarker with diagnostic and/or prognostic potential are also discussed. More studies of ANKRD1 are warranted to obtain deeper functional insights into this molecule to allow assessment of its potential clinical applications as a diagnostic or prognostic marker and/or as a possible therapeutic target.
Collapse
|
12
|
17β-Estradiol and/or estrogen receptor alpha blocks isoproterenol-induced calcium accumulation and hypertrophy via GSK3β/PP2A/NFAT3/ANP pathway. Mol Cell Biochem 2017; 434:181-195. [DOI: 10.1007/s11010-017-3048-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/25/2017] [Indexed: 12/13/2022]
|
13
|
Wang LX, Yang X, Yue Y, Fan T, Hou J, Chen GX, Liang MY, Wu ZK. Imatinib attenuates cardiac fibrosis by inhibiting platelet-derived growth factor receptors activation in isoproterenol induced model. PLoS One 2017; 12:e0178619. [PMID: 28570599 PMCID: PMC5453565 DOI: 10.1371/journal.pone.0178619] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/16/2017] [Indexed: 11/19/2022] Open
Abstract
Cardiac fibrosis is a significant global health problem with limited treatment choices. Although previous studies have shown that imatinib (IMA) inhibited cardiac fibrosis, the anti-fibrotic mechanisms have not been clearly uncovered. The aim of this study is to evaluate whether IMA attenuates cardiac fibrosis by inhibiting platelet-derived growth factor receptors (PDGFR) on isoproterenol (ISO)-induced mice. Adult male C57BL/6 mice were treated with vehicle or ISO ± IMA for one week. After echocardiography examination, the hearts of mice were used for histopathologic, RT-qPCR, and western blot analyses. We found that the ventricular wall thickness, cardiac hypertrophy, and apoptosis were enhanced following ISO treatment. IMA decreased the left ventricular wall thickness, prevented hypertrophy, and inhibited apoptosis induced by ISO. In addition, IMA attenuated the accumulation of collagens and α-smooth muscle actin (α-SMA) (the markers of fibrosis) caused by ISO treatment. Moreover, the expression of fibrosis related genes, and the phosphorylation of PDGFRs in ISO-treated mice hearts were inhibited by IMA as well. However, IMA did not change the expression of the matrix metalloproteinase-9 (MMP-9) in ISO-treated hearts. Furthermore, IMA reduced the expressions of collagens as well as α-SMA caused by activation of PDGFRα in cardiac fibroblasts. Taken together, our data demonstrate that IMA attenuated the cardiac fibrosis by blocking the phosphorylation of PDGFRs in the ISO-induced mice model. This study indicates that IMA could be a potentially therapeutic option for cardiac fibrosis in clinical application.
Collapse
Affiliation(s)
- Le-Xun Wang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao Yang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan Yue
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tian Fan
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Jian Hou
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guang-Xian Chen
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meng-Ya Liang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong-Kai Wu
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
14
|
The role of cytochrome P450 1B1 and its associated mid-chain hydroxyeicosatetraenoic acid metabolites in the development of cardiac hypertrophy induced by isoproterenol. Mol Cell Biochem 2017; 429:151-165. [DOI: 10.1007/s11010-017-2943-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 10/20/2022]
|
15
|
Prohibitin confers cytoprotection against ISO-induced hypertrophy in H9c2 cells via attenuation of oxidative stress and modulation of Akt/Gsk-3β signaling. Mol Cell Biochem 2016; 425:155-168. [PMID: 27854077 DOI: 10.1007/s11010-016-2870-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 11/02/2016] [Indexed: 12/14/2022]
Abstract
Numerous hypertrophic stimuli, including β-adrenergic agonists such as isoproterenol (ISO), result in generation of reactive oxygen species (ROS) and alteration in the mitochondrial membrane potential (Δψ) leading to oxidative stress. This process is well associated with phosphorylation of thymoma viral proto-oncogene Akt (Ser473) and glycogen synthase kinase-3β (Gsk-3β) (Ser9), with resultant inactivation of Gsk-3β. In the present study, we found that the protective defensive role of prohibitin (PHB) against ISO-induced hypertrophic response in rat H9c2 cells is via attenuation of oxidative stress-dependent signaling pathways. The intracellular levels of mitochondrial membrane potential along with cellular ROS levels and mitochondrial superoxide generation were determined. In order to understand the regulation of Akt/Gsk-3β signaling pathway, we carried out immmunoblotting for key proteins of the pathway such as PTEN, PI3K, phosphorylated, and unphosphorylated forms of Akt, Gsk-3β, and immunofluorescence experiments of p-Gsk-3β. Enforced expression of PHB in ISO-treated H9c2 cells suppressed cellular ROS production with mitochondrial superoxide generation and enhanced the mitochondrial membrane potential resulting in suppression of oxidative stress which likely offered potent cellular protection, led to the availability of more healthy cells, and also, significant constitutive activation of Gsk-3β via inactivation of Akt was observed. Knockdown of PHB expression using PHB siRNA in control H9c2 cells reversed these effects. Overall, our results demonstrate that PHB confers cytoprotection against oxidative stress in ISO-induced hypertrophy and this process is associated with modulation of Akt/Gsk-3β signaling mechanisms as evident from our PHB overexpression and knockdown experiments.
Collapse
|
16
|
Peters EL, Offringa C, Kos D, Van der Laarse WJ, Jaspers RT. Regulation of myoglobin in hypertrophied rat cardiomyocytes in experimental pulmonary hypertension. Pflugers Arch 2016; 468:1697-707. [PMID: 27572699 PMCID: PMC5026723 DOI: 10.1007/s00424-016-1865-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/21/2016] [Accepted: 08/04/2016] [Indexed: 01/29/2023]
Abstract
A major problem in chronic heart failure is the inability of hypertrophied cardiomyocytes to maintain the required power output. A Hill-type oxygen diffusion model predicts that oxygen supply is limiting in hypertrophied cardiomyocytes at maximal rates of oxygen consumption and that this limitation can be reduced by increasing the myoglobin (Mb) concentration. We explored how cardiac hypertrophy, oxidative capacity, and Mb expression in right ventricular cardiomyocytes are regulated at the transcriptional and translational levels in an early stage of experimental pulmonary hypertension, in order to identify targets to improve the oxygen supply/demand ratio. Male Wistar rats were injected with monocrotaline to induce pulmonary hypertension (PH) and right ventricular heart failure. The messenger RNA (mRNA) expression levels per nucleus of growth factors insulin-like growth factor-1Ea (IGF-1Ea) and mechano growth factor (MGF) were higher in PH than in healthy controls, consistent with a doubling in cardiomyocyte cross-sectional area (CSA). Succinate dehydrogenase (SDH) activity was unaltered, indicating that oxidative capacity per cell increased. Although the Mb protein concentration was unchanged, Mb mRNA concentration was reduced. However, total RNA per nucleus was about threefold higher in PH rats versus controls, and Mb mRNA content expressed per nucleus was similar in the two groups. The increase in oxidative capacity without an increase in oxygen supply via Mb-facilitated diffusion caused a doubling of the critical extracellular oxygen tension required to prevent hypoxia (PO2crit). We conclude that Mb mRNA expression is not increased during pressure overload-induced right ventricular hypertrophy and that the increase in myoglobin content per myocyte is likely due to increased translation. We conclude that increasing Mb mRNA expression may be beneficial in the treatment of experimental PH.
Collapse
Affiliation(s)
- E L Peters
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, van der Boechorststraat 7, 1081, BT, Amsterdam, The Netherlands
| | - C Offringa
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, van der Boechorststraat 7, 1081, BT, Amsterdam, The Netherlands
| | - D Kos
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, van der Boechorststraat 7, 1081, BT, Amsterdam, The Netherlands.,Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - W J Van der Laarse
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - R T Jaspers
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, van der Boechorststraat 7, 1081, BT, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Wang JJC, Rau C, Avetisyan R, Ren S, Romay MC, Stolin G, Gong KW, Wang Y, Lusis AJ. Genetic Dissection of Cardiac Remodeling in an Isoproterenol-Induced Heart Failure Mouse Model. PLoS Genet 2016; 12:e1006038. [PMID: 27385019 PMCID: PMC4934852 DOI: 10.1371/journal.pgen.1006038] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
We aimed to understand the genetic control of cardiac remodeling using an isoproterenol-induced heart failure model in mice, which allowed control of confounding factors in an experimental setting. We characterized the changes in cardiac structure and function in response to chronic isoproterenol infusion using echocardiography in a panel of 104 inbred mouse strains. We showed that cardiac structure and function, whether under normal or stress conditions, has a strong genetic component, with heritability estimates of left ventricular mass between 61% and 81%. Association analyses of cardiac remodeling traits, corrected for population structure, body size and heart rate, revealed 17 genome-wide significant loci, including several loci containing previously implicated genes. Cardiac tissue gene expression profiling, expression quantitative trait loci, expression-phenotype correlation, and coding sequence variation analyses were performed to prioritize candidate genes and to generate hypotheses for downstream mechanistic studies. Using this approach, we have validated a novel gene, Myh14, as a negative regulator of ISO-induced left ventricular mass hypertrophy in an in vivo mouse model and demonstrated the up-regulation of immediate early gene Myc, fetal gene Nppb, and fibrosis gene Lgals3 in ISO-treated Myh14 deficient hearts compared to controls. Heart failure is the most common cause of morbidity and mortality in the aging population. Previous large-scale human genome-wide association studies have yielded only a handful of genetic loci contributing to heart failure-related traits. Using a panel of diverse inbred mouse strains, treated with a β-adrenergic agonist isoproterenol to mimic the heart failure state, we sought to uncover the contribution of common genetic variation in heart failure. We found that heart failure has a strong genetic component. We successfully identified 17 genome-wide significant loci associated with indices of heart failure. We showed that genetic variation in a novel gene Myh14 affects heart failure by altering the mechanical responses of heart muscles to isoproterenol-induced stress. Follow-up studies of this gene and additional candidate genes and loci should reveal potential mechanisms by which genetic variations contribute to heart failure in the general human population. Such insights may lead to improved diagnosis and tailor treatment based on the genetic makeup of individuals in the population.
Collapse
Affiliation(s)
- Jessica Jen-Chu Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail: (JJCW); (AJL)
| | - Christoph Rau
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Rozeta Avetisyan
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Shuxun Ren
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Milagros C. Romay
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Gabriel Stolin
- Department of Molecular, Cell, and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ke Wei Gong
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Yibin Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Aldons J. Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail: (JJCW); (AJL)
| |
Collapse
|
18
|
Al-Rasheed NM, Al-Oteibi MM, Al-Manee RZ, Al-Shareef SA, Al-Rasheed NM, Hasan IH, Mohamad RA, Mahmoud AM. Simvastatin prevents isoproterenol-induced cardiac hypertrophy through modulation of the JAK/STAT pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3217-29. [PMID: 26150695 PMCID: PMC4484667 DOI: 10.2147/dddt.s86431] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Simvastatin (SIM) is a lipid-soluble inhibitor of hydroxy-3-methylglutaryl coenzyme A reductase with multiple reported therapeutic benefits. The present study was designed to investigate the effect of pretreatment with SIM on isoproterenol (ISO)-induced cardiac hypertrophy in rats. Twenty-four male albino Wistar rats weighing 180–200 g were divided into four groups. Groups I and III received normal saline while groups II and IV received SIM (10 mg/kg body weight) for 30 days per gavage. In the last 7 days, rats of groups III and IV were administered ISO (5 mg/kg) intraperitoneally to induce cardiac hypertrophy. Administration of ISO induced an increase in heart-to-body weight (HW/BW) ratio, an increase in serum interleukin-6, and elevated systolic and diastolic blood pressure. Serum levels of lipids, cardiovascular risk indices, and cardiac troponin I and creatine phosphokinase-MB showed significant increase in ISO-induced hypertrophic rats. Histopathological examination of heart tissue revealed focal areas of subendocardium degeneration, mononuclear cellular infiltrations, fibrous tissue deposition, and increased thickness of the myocardium of left ventricle. In addition, ISO-administered rats exhibited significant upregulation of cardiac Janus kinase, phosphorylated signal transducer and activator of transcription, and nuclear factor-kappa B. Pretreatment with SIM significantly prevented ISO-induced cardiac hypertrophy, alleviated the altered biochemical parameters, and improved the heart architecture. In conclusion, our study provides evidence that SIM prevented the development of cardiac hypertrophy via modulation of the Janus kinase/signal transducer and activator of transcription-signaling pathway in the heart of ISO-administered animals.
Collapse
Affiliation(s)
- Nouf M Al-Rasheed
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maha M Al-Oteibi
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Reem Z Al-Manee
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarah A Al-Shareef
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nawal M Al-Rasheed
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Iman H Hasan
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raeesa A Mohamad
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ayman M Mahmoud
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Egypt
| |
Collapse
|
19
|
Yang S, Chen L, Sun S, Shah P, Yang W, Zhang B, Zhang Z, Chan DW, Kass DA, van Eyk JE, Zhang H. Glycoproteins identified from heart failure and treatment models. Proteomics 2015; 15:567-79. [PMID: 25141849 PMCID: PMC4492696 DOI: 10.1002/pmic.201400151] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 07/28/2014] [Accepted: 08/15/2014] [Indexed: 01/12/2023]
Abstract
Conduction abnormalities can lead to dyssynchronous contraction, which significantly worsens morbidity and mortality of heart failure. Cardiac resynchronization therapy (CRT) can reverse ventricular remodeling and improve cardiac function. Although the underlying molecular changes are unknown, the use of a canine model of dyssynchronous heart failure (DHF) and CRT has shown that there are global changes across the cardiac proteome. This study determines changes in serum glycoprotein concentration from DHF and CRT compared to normal. We hypothesize that CRT invokes protective or advantageous pathways that can be reflected in the circulating proteome. Two prong discovery approaches were carried out on pooled normal, DHF, and CRT samples composed of individual canine serum to determine the overall protein concentration and the N-linked glycosites of circulating glycoproteins. The level of the glycoproteins was altered in DHF and CRT compared to control sera, with 63 glycopeptides substantially increased in DHF and/or CRT. Among the 32 elevated glycosite-containing peptides in DHF, 13 glycopeptides were reverted to normal level after CRT therapy. We further verify the changes of glycopeptides using label-free LC-MS from individual canine serum. Circulating glycoproteins such as alpha-fetoprotein, alpha-2-macroglobulin, galectin-3-binding protein, and collectin-10 show association to failing heart and CRT treatment model.
Collapse
Affiliation(s)
- Shuang Yang
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Shisheng Sun
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Punit Shah
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Weiming Yang
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Bai Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - Daniel W. Chan
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| | - David A. Kass
- Cellular and Molecular Medicine, Johns Hopkins University, Baltimore, 21205
| | - Jennifer E. van Eyk
- Department of Medicine, Johns Hopkins University, Baltimore, 21224
- Advanced Clinical Biosystems Research Institute, Cedar Sinai Medical Center, Los Angeles
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, 21231
| |
Collapse
|
20
|
Grisanti LA, Repas AA, Talarico JA, Gold JI, Carter RL, Koch WJ, Tilley DG. Temporal and gefitinib-sensitive regulation of cardiac cytokine expression via chronic β-adrenergic receptor stimulation. Am J Physiol Heart Circ Physiol 2014; 308:H316-30. [PMID: 25485901 DOI: 10.1152/ajpheart.00635.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic stimulation of β-adrenergic receptors (βAR) can promote survival signaling via transactivation of epidermal growth factor receptor (EGFR) but ultimately alters cardiac structure and contractility over time, in part via enhanced cytokine signaling. We hypothesized that chronic catecholamine signaling will have a temporal impact on cardiac transcript expression in vivo, in particular cytokines, and that EGFR transactivation plays a role in this process. C57BL/6 mice underwent infusion with vehicle or isoproterenol (Iso)±gefitinib (Gef) for 1 or 2 wk. Cardiac contractility decreased following 2 wk of Iso treatment, while cardiac hypertrophy, fibrosis, and apoptosis were enhanced at both timepoints. Inclusion of Gef preserved contractility, blocked Iso-induced apoptosis, and prevented hypertrophy at the 2-wk timepoint, but caused fibrosis on its own. RNAseq analysis revealed hundreds of cardiac transcripts altered by Iso at each timepoint with subsequent RT-quantitative PCR validation confirming distinct temporal patterns of transcript regulation, including those involved in cardiac remodeling and survival signaling, as well as numerous cytokines. Although Gef infusion alone did not significantly alter cytokine expression, it abrogated the Iso-mediated changes in a majority of the βAR-sensitive cytokines, including CCL2 and TNF-α. Additionally, the impact of βAR-dependent EGFR transactivation on the acute regulation of cytokine transcript expression was assessed in isolated cardiomyocytes and in cardiac fibroblasts, where the majority of Iso-dependent, and EGFR-sensitive, changes in cytokines occurred. Overall, coincident with changes in cardiac structure and contractility, βAR stimulation dynamically alters cardiac transcript expression over time, including numerous cytokines that are regulated via EGFR-dependent signaling.
Collapse
Affiliation(s)
- Laurel A Grisanti
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Ashley A Repas
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Jennifer A Talarico
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jessica I Gold
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rhonda L Carter
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Walter J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Douglas G Tilley
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
21
|
Different effects of prolonged β-adrenergic stimulation on heart and cerebral artery. Integr Med Res 2014; 3:204-210. [PMID: 28664099 PMCID: PMC5481746 DOI: 10.1016/j.imr.2014.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/01/2014] [Accepted: 10/01/2014] [Indexed: 01/19/2023] Open
Abstract
The aim of this review was to understand the effects of β-adrenergic stimulation on oxidative stress, structural remodeling, and functional alterations in the heart and cerebral artery. Diverse stimuli activate the sympathetic nervous system, leading to increased levels of catecholamines. Long-term overstimulation of the β-adrenergic receptor (βAR) in response to catecholamines causes cardiovascular diseases, including cardiac hypertrophy, stroke, coronary artery disease, and heart failure. Although catecholamines have identical sites of action in the heart and cerebral artery, the structural and functional modifications differentially activate intracellular signaling cascades. βAR-stimulation can increase oxidative stress in the heart and cerebral artery, but has also been shown to induce different cytoskeletal and functional modifications by modulating various components of the βAR signal transduction pathways. Stimulation of βAR leads to cardiac dysfunction due to an overload of intracellular Ca2+ in cardiomyocytes. However, this stimulation induces vascular dysfunction through disruption of actin cytoskeleton in vascular smooth muscle cells. Many studies have shown that excessive concentrations of catecholamines during stressful conditions can produce coronary spasms or arrhythmias by inducing Ca2+-handling abnormalities and impairing energy production in mitochondria, In this article, we highlight the different fates caused by excessive oxidative stress and disruptions in the cytoskeletal proteome network in the heart and the cerebral artery in responsed to prolonged βAR-stimulation.
Collapse
|
22
|
Talarico JA, Carter RL, Grisanti LA, Yu JE, Repas AA, Tilley DG. β-adrenergic receptor-dependent alterations in murine cardiac transcript expression are differentially regulated by gefitinib in vivo. PLoS One 2014; 9:e99195. [PMID: 24901703 PMCID: PMC4047088 DOI: 10.1371/journal.pone.0099195] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/12/2014] [Indexed: 11/18/2022] Open
Abstract
β-adrenergic receptor (βAR)-mediated transactivation of epidermal growth factor receptor (EGFR) has been shown to promote cardioprotection in a mouse model of heart failure and we recently showed that this mechanism leads to enhanced cell survival in part via regulation of apoptotic transcript expression in isolated primary rat neonatal cardiomyocytes. Thus, we hypothesized that this process could regulate cardiac transcript expression in vivo. To comprehensively assess cardiac transcript alterations in response to acute βAR-dependent EGFR transactivation, we performed whole transcriptome analysis of hearts from C57BL/6 mice given i.p. injections of the βAR agonist isoproterenol in the presence or absence of the EGFR antagonist gefitinib for 1 hour. Total cardiac RNA from each treatment group underwent transcriptome analysis, revealing a substantial number of transcripts regulated by each treatment. Gefitinib alone significantly altered the expression of 405 transcripts, while isoproterenol either alone or in conjunction with gefitinib significantly altered 493 and 698 distinct transcripts, respectively. Further statistical analysis was performed, confirming 473 transcripts whose regulation by isoproterenol were significantly altered by gefitinib (isoproterenol-induced up/downregulation antagonized/promoted by gefinitib), including several known to be involved in the regulation of numerous processes including cell death and survival. Thus, βAR-dependent regulation of cardiac transcript expression in vivo can be modulated by the EGFR antagonist gefitinib.
Collapse
Affiliation(s)
- Jennifer A. Talarico
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rhonda L. Carter
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Laurel A. Grisanti
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Justine E. Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Ashley A. Repas
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Douglas G. Tilley
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
23
|
Szibor M, Pöling J, Warnecke H, Kubin T, Braun T. Remodeling and dedifferentiation of adult cardiomyocytes during disease and regeneration. Cell Mol Life Sci 2014; 71:1907-16. [PMID: 24322910 PMCID: PMC11113405 DOI: 10.1007/s00018-013-1535-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 12/20/2022]
Abstract
Cardiomyocytes continuously generate the contractile force to circulate blood through the body. Imbalances in contractile performance or energy supply cause adaptive responses of the heart resulting in adverse rearrangement of regular structures, which in turn might lead to heart failure. At the cellular level, cardiomyocyte remodeling includes (1) restructuring of the contractile apparatus; (2) rearrangement of the cytoskeleton; and (3) changes in energy metabolism. Dedifferentiation represents a key feature of cardiomyocyte remodeling. It is characterized by reciprocal changes in the expression pattern of "mature" and "immature" cardiomyocyte-specific genes. Dedifferentiation may enable cardiomyocytes to cope with hypoxic stress by disassembly of the energy demanding contractile machinery and by reduction of the cellular energy demand. Dedifferentiation during myocardial repair might provide cardiomyocytes with additional plasticity, enabling survival under hypoxic conditions and increasing the propensity to enter the cell cycle. Although dedifferentiation of cardiomyocytes has been described during tissue regeneration in zebrafish and newts, little is known about corresponding mechanisms and regulatory circuits in mammals. The recent finding that the cytokine oncostatin M (OSM) is pivotal for cardiomyocyte dedifferentiation and exerts strong protective effects during myocardial infarction highlights the role of cytokines as potent stimulators of cardiac remodeling. Here, we summarize the current knowledge about transient dedifferentiation of cardiomyocytes in the context of myocardial remodeling, and propose a model for the role of OSM in this process.
Collapse
Affiliation(s)
- Marten Szibor
- Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- Research Program of Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Jochen Pöling
- Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- Department of Cardiac Surgery, Schüchtermann Clinic, Bad Rothenfelde, Germany
| | - Henning Warnecke
- Department of Cardiac Surgery, Schüchtermann Clinic, Bad Rothenfelde, Germany
| | - Thomas Kubin
- Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| |
Collapse
|
24
|
Effects of hypertension and exercise on cardiac proteome remodelling. BIOMED RESEARCH INTERNATIONAL 2014; 2014:634132. [PMID: 24877123 PMCID: PMC4022191 DOI: 10.1155/2014/634132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/14/2014] [Indexed: 12/29/2022]
Abstract
Left ventricle hypertrophy is a common outcome of pressure overload stimulus closely associated with hypertension. This process is triggered by adverse molecular signalling, gene expression, and proteome alteration. Proteomic research has revealed that several molecular targets are associated with pathologic cardiac hypertrophy, including angiotensin II, endothelin-1 and isoproterenol. Several metabolic, contractile, and stress-related proteins are shown to be altered in cardiac hypertrophy derived by hypertension. On the other hand, exercise is a nonpharmacologic agent used for hypertension treatment, where cardiac hypertrophy induced by exercise training is characterized by improvement in cardiac function and resistance against ischemic insult. Despite the scarcity of proteomic research performed with exercise, healthy and pathologic heart proteomes are shown to be modulated in a completely different way. Hence, the altered proteome induced by exercise is mostly associated with cardioprotective aspects such as contractile and metabolic improvement and physiologic cardiac hypertrophy. The present review, therefore, describes relevant studies involving the molecular characteristics and alterations from hypertensive-induced and exercise-induced hypertrophy, as well as the main proteomic research performed in this field. Furthermore, proteomic research into the effect of hypertension on other target-demerged organs is examined.
Collapse
|
25
|
The elusive importance of being a mitochondrial Ca2+ uniporter. Cell Calcium 2014; 55:139-45. [DOI: 10.1016/j.ceca.2014.02.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 12/20/2022]
|
26
|
Li Y, Shen D, Tang X, Li X, Wo D, Yan H, Song R, Feng J, Li P, Zhang J, Li J. Chlorogenic acid prevents isoproterenol-induced hypertrophy in neonatal rat myocytes. Toxicol Lett 2014; 226:257-63. [PMID: 24583048 DOI: 10.1016/j.toxlet.2014.02.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/19/2014] [Accepted: 02/19/2014] [Indexed: 11/18/2022]
Abstract
Cardiac hypertrophy is an independent risk factor for cardiovascular disease and its subsequent progression to heart failure represents a major cause of morbidity and mortality in the world. CGA is an important component of Chinese herbal medicine, acting as an antioxidant, scavenging free radicals and preventing inflammation. This study found that with the pre-treatment of chlorogenic acid in Iso-induced neonatal rat myocytes, the levels of the hypertrophic markers, ANP, BNP and β-MHC decreased. The nuclear translocation of NF-κB was blocked, whereas NF-κBIA, an inhibitor of NF-κB, was upregulated accordingly. And the level of the intracellular ROS was also reduced. These data reveal that chlorogenic acid may inhibit Iso-induced cardiac hypertrophy by attenuating NF-κB signaling pathway and suppressing ROS.
Collapse
Affiliation(s)
- Yanfei Li
- School of Life Sciences and Technology, Tongji University, Shanghai, China; Tongji University School of Medicine, Shanghai, China
| | - Dan Shen
- Tongji University School of Medicine, Shanghai, China
| | - Xiaomei Tang
- Tongji University School of Medicine, Shanghai, China
| | - Xin Li
- Tongji University School of Medicine, Shanghai, China
| | - Da Wo
- Tongji University School of Medicine, Shanghai, China
| | - Hongwei Yan
- Tongji University School of Medicine, Shanghai, China
| | - Rui Song
- Tongji University School of Medicine, Shanghai, China
| | - Jian Feng
- Tongji University School of Medicine, Shanghai, China
| | - Ping Li
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jie Zhang
- Tongji University School of Medicine, Shanghai, China.
| | - Jue Li
- Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Chowdhury D, Tangutur AD, Khatua TN, Saxena P, Banerjee SK, Bhadra MP. A proteomic view of isoproterenol induced cardiac hypertrophy: prohibitin identified as a potential biomarker in rats. J Transl Med 2013; 11:130. [PMID: 23706090 PMCID: PMC3667141 DOI: 10.1186/1479-5876-11-130] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 05/01/2013] [Indexed: 01/15/2023] Open
Abstract
Background The present study aimed at using a proteomics based approach to: a) analyze and contrast the proteome of the healthy and isoproterenol induced hypertrophied hearts and b) identify potential biomarkers for diagnosis of cardiac hypertrophy. Methods Male Sprague Dawley (SD) rats were administered isoproterenol (ISO, 5 mg/kg, sc, once daily) for 14 days to induce cardiac hypertrophy. There was a significant (p<0.05) increase (~ 55%) in the heart weight to tail length ratio after 14 days of treatment and cardiac hypertrophy was evidenced by significant increase of β-MHC and ANP, two indicative markers of cardiac hypertrophy, in the treated heart compared to that of control. Following confirmation of hypertrophy, 2DE of the tissue samples was done followed by MS/MS analysis of the protein spots to obtain a proteomic view for identification of novel biomarkers. Results Several important proteins were identified by proteomics analysis. They belong to the major functional categories such as cholesterol and protein metabolism, muscle contraction and development, transport, TCAcycle, ATP-biosynthesis, chaperone, signal transduction, DNA synthesis and ubiquitinisation. Careful examination of these protein spots by image analysis led to the successful identification of 7 differentially expressed proteins in the diseased sample. Further extension of this work for validation of differential expression of these proteins was also achieved by RTPCR and western blotting. Conclusions Our results demonstrate characteristic protein expression profile in control and hypertrophy condition in SD rats and also expand the existing knowledge on differentially expressed proteins in hypertrophy. The study signifies the importance of reduced expression of a novel protein such as Prohibitin (PHB) which may be associated with the cardiomyocytes growth and cardiac hypertrophy. However, further work is necessary to confirm the role of PHB in human heart and its potential role in diagnostic and therapeutic intervention in the clinic.
Collapse
Affiliation(s)
- Debabrata Chowdhury
- Centre for Chemical Biology, Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500607, India
| | | | | | | | | | | |
Collapse
|
28
|
Söker T, Gödecke A. Expression of the murine Nr4a1 gene is controlled by three distinct genomic loci. Gene 2012; 512:517-20. [PMID: 23078765 DOI: 10.1016/j.gene.2012.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 10/02/2012] [Accepted: 10/03/2012] [Indexed: 11/18/2022]
Abstract
The transcription factor NR4A1 belongs to the class of orphan nuclear receptors without known ligand which might control its activity. Here we examined its transcriptional regulation in response to elevated cAMP levels in HL-1 cardiac myocytes and in the heart in vivo. We report, that murine Nr4a1 is expressed from distinct promoters located upto -11.5 kb from the first coding exon. Whereas in HL-1 cells only two of the three distinct transcripts were induced by cAMP, in the heart all transcripts were regulated in response to β-adrenergic stimulation.
Collapse
Affiliation(s)
- Torben Söker
- Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität, Universitätsklinikum Düsseldorf, D-40225 Düsseldorf, Germany.
| | | |
Collapse
|
29
|
Moellendorf S, Kessels C, Peiseler L, Raupach A, Jacoby C, Vogt N, Lindecke A, Koch L, Brüning J, Heger J, Köhrer K, Gödecke A. IGF-IR signaling attenuates the age-related decline of diastolic cardiac function. Am J Physiol Endocrinol Metab 2012; 303:E213-22. [PMID: 22589390 DOI: 10.1152/ajpendo.00538.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Insulin-like growth factor (IGF-I) signaling has been implicated to play an important role in regulation of cardiac growth, hypertrophy, and contractile function and has been linked to the development of age-related congestive heart failure. Here, we address the question to what extent cardiomyocyte-specific IGF-I signaling is essential for maintenance of the structural and functional integrity of the adult murine heart. To investigate the effects of IGF-I signaling in the adult heart without confounding effects due to IGF-I overexpression or adaptation during embryonic and early postnatal development, we inactivated the IGF-I receptor (IGF-IR) by a 4-hydroxytamoxifen-inducible Cre recombinase in adult cardiac myocytes. Efficient inactivation of the IGF-IR (iCMIGF-IRKO) as assessed by Western analysis and real-time PCR went along with reduced IGF-I-dependent Akt and GSK3β phosphorylation. Functional analysis by conductance manometry and MRI revealed no functional alterations in young adult iCMIGF-IRKO mice (age 3 mo). However, when induced in aging mice (11 mo) diastolic cardiac function was depressed. To address the question whether insulin signaling might compensate for the defective IGF-IR signaling, we inactivated β-cells by streptozotocin. However, the diabetes-associated functional depression was similar in control and iCMIGF-IRKO mice. Similarly, analysis of the cardiac gene expression profile on 44K microarrays did not reveal activation of overt adaptive processes. Endogenous IGF-IR signaling is required for conservation of cardiac function of the aging heart, but not for the integrity of cardiac structure and function of young hearts.
Collapse
Affiliation(s)
- Sarah Moellendorf
- Institut für Herz- und Kreislaufphysiologie, Universitätsklinikum, Heinrich-Heine-Universität Düsseldorf, Dusseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|