51
|
Recombinant neuregulin 1 does not activate cardiomyocyte DNA synthesis in normal or infarcted adult mice. PLoS One 2014; 9:e115871. [PMID: 25545368 PMCID: PMC4278834 DOI: 10.1371/journal.pone.0115871] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 11/28/2014] [Indexed: 11/19/2022] Open
Abstract
Objectives Neuregulin 1 signaling plays an important role in cardiac trabecular development, and in sustaining functional integrity in adult hearts. Treatment with neuregulin 1 enhances adult cardiomyocyte differentiation, survival and/or function in vitro and in vivo. It has also been suggested that recombinant neuregulin 1β1 (NRG1β1) induces cardiomyocyte proliferation in normal and injured adult hearts. Here we further explore the impact of neuregulin 1 signaling on adult cardiomyocyte cell cycle activity. Methods and Results Adult mice were subjected to 9 consecutive daily injections of recombinant NRG1β1 or vehicle, and cardiomyocyte DNA synthesis was quantitated via bromodeoxyuridine (BrdU) incorporation, which was delivered using mini-osmotic pumps over the entire duration of NRG1β1 treatment. NRG1β1 treatment inhibited baseline rates of cardiomyocyte DNA synthesis in normal mice (cardiomyocyte labelling index: 0.019±0.005% vs. 0.003±0.001%, saline vs. NRG1β1, P<0.05). Acute NRG1β1 treatment did result in activation of Erk1/2 and cardiac myosin regulatory light chain (down-stream mediators of neuregulin signalling), as well as activation of DNA synthesis in non-cardiomyocytes, validating the biological activity of the recombinant protein. In other studies, mice were subjected to permanent coronary artery occlusion, and cardiomyocyte DNA synthesis was monitored via tritiated thymidine incorporation which was delivered as a single injection 7 days post-infarction. Daily NRG1β1 treatment had no impact on cardiomyocyte DNA synthesis in the infarcted myocardium (cardiomyocyte labelling index: 0.039±0.011% vs. 0.027±0.021%, saline vs. NRG1β1, P>0.05). Summary These data indicate that NRG1β1 treatment does not increase cardiomyocyte DNA synthesis (and consequently does not increase the rate of cardiomyocyte renewal) in normal or infarcted adult mouse hearts. Thus, any improvement in cardiac structure and function observed following neuregulin treatment of injured hearts likely occurs independently of overt myocardial regeneration.
Collapse
|
52
|
Abstract
The beta isoform of Neuregulin-1 (NRG-1β), along with its receptors (ErbB2-4), is required for cardiac development. NRG-1β, as well as the ErbB2 and ErbB4 receptors, is also essential for maintenance of adult heart function. These observations have led to its evaluation as a therapeutic for heart failure. Animal studies and ongoing clinical trials have demonstrated beneficial effects of two forms of recombinant NRG-1β on cardiac function. In addition to the possible role for recombinant NRG-1βs as heart failure therapies, endogenous NRG-1β/ErbB signaling appears to play a role in restoring cardiac function after injury. The potential mechanisms by which NRG-1β may act as both a therapy and a mediator of reverse remodeling remain incompletely understood. In addition to direct effects on cardiac myocytes NRG-1β acts on the vasculature, interstitium, cardiac fibroblasts, and hematopoietic and immune cells, which, collectively, may contribute to NRG-1β's role in maintaining cardiac structure and function, as well as mediating reverse remodeling.
Collapse
|
53
|
Vasti C, Hertig CM. Neuregulin-1/erbB activities with focus on the susceptibility of the heart to anthracyclines. World J Cardiol 2014; 6:653-662. [PMID: 25068025 PMCID: PMC4110613 DOI: 10.4330/wjc.v6.i7.653] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 02/11/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
Neuregulin-1 (NRG1) signaling through the tyrosine kinase receptors erbB2 and erbB4 is required for cardiac morphogenesis, and it plays an essential role in maintaining the myocardial architecture during adulthood. The tyrosine kinase receptor erbB2 was first linked to the amplification and overexpression of erbb2 gene in a subtype of breast tumor cells, which is indicative of highly proliferative cells and likely a poor prognosis following conventional chemotherapy. The development of targeted therapies to block the survival of erbB2-positive cancer cells revealed that impaired NRG1 signaling through erbB2/erbB4 heterodimers combined with anthracycline chemotherapy may lead to dilated cardiomyopathy in a subpopulation of treated patients. The ventricular-specific deletion of either erbb2 or erbb4 manifested dilated cardiomyopathy, which is aggravated by the administration of doxorubicin. Based on the exacerbated toxicity displayed by the combined treatment, it is expected that the relevant pathways would be affected in a synergistic manner. This review examines the NRG1 activities that were monitored in different model systems, focusing on the emerging pathways and molecular targets, which may aid in understanding the acquired dilated cardiomyopathy that occurs under the conditions of NRG1-deficient signaling.
Collapse
|
54
|
Paatero I, Seagroves TN, Vaparanta K, Han W, Jones FE, Johnson RS, Elenius K. Hypoxia-inducible factor-1α induces ErbB4 signaling in the differentiating mammary gland. J Biol Chem 2014; 289:22459-69. [PMID: 24966332 DOI: 10.1074/jbc.m113.533497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Conditional knock-out of Hif1a in the mouse mammary gland impairs lobuloalveolar differentiation during lactation. Here, we demonstrate that expression of ErbB4 was reduced in the lobulalveoli of mice with mammary gland-specific deletion of Hif1a. Erbb4 was not, however, a direct target gene for transcriptional regulation by HIF-1α in vitro. HIF-1α overexpression or HIF accumulating prolyl hydroxylase inhibitors reduced ErbB4 endocytosis, promoted transcriptional co-regulatory activity of ErbB4, and stimulated ErbB4-induced differentiation of mammary carcinoma cells. Consistently, RNA interference-mediated down-regulation of HIF-1α resulted in reduced ErbB4 protein amount and reduced mammary carcinoma cell differentiation. These findings indicate that HIF-1α is a physiologically relevant regulator of ErbB4 and that ErbB4 is involved in HIF-regulated differentiation of the mammary gland.
Collapse
Affiliation(s)
- Ilkka Paatero
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, 20520 Turku, Finland, the Turku Graduate School of Biomedical Sciences, 20520 Turku, Finland
| | - Tiffany N Seagroves
- the Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Katri Vaparanta
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Wen Han
- the Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Frank E Jones
- the Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Randall S Johnson
- the Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge CB2 1TN, United Kingdom, and
| | - Klaus Elenius
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, 20520 Turku, Finland, the Department of Oncology, Turku University Hospital, 20520 Turku, Finland
| |
Collapse
|
55
|
Central neuregulin-1/ErbB signaling modulates cardiac function via sympathetic activity in pressure overload-induced heart failure. J Hypertens 2014; 32:817-25. [DOI: 10.1097/hjh.0000000000000072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
56
|
Ryall KA, Bezzerides VJ, Rosenzweig A, Saucerman JJ. Phenotypic screen quantifying differential regulation of cardiac myocyte hypertrophy identifies CITED4 regulation of myocyte elongation. J Mol Cell Cardiol 2014; 72:74-84. [PMID: 24613264 DOI: 10.1016/j.yjmcc.2014.02.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/18/2014] [Accepted: 02/25/2014] [Indexed: 01/19/2023]
Abstract
Cardiac hypertrophy is controlled by a highly connected signaling network with many effectors of cardiac myocyte size. Quantification of the contribution of individual pathways to specific changes in shape and transcript abundance is needed to better understand hypertrophy signaling and to improve heart failure therapies. We stimulated cardiac myocytes with 15 hypertrophic agonists and quantitatively characterized differential regulation of 5 shape features using high-throughput microscopy and transcript levels of 12 genes using qPCR. Transcripts measured were associated with phenotypes including fibrosis, cell death, contractility, proliferation, angiogenesis, inflammation, and the fetal cardiac gene program. While hypertrophy pathways are highly connected, the agonist screen revealed distinct hypertrophy phenotypic signatures for the 15 receptor agonists. We then used k-means clustering of inputs and outputs to identify a network map linking input modules to output modules. Five modules were identified within inputs and outputs with many maladaptive outputs grouping together in one module: Bax, C/EBPβ, Serca2a, TNFα, and CTGF. Subsequently, we identified mechanisms underlying two correlations revealed in the agonist screen: correlation between regulators of fibrosis and cell death signaling (CTGF and Bax mRNA) caused by AngII; and myocyte proliferation (CITED4 mRNA) and elongation caused by Nrg1. Follow-up experiments revealed positive regulation of Bax mRNA level by CTGF and an incoherent feedforward loop linking Nrg1, CITED4 and elongation. With this agonist screen, we identified the most influential inputs in the cardiac hypertrophy signaling network for a variety of features related to pathological and protective hypertrophy signaling and shared regulation among cardiac myocyte phenotypes.
Collapse
Affiliation(s)
- Karen A Ryall
- Department of Biomedical Engineering, University of Virginia, VA, USA
| | - Vassilios J Bezzerides
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Anthony Rosenzweig
- Cardiovascular Division of the Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
57
|
|
58
|
Patel N, Barrientos A, Landgraf R. The growth factor receptor ERBB2 regulates mitochondrial activity on a signaling time scale. J Biol Chem 2013; 288:35253-65. [PMID: 24142693 DOI: 10.1074/jbc.m113.478271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Overexpression of the ERBB2 receptor tyrosine kinase and the mitochondrial inner membrane protein UCP2 occurs frequently in aggressive cancers with dysfunctional mitochondria. Overexpressed ERBB2 signals constitutively and elevated UCP2 can uncouple mitochondria and alleviate oxidative stress. However, the physiological contributions of UCP2 and ERBB2 at the low expression levels that are typical of most tissues, as well as the path to oncogenic deregulation, are poorly understood. We now show that ERBB2 directly controls UCP2 levels, both at low physiological levels and oncogenic overexpression. At low levels of receptor and UCP2, ligand stimulation creates a distinct temporal response pattern driven by the opposing forces of translational suppression of the exceptionally short lived UCP2 protein and a time delayed transcriptional up-regulation. The latter becomes dominant through constitutive signaling by overexpressed ERBB2, resulting in high levels of UCP2 that contribute mitochondrial uncoupling. By contrast, ligand stimulation of non-overexpressed ERBB2 transiently removes UCP2 and paradoxically reduces the mitochondrial membrane potential, oxygen consumption, and OXPHOS on a signaling time scale. However, neither the transporter activity nor down-regulation of already low UCP2 levels drive this reduction in mitochondrial activity. Instead, UCP2 is required to establish mitochondria that are capable of responding to ligand. UCP2 knockdown impairs proliferation at high glucose but its absence specifically impairs ligand-induced growth when glucose levels fluctuate. These findings demonstrate the ability of growth factor signaling to control oxidative phosphorylation on a signaling time scale and point toward a non-transporter role for low levels of UCP2 in establishing dynamic response capability.
Collapse
Affiliation(s)
- Nirav Patel
- From the Department of Biochemistry and Molecular Biology
| | | | | |
Collapse
|
59
|
Seemann I, te Poele JAM, Song JY, Hoving S, Russell NS, Stewart FA. Radiation- and anthracycline-induced cardiac toxicity and the influence of ErbB2 blocking agents. Breast Cancer Res Treat 2013; 141:385-95. [PMID: 24091769 PMCID: PMC3824562 DOI: 10.1007/s10549-013-2707-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
In Her2-positive breast cancer patients, inhibition of epidermal growth factor receptor 2 (ErbB2)-signaling is often combined with chemotherapy and radiotherapy. The risk of cardiac toxicity after anthracyclines and radiotherapy is recognized, but little is known about increased risk when these treatments are combined with ErbB2 inhibition. This study investigated whether ErbB2 inhibition increased radiation or anthracycline-induced toxicity. In an in vitro study, human cardiomyocytes were treated with irradiation or doxorubicin, alone or in combination with trastuzumab, and evaluated for cell survival and growth. Groups of mice received 0 or 14 Gy to the heart, alone or in combination with lapatinib, or 3 × 4 mg/kg doxorubicin alone or in combination with lapatinib. Mice were evaluated 40 weeks after treatment for cardiac damage. Changes in cardiac function ((99m)Tc-Myoview gated SPECT) were related to histomorphology and microvascular damage. Radiation or doxorubicin-induced cardiomyocyte toxicity (in vitro) were not exacerbated by trastuzumab. Cardiac irradiation of mice decreased microvascular density (MVD) and increased endothelial damage in surviving capillaries (decrease alkaline phosphatase expression and increased von Willebrand factor), but these changes were not exacerbated by lapatinib. Inflammatory responses in the irradiated epicardium (CD45+ and F4/80+ cells) were significantly reduced in combination with lapatinib. Irradiation, doxorubicin, and lapatinib each induced cardiac fibrosis but this was not further enhanced when treatments were combined. At the ultra-structural level, both lapatinib and doxorubicin induced mitochondrial damage, which was enhanced in combined treatments. Lapatinib alone also induced mild changes in cardiac function but this was not enhanced in the combined treatments. Trastuzumab did not enhance direct radiation or anthracycline toxicity of cardiomyocytes in vitro. Lapatinib did not enhance the risk of radiation or anthracycline-induced cardiac toxicity in mice up to 40 weeks after treatment, but mitochondrial damage was more severe after doxorubicin combined with lapatinib.
Collapse
Affiliation(s)
- Ingar Seemann
- Division of Biological Stress Response (H3), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
60
|
Mendes-Ferreira P, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Therapeutic potential of neuregulin-1 in cardiovascular disease. Drug Discov Today 2013; 18:836-42. [DOI: 10.1016/j.drudis.2013.01.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/16/2013] [Accepted: 01/28/2013] [Indexed: 11/29/2022]
|
61
|
Sridharan V, Sharma SK, Moros EG, Corry PM, Tripathi P, Lieblong BJ, Guha C, Hauer-Jensen M, Boerma M. Effects of radiation on the epidermal growth factor receptor pathway in the heart. Int J Radiat Biol 2013; 89:539-47. [PMID: 23488537 PMCID: PMC3700655 DOI: 10.3109/09553002.2013.782110] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Radiation-induced heart disease (RIHD) is a serious side-effect of thoracic radiotherapy. The epidermal growth factor receptor (EGFR) pathway is essential for the function and survival of cardiomyocytes. Hence, agents that target the EGFR pathway are cardiotoxic. Tocotrienols protect from radiation injury, but may also enhance the therapeutic effects of EGFR pathway inhibitors in cancer treatment. This study investigated the effects of local irradiation on the EGFR pathway in the heart and tests whether tocotrienols may modify radiation-induced changes in this pathway. METHODS Male Sprague-Dawley rats received image-guided localized heart irradiation with 21 Gy. Twenty four hours before irradiation, rats received a single dose of tocotrienol-enriched formulation or vehicle by oral gavage. At time points from 2 h to 9 months after irradiation, left ventricular expression of EGFR pathway mediators was studied. RESULTS Irradiation caused a decrease in the expression of epidermal growth factor (EGF) and neuregulin-1 (Nrg-1) mRNA from 6 h up to 10 weeks, followed by an upregulation of these ligands and the receptor erythroblastic leukemia viral oncogene homolog (ErbB)4 at 6 months. In addition, the upregulation of Nrg-1 was statistically significant up to 9 months after irradiation. A long-term upregulation of ErbB2 protein did not coincide with changes in transcription or post-translational interaction with the chaperone heat shock protein 90 (HSP90). Pretreatment with tocotrienols prevented radiation-induced changes at 2 weeks. CONCLUSIONS Local heart irradiation causes long-term changes in the EGFR pathway. Studies have to address how radiation may interact with cardiotoxic effects of EGFR inhibitors.
Collapse
Affiliation(s)
- Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas 72205, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
|
63
|
Akhtar S, Benter IF. The role of epidermal growth factor receptor in diabetes-induced cardiac dysfunction. BIOIMPACTS : BI 2013; 3:5-9. [PMID: 23678464 DOI: 10.5681/bi.2013.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 02/06/2023]
Abstract
The incidence of diabetes mellitus is increasing rapidly and set to reach near epidemic proportions with the latest estimates suggesting that by 2030 there will be over 550 million people with this debilitating disease. Cardiovascular complications and dysfunctions are three- to eight-folds more likely in diabetic patients and are major causes of increased mortality. The exact underlying mechanisms for the development of complications of the diabetic heart are poorly understood and may involve multiple signaling pathways that are affected by hyperglycemia. This focused article reviews the recent evidence for a possible dual role of epidermal growth factor receptor signaling in diabetes-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Saghir Akhtar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait
| | | |
Collapse
|
64
|
Abstract
Studies in genetically modified mice have demonstrated that neuregulin-1 (NRG-1), along with the erythroblastic leukemia viral oncogene homolog (ErbB) 2, 3, and 4 receptor tyrosine kinases, is necessary for multiple aspects of cardiovascular development. These observations stimulated in vitro and in vivo animal studies, implicating NRG-1/ErbB signaling in the regulation of cardiac cell biology throughout life. Cardiovascular effects of ErbB2-targeted cancer therapies provide evidence in humans that ErbB signaling plays a role in the maintenance of cardiac function. These and other studies suggest a conceptual model in which a key function of NRG-1/ErbB signaling is to mediate adaptations of the heart to physiological and pathological stimuli through activation of intracellular kinase cascades that regulate tissue plasticity. Recent work implicates NRG-1/ErbB signaling in the regulation of multiple aspects of cardiovascular biology, including angiogenesis, blood pressure, and skeletal muscle responses to exercise. The therapeutic potential of recombinant NRG-1 as a potential treatment for heart failure has been demonstrated in animal models and is now being explored in clinical studies. NRG-1 is found in human serum and plasma, and it correlates with some clinical parameters, suggesting that it may have value as an indicator of prognosis. In this review, we bring together this growing literature on NRG-1 and its significance in cardiovascular development and disease.
Collapse
Affiliation(s)
- Oghenerukevwe Odiete
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | |
Collapse
|
65
|
Pentassuglia L, Sawyer DB. ErbB/integrin signaling interactions in regulation of myocardial cell-cell and cell-matrix interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:909-16. [PMID: 23261977 DOI: 10.1016/j.bbamcr.2012.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 12/02/2012] [Accepted: 12/03/2012] [Indexed: 12/17/2022]
Abstract
Neuregulin (Nrg)/ErbB and integrin signaling pathways are critical for the normal function of the embryonic and adult heart. Both systems activate several downstream signaling pathways, with different physiological outputs: cell survival, fibrosis, excitation-contraction coupling, myofilament structure, cell-cell and cell-matrix interaction. Activation of ErbB2 by Nrg1β in cardiomycytes or its overexpression in cancer cells induces phosphorylation of FAK (Focal Adhesion Kinase) at specific sites with modulation of survival, invasion and cell-cell contacts. FAK is also a critical mediator of integrin receptors, converting extracellular matrix alterations into intracellular signaling. Systemic FAK deletion is lethal and is associated with left ventricular non-compaction whereas cardiac restriction in adult hearts is well tolerated. Nevertheless, these hearts are more susceptible to stress conditions like trans-aortic constriction, hypertrophy, and ischemic injury. As FAK is both downstream and specifically activated by integrins and Nrg-1β, here we will explore the role of FAK in the heart as a protective factor and as possible mediator of the crosstalk between the ErbB and Integrin receptors. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
|
66
|
Affiliation(s)
- Gregory M Cote
- Massachusetts General Hospital Cancer Center, Boston, USA
| | | | | |
Collapse
|
67
|
Vasti C, Witt H, Said M, Sorroche P, García-Rivello H, Ruiz-Noppinger P, Hertig CM. Doxorubicin and NRG-1/erbB4-Deficiency Affect Gene Expression Profile: Involving Protein Homeostasis in Mouse. ISRN CARDIOLOGY 2012; 2012:745185. [PMID: 22970387 PMCID: PMC3437290 DOI: 10.5402/2012/745185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/01/2012] [Indexed: 12/17/2022]
Abstract
The accumulating evidence demonstrates the essential role of neuregulin-1 signaling in the adult heart, and, moreover, indicates that an impaired neuregulin signaling exacerbates the doxorubicin-mediated cardiac toxicity. Despite this strong data, the specific cardiomyocyte targets of the active erbB2/erbB4 heterodimer remain unknown. In this paper, we examined pathways involved in cardiomyocyte damage as a result of the cardiac sensitization to anthracycline toxicity in the ventricular muscle-specific erbB4 knockout mouse. We performed morphological analyses to evaluate the ventricular remodeling and employed a cDNA microarray to assess the characteristic gene expression profile, verified data by real-time RT-PCR, and then grouped into functional categories and pathways. We confirm the upregulation of genes related to the classical signature of a hypertrophic response, implicating an erbB2-dependent mechanism in doxorubicin-treated erbB4-KO hearts. Our results indicate the remarkable downregulation of IGF-I/PI-3′ kinase pathway and extends our current knowledge by uncovering an altered ubiquitin-proteasome system leading to cardiomyocyte autophagic vacuolization.
Collapse
Affiliation(s)
- Cecilia Vasti
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-(INGEBI), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina
| | | | | | | | | | | | | |
Collapse
|
68
|
Wadugu B, Kühn B. The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol 2012; 302:H2139-47. [PMID: 22427524 DOI: 10.1152/ajpheart.00063.2012] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The signaling complex consisting of the growth factor neuregulin-1 (NRG1) and its tyrosine kinase receptors ErbB2 and ErbB4 has a critical role in cardiac development and homeostasis of the structure and function of the adult heart. Recent research results suggest that targeting this signaling complex may provide a viable strategy for treating heart failure. Clinical trials are currently evaluating the effectiveness and safety of intravenous administration of recombinant NRG1 formulations in heart failure patients. Endogenous as well as administered NRG1 has multiple possible activities in the adult heart, but how these are related is unknown. It has recently been demonstrated that NRG1 administration can stimulate proliferation of cardiomyocytes, which may contribute to repair failing hearts. This review summarizes the current knowledge of how NRG1 and its receptors control cardiac physiology and biology, with special emphasis on its role in cardiomyocyte proliferation during myocardial growth and regeneration.
Collapse
Affiliation(s)
- Brian Wadugu
- Department of Cardiology, Children's Hospital Boston, Massachusetts, USA
| | | |
Collapse
|
69
|
Paatero I, Jokilammi A, Heikkinen PT, Iljin K, Kallioniemi OP, Jones FE, Jaakkola PM, Elenius K. Interaction with ErbB4 promotes hypoxia-inducible factor-1α signaling. J Biol Chem 2012; 287:9659-9671. [PMID: 22308027 DOI: 10.1074/jbc.m111.299537] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The receptor-tyrosine kinase ErbB4 was identified as a direct regulator of hypoxia-inducible factor-1α (HIF-1α) signaling. Cleaved intracellular domain of ErbB4 directly interacted with HIF-1α in the nucleus, and stabilized HIF-1α protein in both normoxic and hypoxic conditions by blocking its proteasomal degradation. The mechanism of HIF stabilization was independent of VHL and proline hydroxylation but dependent on RACK1. ErbB4 activity was necessary for efficient HRE-driven promoter activity, transcription of known HIF-1α target genes, and survival of mammary carcinoma cells in vitro. In addition, mammary epithelial specific targeting of Erbb4 in the mouse significantly reduced the amount of HIF-1α protein in vivo. ERBB4 expression also correlated with the expression of HIF-regulated genes in a series of 4552 human normal and cancer tissue samples. These data demonstrate that soluble ErbB4 intracellular domain promotes HIF-1α stability and signaling via a novel mechanism.
Collapse
Affiliation(s)
- Ilkka Paatero
- Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland,; Turku Doctoral Programme of Biomedical Sciences, FI-20520 Turku, Finland
| | - Anne Jokilammi
- Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland
| | - Pekka T Heikkinen
- Turku Doctoral Programme of Biomedical Sciences, FI-20520 Turku, Finland; Turku Centre for Biotechnology, FI-20520 Turku, Finland
| | - Kristiina Iljin
- Turku Centre for Biotechnology, FI-20520 Turku, Finland; Medical Biotechnology, VTT Technical Research Centre, FI-20520 Turku, Finland
| | - Olli-Pekka Kallioniemi
- Turku Centre for Biotechnology, FI-20520 Turku, Finland; Medical Biotechnology, VTT Technical Research Centre, FI-20520 Turku, Finland,; FIMM - Institute for Molecular Medicine Finland, and the Genome-Scale Biology Research Program, Biomedicum, University of Helsinki, FI-00014 Helsinki, Finland
| | - Frank E Jones
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118, and
| | - Panu M Jaakkola
- Turku Centre for Biotechnology, FI-20520 Turku, Finland; Department of Oncology, Turku University Hospital, FI-20520 Turku, Finland
| | - Klaus Elenius
- Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland,; Department of Oncology, Turku University Hospital, FI-20520 Turku, Finland.
| |
Collapse
|
70
|
Gui C, Zhu L, Hu M, Lei L, Long Q. Neuregulin-1/ErbB signaling is impaired in the rat model of diabetic cardiomyopathy. Cardiovasc Pathol 2012; 21:414-20. [PMID: 22285193 DOI: 10.1016/j.carpath.2011.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 12/13/2011] [Accepted: 12/16/2011] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCP) is one of the leading causes of increased morbidity and mortality in the diabetic population. The neuregulin-1(NRG1)/ErbB signal system plays a critical role in maintenance of adult heart function. But little is known about the changes of NRG1/ErbB signal system in DCP. The aim of this study was to investigate the changes of the NRG1/ErbB signal system in DCP. METHODS A rat model of DCP was established using a single intraperitoneal injection of streptozotocin (STZ). Cardiac function was assessed using echocardiography. The left ventricle fibrosis was evaluated using Masson's trichrome staining. The mRNA expression profiles of ErbB2 and ErbB4 receptors were evaluated using real-time polymerase chain reaction. The protein expression of NRG1 and the phosphorylation of ErbB2 and ErbB4 receptors were assessed using Western blot analysis. RESULTS The results showed dramatic left ventricle fibrosis and impaired left ventricle systolic function at 12 weeks after STZ-induced diabetes. This study also showed that ErbB2 and ErbB4 mRNA expression and NRG1 protein expression in the left ventricular myocardium were significantly decreased. In addition, we observed decreased phosphorylation of the ErbB2 and ErbB4 receptors at 12 weeks after the induction of diabetes. CONCLUSIONS These findings suggest that NRG1/ErbB signaling is impaired in DCP, which may play some roles in the pathogenesis of DCP.
Collapse
Affiliation(s)
- Chun Gui
- Department of Cardiology, the First Affiliated Hospital, Guangxi Medical University, Nanning Guangxi, People's Republic of China.
| | | | | | | | | |
Collapse
|
71
|
Icli B, Bharti A, Pentassuglia L, Peng X, Sawyer DB. ErbB4 localization to cardiac myocyte nuclei, and its role in myocyte DNA damage response. Biochem Biophys Res Commun 2012; 418:116-21. [PMID: 22244893 DOI: 10.1016/j.bbrc.2011.12.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 12/28/2011] [Indexed: 01/04/2023]
Abstract
The intracellular domain of ErbB4 receptor tyrosine kinase is known to translocate to the nucleus of cells where it can regulate p53 transcriptional activity. The purpose of this study was to examine whether ErbB4 can localize to the nucleus of adult rat ventricular myocytes (ARVM), and regulate p53 in these cells. We demonstrate that ErbB4 does locate to the nucleus of cardiac myocytes as a full-length protein, although nuclear location occurs as a full-length protein that does not require Protein Kinase C or γ-secretase activity. Consistent with this we found that only the non-cleavable JM-b isoform of ErbB4 is expressed in ARVM. Doxorubicin was used to examine ErbB4 role in regulation of a DNA damage response in ARVM. Doxorubicin induced p53 and p21 was suppressed by treatment with AG1478, an EGFR and ErbB4 kinase inhibitor, or suppression of ErbB4 expression with small interfering RNA. Thus ErbB4 localizes to the nucleus as a full-length protein, and plays a role in the DNA damage response induced by doxorubicin in cardiac myocytes.
Collapse
Affiliation(s)
- Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
72
|
Neuregulin 1 regulates excitability of fast-spiking neurons through Kv1.1 and acts in epilepsy. Nat Neurosci 2011; 15:267-73. [DOI: 10.1038/nn.3006] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 11/07/2011] [Indexed: 02/08/2023]
|
73
|
Tan GH, Liu YY, Hu XL, Yin DM, Mei L, Xiong ZQ. Neuregulin 1 represses limbic epileptogenesis through ErbB4 in parvalbumin-expressing interneurons. Nat Neurosci 2011; 15:258-66. [DOI: 10.1038/nn.3005] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 11/07/2011] [Indexed: 02/08/2023]
|
74
|
Hedhli N, Huang Q, Kalinowski A, Palmeri M, Hu X, Russell RR, Russell KS. Endothelium-derived neuregulin protects the heart against ischemic injury. Circulation 2011; 123:2254-62. [PMID: 21555713 DOI: 10.1161/circulationaha.110.991125] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Removal of cardiac endothelial cells (EC) has been shown to produce significant detrimental effects on the function of adjacent cardiac myocytes, suggesting that EC play a critical role in autocrine/paracrine regulation of the heart. Despite this important observation, the mediators of the protective function of EC remain obscure. Neuregulin (NRG, a member of the epidermal growth factor family) is produced by EC and cardiac myocytes contain receptors (erbB) for this ligand. We hypothesized that NRG is an essential factor produced by EC, which promotes cardioprotection against ischemic injury. METHODS AND RESULTS We demonstrate that human cardiac EC express and release NRG in response to hypoxia-reoxygenation. Under conditions where hypoxia--reoxygenation causes significant cardiac myocyte cell death, NRG can significantly decrease apoptosis of isolated adult ventricular myocytes. Coculturing adult murine myocytes with human umbilical vein, murine lung microvascular, or human coronary artery EC can also protect myocytes against hypoxia--reoxygenation--induced apoptosis. These protective effects are abolished by NRG gene deletion or silencing of NRG expression in EC. Finally, endothelium-selective deletion of NRG in vivo leads to significantly decreased tolerance to ischemic insult, as demonstrated by impaired postischemic contractile recovery in a perfused whole-organ preparation and larger infarct sizes after coronary artery ligation. CONCLUSION Together, these data demonstrate that EC-derived NRG plays an important role in cardiac myocyte protection against ischemic injury in the heart and supports the idea that manipulation of this signaling pathway may be an important clinical target in this setting.
Collapse
Affiliation(s)
- Nadia Hedhli
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Neuregulin 1 promotes excitatory synapse development and function in GABAergic interneurons. J Neurosci 2011; 31:15-25. [PMID: 21209185 DOI: 10.1523/jneurosci.2538-10.2011] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neuregulin 1 (NRG1) and its receptor ErbB4 are both susceptibility genes of schizophrenia. However, little is known about the underlying mechanisms of their malfunction. Although ErbB4 is enriched in GABAergic interneurons, the role of NRG1 in excitatory synapse formation in these neurons remains poorly understood. We showed that NRG1 increased both the number and size of PSD-95 puncta and the frequency and amplitude of miniature EPSCs (mEPSCs) in GABAergic interneurons, indicating that NRG1 stimulates the formation of new synapses and strengthens existing synapses. In contrast, NRG1 treatment had no effect on either the number or size of excitatory synapses in glutamatergic neurons, suggesting its synaptogenic effect is specific to GABAergic interneurons. Ecto-ErbB4 treatment diminished both the number and size of excitatory synapses, suggesting that endogenous NRG1 may be critical for basal synapse formation. NRG1 could stimulate the stability of PSD-95 in the manner that requires tyrosine kinase activity of ErbB4. Finally, deletion of ErbB4 in parvalbumin-positive interneurons led to reduced frequency and amplitude of mEPSCs, providing in vivo evidence that ErbB4 is important in excitatory synaptogenesis in interneurons. Together, our findings suggested a novel synaptogenic role of NRG1 in excitatory synapse development, possibly via stabilizing PSD-95, and this effect is specific to GABAergic interneurons. In light of the association of the genes of both NRG1 and ErbB4 with schizophrenia and dysfunction of GABAergic system in this disorder, these results provide insight into its potential pathological mechanism.
Collapse
|
76
|
Campreciós G, Lorita J, Pardina E, Peinado-Onsurbe J, Soley M, Ramírez I. Expression, localization, and regulation of the neuregulin receptor ErbB3 in mouse heart. J Cell Physiol 2011; 226:450-5. [PMID: 20672328 DOI: 10.1002/jcp.22354] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuregulins (NRG) belong to the EGF family of growth factors, which are ligands of the ErbB receptors. Their expression in the adult heart is essential, especially when the heart is submitted to cardiotoxic stress such as that produced by anthracyclines. It is considered that ErbB4 is the only NRG receptor expressed by the adult heart. Upon binding, ErbB4 may dimerize with ErbB2 to generate signals inside cells. However, here we show the presence of ErbB3 in the mouse heart from birth to adulthood by Western blotting and real-time RT-PCR. The expression level of ErbB3 mRNA was lower than that of ErbB2 or ErbB4, but was more stable throughout postnatal development. In isolated heart myocytes, ErbB3 localized to the Z-lines similarly to ErbB1. Perfusion of isolated hearts with NRG-1β induced phosphorylation of ErbB3, as well as ErbB2 and ErbB4. In adult mice, both ErbB2 and ErbB3, but not ErbB1 or ErbB4, were rapidly down-regulated upon the induction of heart hypertrophy. In conclusion, our results demonstrate that ErbB3, in addition to ErbB4, is a receptor for neuregulin-1β in the adult mouse heart.
Collapse
Affiliation(s)
- Genís Campreciós
- Faculty of Biology, Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Catalonia, Spain
| | | | | | | | | | | |
Collapse
|
77
|
Lemmens K, Doggen K, De Keulenaer GW. Activation of the neuregulin/ErbB system during physiological ventricular remodeling in pregnancy. Am J Physiol Heart Circ Physiol 2010; 300:H931-42. [PMID: 21186272 DOI: 10.1152/ajpheart.00385.2010] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The neuregulin-1 (NRG1)/ErbB system has emerged as a paracrine endothelium-controlled system in the heart, which preserves left ventricular (LV) performance in pathophysiological conditions. Here, we analyze the activity and function of this system in pregnancy, which imparts a physiological condition of LV hemodynamic overload. NRG1 expression and ErbB receptor activation were studied by Western blot analyses in rats and mice at different stages of pregnancy. LV performance was evaluated by transthoracic echocardiography, and myocardial performance was assessed from twitches of isolated papillary muscles. NRG1/ErbB signaling was inhibited by oral treatment of animals with the dual ErbB1/ErbB2 tyrosine kinase inhibitor lapatinib. Analyses of LV tissue revealed that protein expression of different NRG1 isoforms and levels of phosphorylated ErbB2 and ErbB4 significantly increased after 1-2 wk of pregnancy. Lapatinib prevented phosphorylation of ErbB2 and ERK1/2, but not of ErbB4 and protein kinase B (Akt), revealing that lapatinib only partially inhibited NRG1/ErbB signaling in the LV. Lapatinib did not prevent pregnancy-induced changes in LV mass and did not cause apoptotic cell death or fibrosis in the LV. Nevertheless, lapatinib led to premature maternal death of ∼25% during pregnancy and it accentuated pregnancy-induced LV dilatation, significantly reduced LV fractional shortening, and induced abnormalities of twitch relaxation (but not twitch amplitude) of isolated papillary muscles. This is the first study showing that the NRG1/ErbB system is activated, and plays a modulatory role, during physiological hemodynamic overload associated with pregnancy. Inhibiting this system during physiological overload may cause LV dysfunction in the absence of myocardial cell death.
Collapse
Affiliation(s)
- Katrien Lemmens
- Laboratory of Physiology, University of Antwerp, Antwerp, Belgium.
| | | | | |
Collapse
|
78
|
ErbB4 in parvalbumin-positive interneurons is critical for neuregulin 1 regulation of long-term potentiation. Proc Natl Acad Sci U S A 2010; 107:21818-23. [PMID: 21106764 DOI: 10.1073/pnas.1010669107] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neuregulin 1 (NRG1) is a trophic factor that acts by stimulating ErbB receptor tyrosine kinases and has been implicated in neural development and synaptic plasticity. In this study, we investigated mechanisms of its suppression of long-term potentiation (LTP) in the hippocampus. We found that NRG1 did not alter glutamatergic transmission at SC-CA1 synapses but increased the GABA(A) receptor-mediated synaptic currents in CA1 pyramidal cells via a presynaptic mechanism. Inhibition of GABA(A) receptors blocked the suppressing effect of NRG1 on LTP and prevented ecto-ErbB4 from enhancing LTP, implicating a role of GABAergic transmission. To test this hypothesis further, we generated parvalbumin (PV)-Cre;ErbB4(-/-) mice in which ErbB4, an NRG1 receptor in the brain, is ablated specifically in PV-positive interneurons. NRG1 was no longer able to increase inhibitory postsynaptic currents and to suppress LTP in PV-Cre;ErbB4(-/-) hippocampus. Accordingly, contextual fear conditioning, a hippocampus-dependent test, was impaired in PV-Cre;ErbB4(-/-) mice. In contrast, ablation of ErbB4 in pyramidal neurons had no effect on NRG1 regulation of hippocampal LTP or contextual fear conditioning. These results demonstrate a critical role of ErbB4 in PV-positive interneurons but not in pyramidal neurons in synaptic plasticity and support a working model that NRG1 suppresses LTP by enhancing GABA release. Considering that NRG1 and ErbB4 are susceptibility genes of schizophrenia, these observations contribute to a better understanding of how abnormal NRG1/ErbB4 signaling may be involved in the pathogenesis of schizophrenia.
Collapse
|
79
|
Sanchez-Soria P, Camenisch TD. ErbB signaling in cardiac development and disease. Semin Cell Dev Biol 2010; 21:929-35. [PMID: 20933094 DOI: 10.1016/j.semcdb.2010.09.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 09/24/2010] [Accepted: 09/28/2010] [Indexed: 01/01/2023]
Abstract
The ErbB family of receptor tyrosine kinases (RTKs) is a family of receptors that allow cells to interact with the extracellular environment and transduce signals to the nucleus that promote differentiation, migration and proliferation necessary for proper heart morphogenesis and function. This review focuses on the role of the ErbB family of receptor tyrosine kinases, and their importance in proper heart morphogenesis, as well as their role in maintenance and function of the adult heart. Studies from transgenic mouse models have shown the importance of ErbB receptors in heart development, and provide insight into potential future therapeutic targets to help reduce congenital heart defect (CHD) mortality rates and prevent disease in adults. Cancer therapeutics have also shed light to the ErbB receptors and signaling network, as undesired side effects have demonstrated their importance in adult cardiomyocytes and prevention of cardiomyopathies. This review will discuss ErbB receptor tyrosine kinases (RTK) in heart development and disease including valve formation and partitioning of a four-chambered heart as well as cardiotoxicity when ErbB signaling is attenuated in adults.
Collapse
Affiliation(s)
- Pablo Sanchez-Soria
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207, USA.
| | | |
Collapse
|
80
|
Abstract
Neuregulin-1 (NRG-1), a ligand of receptor tyrosine kinases of the ErbB family, plays a critical role in cardiovascular development and maintenance of adult heart function. Results from cellular, animal, and clinical experiments have shown NRG-1 to be a promising drug candidate for restoring cardiac function after cardiac injury. Various mechanisms have been suggested to be involved in this process, such as improving sarcomeric structure or cell-cell adhesion, promoting proliferation and survival of cardiac myocytes, balancing Ca(2+) homeostasis, modulating inotropic effects, promoting angiogenesis, and preventing atherosclerosis. However, the contribution of these effects to the restoration of cardiac function remains to be estimated, and it may depend on the specific events that led to heart failure. Meanwhile, distinct and crossed signaling pathways downstream of NRG-1 may play a role in these underlying mechanisms, resulting in a complicated network of signaling mediating the function of NRG-1.
Collapse
Affiliation(s)
- Zhenggang Jiang
- Zensun (Shanghai) Sci & Tech Ltd, No. 68 Ju Li Road, Zhangjiang Hi-Tech Park, Pudong District, Shanghai, 201203, China
| | | |
Collapse
|
81
|
Brero A, Ramella R, Fitou A, Dati C, Alloatti G, Gallo MP, Levi R. Neuregulin-1beta1 rapidly modulates nitric oxide synthesis and calcium handling in rat cardiomyocytes. Cardiovasc Res 2010; 88:443-52. [PMID: 20634213 DOI: 10.1093/cvr/cvq238] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS The ErbB-neuregulin-1β1 (Nrg1β1) pathway is required for cardiac development and exerts chronic effects on the postnatal adult heart. Long-term application of Nrg1β1 results in hypertrophy and protection against oxidative stress and cytotoxic agents. We performed experiments with acute Nrg1β1 treatment to find evidence for a further protective role due to rapid modulation of adult cardiomyocyte function. METHODS AND RESULTS In confocal fluorimetric measurements, Nrg1β1 induced a calcium-independent increase in nitric oxide (NO) production in isolated adult rat ventricular myocytes (ARVCMs) that was blocked by the phosphoinositide-3-kinase (PI3K) inhibitor Wortmannin. Western blot analysis showed enhancement of endothelial nitric oxide synthase phosphorylation in Nrg1β1-treated ARVCMs, which was attenuated by Wortmannin. Nrg1β1 induced a significant increase in calcium transient amplitude (indo-1 ratiometric measurement) and accelerated the recovery of cytosolic calcium in the sarcoplasmic reticulum without affecting whole-cell L-type calcium current. Wortmannin or the protein kinase G inhibiting peptide (DT-2) abolished the increase in calcium transient amplitude and the acceleration of calcium recovery induced by Nrg1β1 treatment. Immunofluorescence analysis revealed that Nrg1β1 treatment increased phospholamban phosphorylation, and the effect was blocked by PI3K and protein kinase G inhibition. Caffeine-releasable sarcoplasmic reticulum calcium content was also higher during Nrg1β1 administration. CONCLUSION Rapid activation of PI3K, endothelial nitric oxide synthase and protein kinase G and a consequent improvement in diastolic calcium can be added to established Nrg1 protective roles.
Collapse
Affiliation(s)
- Alessia Brero
- Department of Animal and Human Biology, University of Turin, Via Accademia Albertina 13, 10123, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
82
|
Horie T, Ono K, Nishi H, Nagao K, Kinoshita M, Watanabe S, Kuwabara Y, Nakashima Y, Takanabe-Mori R, Nishi E, Hasegawa K, Kita T, Kimura T. Acute doxorubicin cardiotoxicity is associated with miR-146a-induced inhibition of the neuregulin-ErbB pathway. Cardiovasc Res 2010; 87:656-64. [PMID: 20495188 PMCID: PMC2920811 DOI: 10.1093/cvr/cvq148] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aims A significant increase in congestive heart failure (CHF) was reported when the anti-ErbB2 antibody trastuzumab was used in combination with the chemotherapy drug doxorubicin (Dox). The aim of the present study was to investigate the role(s) of miRNAs in acute Dox-induced cardiotoxicity. Methods and results Neuregulin-1-ErbB signalling is essential for maintaining adult cardiac function. We found a significant reduction in ErbB4 expression in the hearts of mice after Dox treatment. Because the proteasome pathway was only partially involved in the reduction of ErbB4 expression, we examined the involvement of microRNAs (miRs) in the reduction of ErbB4 expression. miR-146a was shown to be up-regulated by Dox in neonatal rat cardiac myocytes. Using a luciferase reporter assay and overexpression of miR-146a, we confirmed that miR-146a targets the ErbB4 3′UTR. After Dox treatment, overexpression of miR-146a, as well as that of siRNA against ErbB4, induced cell death in cardiomyocytes. Re-expression of ErbB4 in miR-146a-overexpressing cardiomyocytes ameliorated Dox-induced cell death. To examine the loss of miR-146a function, we constructed ‘decoy’ genes that had tandem complementary sequences for miR-146a in the 3′UTR of a luciferase gene. When miR-146a ‘decoy’ genes were introduced into cardiomyocytes, ErbB4 expression was up-regulated and Dox-induced cell death was reduced. Conclusion These findings suggested that the up-regulation of miR-146a after Dox treatment is involved in acute Dox-induced cardiotoxicity by targeting ErbB4. Inhibition of both ErbB2 and ErbB4 signalling may be one of the reasons why those patients who receive concurrent therapy with Dox and trastuzumab suffer from CHF.
Collapse
Affiliation(s)
- Takahiro Horie
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
- Corresponding author. Tel: +81 75 751 3190, Fax: +81 75 751 3203,
| | - Hitoo Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuya Nagao
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Minako Kinoshita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin Watanabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhide Kuwabara
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiro Nakashima
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Rieko Takanabe-Mori
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto 612-8555, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koji Hasegawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization, Kyoto 612-8555, Japan
| | - Toru Kita
- Kobe City Medical Center General Hospital, Kobe, Hyogo 650-0046, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
83
|
Wen L, Lu YS, Zhu XH, Li XM, Woo RS, Chen YJ, Yin DM, Lai C, Terry AV, Vazdarjanova A, Xiong WC, Mei L. Neuregulin 1 regulates pyramidal neuron activity via ErbB4 in parvalbumin-positive interneurons. Proc Natl Acad Sci U S A 2010; 107:1211-1216. [PMID: 20080551 PMCID: PMC2824309 DOI: 10.1073/pnas.0910302107] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neuregulin 1 (NRG1) is a trophic factor thought to play a role in neural development. Recent studies suggest that it may regulate neurotransmission, mechanisms of which remain elusive. Here we show that NRG1, via stimulating GABA release from interneurons, inhibits pyramidal neurons in the prefrontal cortex (PFC). Ablation of the NRG1 receptor ErbB4 in parvalbumin (PV)-positive interneurons prevented NRG1 from stimulating GABA release and from inhibiting pyramidal neurons. PV-ErbB4(-/-) mice exhibited schizophrenia-relevant phenotypes similar to those observed in NRG1 or ErbB4 null mutant mice, including hyperactivity, impaired working memory, and deficit in prepulse inhibition (PPI) that was ameliorated by diazepam, a GABA enhancer. These results indicate that NRG1 regulates the activity of pyramidal neurons by promoting GABA release from PV-positive interneurons, identifying a critical function of NRG1 in balancing brain activity. Because both NRG1 and ErbB4 are susceptibility genes of schizophrenia, our study provides insight into potential pathogenic mechanisms of schizophrenia and suggests that PV-ErbB4(-/-) mice may serve as a model in the study of this and relevant brain disorders.
Collapse
Affiliation(s)
- Lei Wen
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Yi-Sheng Lu
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Xin-Hong Zhu
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Xiao-Ming Li
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Ran-Sook Woo
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Yong-Jun Chen
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Dong-Min Yin
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Cary Lai
- Department of Psychological and Brain Sciences and Program in Neuroscience, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405
| | | | - Almira Vazdarjanova
- Synapses and Cognitive Neuroscience Center and Department of Neurology, Medical College of Georgia, Augusta, GA 30912; and
| | - Wen-Cheng Xiong
- Institute of Molecular Medicine and Genetics and Department of Neurology
| | - Lin Mei
- Institute of Molecular Medicine and Genetics and Department of Neurology
| |
Collapse
|
84
|
Neuregulin-1/ErbB signaling and chronic heart failure. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 59:31-51. [PMID: 20933198 DOI: 10.1016/s1054-3589(10)59002-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuregulin-1 (NRG-1), a cardioactive growth factor released from endothelial cells, is indispensable for cardiac development, structural maintenance, and functional integrity of the heart. In recent years, a growing number of studies have focused on NRG-1 and members of the ErbB family that serve as receptors for NRG-1 in order to better understand the role of this signaling pathway in physiology and pathophysiology of the heart. An essential role for NRG-1 and ErbB in heart development and functionality has been suggested by studies in conditional NRG-1/ErbB-deficient mice and by the cardiac-related side effects of anti-ErbB2 antibody therapies used for treatment of breast cancer. In vitro and in vivo studies using recombinant human neuregulin-1 (rhNRG-1), which contains the epidermal growth factor (EGF)-like domain (necessary for ErbB2/ErbB4 activation), have further supported the hypothesis that NRG-1 plays an important role in heart function. Consistent with other studies, expression of rhNRG-1 not only restored normal cardiomyocytic structure altered by nutritional deficiency in cell cultures, but also improved the pumping function of the heart in several animal models of chronic heart failure (CHF). As a result of these findings, proteins involved in the NRG-1/ErbB-signaling pathway have been explored as potential drug targets for treatment of heart failure. Clinical trials to evaluate the safety and efficacy of rhNRG-1 have been conducted in both China and Australia. As predicted, rhNRG-1 treatment improved both cardiac function and reversed remodeling of the heart. Therefore, rhNRG-1 may represent a new drug for treatment of CHF with a novel therapeutic mechanism.
Collapse
|
85
|
Dasgupta C, Sakurai R, Wang Y, Guo P, Ambalavanan N, Torday JS, Rehan VK. Hyperoxia-induced neonatal rat lung injury involves activation of TGF-{beta} and Wnt signaling and is protected by rosiglitazone. Am J Physiol Lung Cell Mol Physiol 2009; 296:L1031-41. [PMID: 19304912 PMCID: PMC3286237 DOI: 10.1152/ajplung.90392.2008] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 03/05/2009] [Indexed: 01/09/2023] Open
Abstract
Despite tremendous technological and therapeutic advances, bronchopulmonary dysplasia (BPD) remains a leading cause of respiratory morbidity in very low birth weight infants, and there are no effective preventive and/or therapeutic options. We have previously reported that hyperoxia-induced neonatal rat lung injury might be prevented by rosiglitazone (RGZ). Here, we characterize 1) perturbations in wingless/Int (Wnt) and transforming growth factor (TGF)-beta signaling, and 2) structural aberrations in lung morphology following 7-day continuous in vivo hyperoxia exposure to neonatal rats. We also tested whether treatment of neonatal pups with RGZ, concomitant to hyperoxia, could prevent such aberrations. Our study revealed that hyperoxia caused significant upregulation of Wnt signaling protein markers lymphoid enhancer factor 1 (Lef-1) and beta-catenin and TGF-beta pathway transducers phosphorylated Smad3 and Smad7 proteins in whole rat lung extracts. These changes were also accompanied by upregulation of myogenic marker proteins alpha-smooth muscle actin (alpha-SMA) and calponin but significant downregulation of the lipogenic marker peroxisome proliferator-activated receptor-gamma (PPARgamma) expression. These molecular perturbations were associated with reduction in alveolar septal thickness, radial alveolar count, and larger alveoli in the hyperoxia-exposed lung. These hyperoxia-induced molecular and morphological changes were prevented by systemic administration of RGZ, with lung sections appearing near normal. This is the first evidence that in vivo hyperoxia induces activation of both Wnt and TGF-beta signal transduction pathways in lung and of its near complete prevention by RGZ. Hyperoxia-induced arrest in alveolar development, a hallmark of BPD, along with these molecular changes strongly implicates these proteins in hyperoxia-induced lung injury. Administration of PPARgamma agonists may thus be a potential strategy to attenuate hyperoxia-induced lung injury and subsequent BPD.
Collapse
Affiliation(s)
- Chiranjib Dasgupta
- Departments of Pediatrics , Harbor-UCLA Medical Center, Los Angeles Biomedical Research Institute at Harbor-UCLA, David Geffen School of Medicine at UCLA, Torrance, California, USA
| | | | | | | | | | | | | |
Collapse
|
86
|
Pentassuglia L, Graf M, Lane H, Kuramochi Y, Cote G, Timolati F, Sawyer DB, Zuppinger C, Suter TM. Inhibition of ErbB2 by receptor tyrosine kinase inhibitors causes myofibrillar structural damage without cell death in adult rat cardiomyocytes. Exp Cell Res 2009; 315:1302-12. [PMID: 19331811 PMCID: PMC4991362 DOI: 10.1016/j.yexcr.2009.02.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 02/01/2009] [Accepted: 02/02/2009] [Indexed: 11/25/2022]
Abstract
Inhibition of ErbB2 (HER2) with monoclonal antibodies, an effective therapy in some forms of breast cancer, is associated with cardiotoxicity, the pathophysiology of which is poorly understood. Recent data suggest, that dual inhibition of ErbB1 (EGFR) and ErbB2 signaling is more efficient in cancer therapy, however, cardiac safety of this therapeutic approach is unknown. We therefore tested an ErbB1-(CGP059326) and an ErbB1/ErbB2-(PKI166) tyrosine kinase inhibitor in an in-vitro system of adult rat ventricular cardiomyocytes and assessed their effects on 1. cell viability, 2. myofibrillar structure, 3. contractile function, and 4. MAPK- and Akt-signaling alone or in combination with Doxorubicin. Neither CGP nor PKI induced cardiomyocyte necrosis or apoptosis. PKI but not CGP caused myofibrillar structural damage that was additive to that induced by Doxorubicin at clinically relevant doses. These changes were associated with an inhibition of excitation-contraction coupling. PKI but not CGP decreased p-Erk1/2, suggesting a role for this MAP-kinase signaling pathway in the maintenance of myofibrils. These data indicate that the ErbB2 signaling pathway is critical for the maintenance of myofibrillar structure and function. Clinical studies using ErbB2-targeted inhibitors for the treatment of cancer should be designed to include careful monitoring for cardiac dysfunction.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Calcium/metabolism
- Cell Death/physiology
- Cells, Cultured
- Connectin
- Doxorubicin/pharmacology
- ErbB Receptors/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- GATA4 Transcription Factor/metabolism
- Male
- Mice
- Mitochondria/metabolism
- Muscle Proteins/metabolism
- Myocardial Contraction/drug effects
- Myocardial Contraction/physiology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myofibrils/metabolism
- Myofibrils/ultrastructure
- Protein Kinase Inhibitors/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Wistar
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-4
- Signal Transduction/physiology
Collapse
Affiliation(s)
| | - Michael Graf
- Swiss Cardiovascular Center, University Hospital, Bern, Switzerland
| | - Heidi Lane
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Yukio Kuramochi
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Gregory Cote
- Boston University Medical Center, Boston, MA, USA
| | | | - Douglas B. Sawyer
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN, USA
| | | | - Thomas M. Suter
- Swiss Cardiovascular Center, University Hospital, Bern, Switzerland
| |
Collapse
|
87
|
Xu Y, Li X, Zhou M. Neuregulin-1/ErbB signaling: a druggable target for treating heart failure. Curr Opin Pharmacol 2008; 9:214-9. [PMID: 19070544 DOI: 10.1016/j.coph.2008.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 11/03/2008] [Accepted: 11/03/2008] [Indexed: 11/28/2022]
Abstract
Neuregulin-1s are widely expressed signaling molecules that are involved in cell differentiation, proliferation, growth, survival, and apoptosis. They transmit their signals by interacting with cell membrane receptors of the ErbB family, resulting in the activation of intracellular signaling cascades, which participate in various physiological and etiological processes. Besides their essential function in the development of the heart and the physiology of cardiac pumping, there is emerging evidence for the involvement of neuregulin-1/ErbB signals in human disease, including heart failure. These reasons prompt the development of new therapeutic agents based on neuregulin-1.
Collapse
Affiliation(s)
- Yabei Xu
- Zensun (Shanghai) Science and Technology Co. Ltd, 328 Bi Bo Road, Zhangjiang Hi-tech Park, Shanghai, PR China
| | | | | |
Collapse
|
88
|
Doggen K, Ray L, Mathieu M, Mc Entee K, Lemmens K, De Keulenaer GW. Ventricular ErbB2/ErbB4 activation and downstream signaling in pacing-induced heart failure. J Mol Cell Cardiol 2008; 46:33-8. [PMID: 19010331 DOI: 10.1016/j.yjmcc.2008.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 10/09/2008] [Accepted: 10/16/2008] [Indexed: 11/25/2022]
Abstract
The neuregulin-1 (NRG-1)/ErbB system has emerged as a cardioprotective system that becomes activated during myocardial stress, most convincingly shown in response to cardiotoxic chemotherapy. Direct evidence of increased ventricular ErbB receptor activity in heart failure unrelated to cardiotoxic drugs is, however, limited. We investigated changes in NRG-1 expression, ErbB receptor phosphorylation and downstream activation of intracellular ErbB targets during rapid pacing and progressive ventricular dysfunction in the dog. Heart failure was induced in dogs by 7 weeks of rapid pacing. Ventricular function was assessed by echocardiography. Messenger RNA expression was investigated in ventricular biopsies using quantitative PCR. Activation of NRG-1/ErbB signaling and of downstream targets was investigated using immunoprecipitation and/or Western blotting. Over the course of 7 weeks of pacing and ventricular dilatation, ventricular levels of NRG-1, but not of other ErbB4 ligands, and of ADAM19, a protease promoting NRG-1 release, progressively increased. In parallel, levels of activated ErbB2 and ErbB4, phosphorylated at tyrosine residues 877/1248 and 1284 respectively, became progressively higher. Similarly, levels of total and phosphorylated PI-3 kinase increased. Surprisingly, however, and in contrast with activation of downstream targets of ErbB receptors in normal hearts, Akt and ERK1/2, remained inactivated. This study shows that ventricular ErbB2 and ErbB4 receptors become activated during the development of pacing-induced heart failure, but that downstream signaling is, at least partly, abrogated. Abrogation of cardioprotective signaling after ErbB activation is an unanticipated phenomenon in the progression of heart failure with possibly major pathophysiological significance. The underlying mechanisms should be further elucidated.
Collapse
Affiliation(s)
- Kris Doggen
- Laboratory of Physiology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
89
|
Pentassuglia L, Sawyer DB. The role of Neuregulin-1beta/ErbB signaling in the heart. Exp Cell Res 2008; 315:627-37. [PMID: 18801360 DOI: 10.1016/j.yexcr.2008.08.015] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Revised: 08/19/2008] [Accepted: 08/19/2008] [Indexed: 01/13/2023]
Abstract
Products of the Neuregulin-1 (Nrg-1) gene, along with the ErbB family of receptor tyrosine kinases through which Nrg-1 ligands signal, play a critical role during cardiovascular development. Through studies of genetically manipulated mice, as well as studies in cells isolated from adult hearts, it appears that Nrg-1/ErbB signaling is an essential paracrine mediator of cell-cell interactions that not only regulates tissue organization during development, but also helps to maintain cardiac function throughout an organism's life. Studies in cells isolated from the heart demonstrate that Nrg-1 can activate a number of signaling pathways, which mediate cellular adaptations to stress in the myocardium. These observations provide insight as to why ErbB2-targeted cancer treatments have deleterious effects on cardiac function in some cancer patients. Moreover emerging data suggest that Nrg-1 ligands might be useful clinically to restore cardiac function after cardiac injury. In this review we will attempt to synthesize the literature behind this rapidly growing and exciting area of research.
Collapse
Affiliation(s)
- Laura Pentassuglia
- Cardiovascular Division, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | | |
Collapse
|
90
|
Rajagopalan V, Zucker IH, Jones JA, Carlson M, Ma YJ. Cardiac ErbB-1/ErbB-2 mutant expression in young adult mice leads to cardiac dysfunction. Am J Physiol Heart Circ Physiol 2008; 295:H543-54. [DOI: 10.1152/ajpheart.91436.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple factors lead to the development and maintenance of chronic heart failure. Blockade of ErbB-2 or ErbB-4 tyrosine kinase receptor signaling leads to dilated cardiomyopathy. ErbB-1 may protect the heart against stress-induced injury and its ligand; epidermal growth factor (EGF) increases myocardial contractility, whereas heparin-binding EGF is essential for normal cardiac function. However, the role of ErbB-1 in control of cardiac function is not clear. We hypothesized that ErbB-1 is essential for maintaining adult cardiac function. Using the ecdysone-inducible gene expression system, we expressed humanized cardiomyocyte-specific dominant-negative ErbB-1 mutant receptors (hErbB-1-mut) in young adult mice that block endogenous cardiac ErbB-1 signaling. Molecular, morphological, and physiological tests (under anesthesia) were performed. As a result, hErbB-1-mut was expressed selectively in cardiomyocytes leading to the blockade of endogenous ErbB-1 phosphorylation and ErbB-2 transphosphorylation. An increase in left ventricular mass, atrial natriuretic factor expression, and histological changes were indicative of cardiac hypertrophy. Cardiac dilation, numerous cardiac lesions, and the loss of the clear boundary between cardiac fibrils were noted histologically. Early and long-term hErbB-1-mut induction led to a significant decrease in fractional shortening and to significant increases in left ventricular end-systolic diameter and volume. The treatment of adenylyl cyclase activator (forskolin analog) normalized the depressed cardiac function. Resting cardiac function returned to normal after reversing mutant expression. A 4-day survival rate of transverse-aortic constricted hErbB-1-mut mice was only 20% compared with 100% in controls. In conclusion, these observations indicate that the blockade of cardiac ErbB-1 signaling leads to the blockade of ErbB-2 signaling and that together they result in cardiac dysfunction.
Collapse
|
91
|
Milam JE, Keshamouni VG, Phan SH, Hu B, Gangireddy SR, Hogaboam CM, Standiford TJ, Thannickal VJ, Reddy RC. PPAR-gamma agonists inhibit profibrotic phenotypes in human lung fibroblasts and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2008; 294:L891-901. [PMID: 18162602 PMCID: PMC5926773 DOI: 10.1152/ajplung.00333.2007] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pulmonary fibrosis is characterized by alterations in fibroblast phenotypes resulting in excessive extracellular matrix accumulation and anatomic remodeling. Current therapies for this condition are largely ineffective. Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a member of the nuclear hormone receptor superfamily, the activation of which produces a number of biological effects, including alterations in metabolic and inflammatory responses. The role of PPAR-gamma as a potential therapeutic target for fibrotic lung diseases remains undefined. In the present study, we show expression of PPAR-gamma in fibroblasts obtained from normal human lungs and lungs of patients with idiopathic interstitial pneumonias. Treatment of lung fibroblasts and myofibroblasts with PPAR-gamma agonists results in inhibition of proliferative responses and induces cell cycle arrest. In addition, PPAR-gamma agonists, including a constitutively active PPAR-gamma construct (VP16-PPAR-gamma), inhibit the ability of transforming growth factor-beta1 to induce myofibroblast differentiation and collagen secretion. PPAR-gamma agonists also inhibit fibrosis in a murine model, even when administration is delayed until after the initial inflammation has largely resolved. These observations indicate that PPAR-gamma is an important regulator of fibroblast/myofibroblast activation and suggest a role for PPAR-gamma ligands as novel therapeutic agents for fibrotic lung diseases.
Collapse
Affiliation(s)
- Jami E Milam
- Univ. of Michigan, Division of Pulmonary and Critical Care Medicine, 109 Zina Pitcher Pl., 4062 BSRB, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Wieduwilt MJ, Moasser MM. The epidermal growth factor receptor family: biology driving targeted therapeutics. Cell Mol Life Sci 2008; 65:1566-84. [PMID: 18259690 PMCID: PMC3060045 DOI: 10.1007/s00018-008-7440-8] [Citation(s) in RCA: 528] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The epidermal growth factor family of receptor tyrosine kinases (ErbBs) plays essential roles in regulating cell proliferation, survival, differentiation and migration. The ErbB receptors carry out both redundant and restricted functions in mammalian development and in the maintenance of tissues in the adult mammal. Loss of regulation of the ErbB receptors underlies many human diseases, most notably cancer. Our understanding of the function and complex regulation of these receptors has fueled the development of targeted therapeutic agents for human malignancies in the last 15 years. Here we review the biology of ErbB receptors, including their structure, signaling, regulation, and roles in development and disease, then briefly touch on their increasing roles as targets for cancer therapy.
Collapse
Affiliation(s)
- M. J. Wieduwilt
- Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, UCSF, Box 0875, San Francisco, CA 94143-0875 USA
| | - M. M. Moasser
- Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, UCSF, Box 0875, San Francisco, CA 94143-0875 USA
| |
Collapse
|
93
|
Barrick CJ, Yu M, Chao HH, Threadgill DW. Chronic pharmacologic inhibition of EGFR leads to cardiac dysfunction in C57BL/6J mice. Toxicol Appl Pharmacol 2008; 228:315-25. [PMID: 18313710 DOI: 10.1016/j.taap.2007.12.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 12/04/2007] [Accepted: 12/04/2007] [Indexed: 11/24/2022]
Abstract
Molecule-targeted therapies like those against the epidermal growth factor receptor (EGFR) are becoming widely used in the oncology clinic. With improvements in treatment efficacy, many cancers are being treated as chronic diseases, with patients having prolonged exposure to several therapies that were previously only given acutely. The consequence of chronic suppression of EGFR activity may lead to unexpected toxicities like altered cardiac physiology, a common organ site for adverse drug effects. To explore this possibility, we treated C57BL/6J (B6) mice with two EGFR small molecule tyrosine kinase inhibitors (TKIs), irreversible EKB-569 and reversible AG-1478, orally for 3 months. In B6 female mice, chronic exposure to both TKIs depressed body weight gain and caused significant changes in left ventricular (LV) wall thickness and cardiac function. No significant differences were observed in heart weight or cardiomyocyte size but histological analysis revealed an increase in fibrosis and in the numbers of TUNEL-positive cells in the hearts from treated female mice. Consistent with histological results, LV apoptotic gene expression was altered, with significant downregulation of the anti-apoptotic gene Bcl2l1. Although there were no significant differences in any of these endpoints in treated male mice, suggesting sex may influence susceptibility to TKI mediated toxicity, the LVs of treated male mice had significant upregulation of Egf, Erbb2 and Nppb over controls. Taken together, these data suggest that chronic dietary exposure to TKIs may result in pathological and physiological changes in the heart.
Collapse
Affiliation(s)
- Cordelia J Barrick
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
94
|
Ruan X, Zheng F, Guan Y. PPARs and the kidney in metabolic syndrome. Am J Physiol Renal Physiol 2008; 294:F1032-47. [PMID: 18234957 DOI: 10.1152/ajprenal.00152.2007] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The metabolic syndrome (MetS) is defined by a set of metabolic risk factors, including insulin resistance, central obesity, dyslipidemia, hyperglycemia, and hypertension for type 2 diabetes and cardiovascular disease. Although both retrospective and prospective clinical studies have revealed that MetS is associated with chronic renal disease, even with a nondiabetic cause, the cellular and molecular mechanisms in this association remain largely uncharacterized. Recently, increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs), a subgroup of the nuclear hormone receptor superfamily of ligand-activated transcription factors, may play an important role in the pathogenesis of MetS. All three members of the PPAR nuclear receptor subfamily, PPARalpha, -beta/delta, and -gamma, are critical in regulating insulin sensitivity, adipogenesis, lipid metabolism, inflammation, and blood pressure. PPARs have also been implicated in many renal pathophysiological conditions, including diabetic nephropathy and glomerulosclerosis. Ligands for PPARs such as hypolipidemic PPARalpha activators, and antidiabetic thiazolidinedione PPARgamma agonists affect not only diverse aspects of MetS but also renal disease progression. Emerging data suggest that PPARs may be potential therapeutic targets for MetS and its related renal complications. This review focuses on current knowledge of the role of PPARs in MetS and discusses the potential therapeutic utility of PPAR modulators in the treatment of kidney diseases associated with MetS.
Collapse
Affiliation(s)
- Xiongzhong Ruan
- Center for Nephrology, University College of London, London, United Kingdom
| | | | | |
Collapse
|
95
|
Nakaoka Y, Nishida K, Narimatsu M, Kamiya A, Minami T, Sawa H, Okawa K, Fujio Y, Koyama T, Maeda M, Sone M, Yamasaki S, Arai Y, Koh GY, Kodama T, Hirota H, Otsu K, Hirano T, Mochizuki N. Gab family proteins are essential for postnatal maintenance of cardiac function via neuregulin-1/ErbB signaling. J Clin Invest 2007; 117:1771-81. [PMID: 17571162 PMCID: PMC1888569 DOI: 10.1172/jci30651] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 04/10/2007] [Indexed: 01/11/2023] Open
Abstract
Grb2-associated binder (Gab) family of scaffolding adaptor proteins coordinate signaling cascades downstream of growth factor and cytokine receptors. In the heart, among EGF family members, neuregulin-1beta (NRG-1beta, a paracrine factor produced from endothelium) induced remarkable tyrosine phosphorylation of Gab1 and Gab2 via erythroblastic leukemia viral oncogene (ErbB) receptors. We examined the role of Gab family proteins in NRG-1beta/ErbB-mediated signal in the heart by creating cardiomyocyte-specific Gab1/Gab2 double knockout mice (DKO mice). Although DKO mice were viable, they exhibited marked ventricular dilatation and reduced contractility with aging. DKO mice showed high mortality after birth because of heart failure. In addition, we noticed remarkable endocardial fibroelastosis and increase of abnormally dilated vessels in the ventricles of DKO mice. NRG-1beta induced activation of both ERK and AKT in the hearts of control mice but not in those of DKO mice. Using DNA microarray analysis, we found that stimulation with NRG-1beta upregulated expression of an endothelium-stabilizing factor, angiopoietin 1, in the hearts of control mice but not in those of DKO mice, which accounted for the pathological abnormalities in the DKO hearts. Taken together, our observations indicated that in the NRG-1beta/ErbB signaling, Gab1 and Gab2 of the myocardium are essential for both maintenance of myocardial function and stabilization of cardiac capillary and endocardial endothelium in the postnatal heart.
Collapse
Affiliation(s)
- Yoshikazu Nakaoka
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keigo Nishida
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Narimatsu
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsunori Kamiya
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Minami
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hirofumi Sawa
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsuya Okawa
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Fujio
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsuya Koyama
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makiko Maeda
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manami Sone
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoru Yamasaki
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuji Arai
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Gou Young Koh
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsuhiko Kodama
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisao Hirota
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kinya Otsu
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshio Hirano
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoki Mochizuki
- Department of Structural Analysis, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology (RCAI), Yokohama, Japan.
Laboratory of Developmental Immunology, Osaka University Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka, Japan.
Department of Cardiovascular Dynamics, National Cardiovascular Center Research Institute, Osaka, Japan.
Laboratory for System Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan.
Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan.
Horizontal Medical Research Organization, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Department of Clinical Evaluation of Medicines and Therapeutics, Osaka University Graduate School of Pharmaceutical Sciences, Osaka, Japan.
Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan.
Biomedical Research Center and Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
96
|
Lemmens K, Doggen K, De Keulenaer GW. Role of neuregulin-1/ErbB signaling in cardiovascular physiology and disease: implications for therapy of heart failure. Circulation 2007; 116:954-60. [PMID: 17709650 DOI: 10.1161/circulationaha.107.690487] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Since the discovery that neuregulin-1 (NRG-1)/ErbB signaling is indispensable in cardiac development, evidence has shown that this system also plays a crucial role in the adult heart. In patients, an inhibitory ErbB2 antibody, trastuzumab, used in the treatment of mammary carcinomas, increases the risk for the development of cardiotoxic cardiomyopathy. Postnatal disruption of NRG-1/ErbB signaling by gene targeting in mice leads to dilated cardiomyopathy. Initially, the search for the mechanisms behind these observations focused mainly on the effects of NRG-1 on cardiomyocyte growth and survival and revealed that NRG-1 has Akt-dependent antiapoptotic effects in cultured cardiomyocytes. In vivo studies, however, did not uniformly reinforce a role for apoptosis in the development of cardiomyopathy induced by impaired NRG-1/ErbB signaling. More recent studies have revealed that NRG-1 is involved in the regulation of cardiac sympathovagal balances by counterbalancing adrenergic stimulation of the adult myocardium and through an obligatory interaction with the muscarinic cholinergic system. NRG-1 is synthesized and released by the endocardial and cardiac microvascular endothelium, dynamically controlled by neurohormonal and biomechanical stimuli. The physiology of the cardiac NRG-1/ErbB system has implications for the treatment of both cancer and heart failure. Clinical studies in breast cancer with novel ErbB inhibitors are currently underway. Novel oncological indications for ErbB inhibition are emerging; cardiovascular side effects need to be carefully monitored. On the other hand, pharmacological activation of ErbB signaling is likely an unrecognized and beneficial effect of currently used drugs in heart failure and a promising therapeutic approach to prevent or reverse myocardial dysfunction.
Collapse
Affiliation(s)
- Katrien Lemmens
- University of Antwerp, Laboratory of Physiology, Universiteitsplein 1, Bldg T, 2nd Floor, 2610 Wilrijk, Belgium
| | | | | |
Collapse
|
97
|
Hoshijima M, Knöll R, Pashmforoush M, Chien KR. Reversal of calcium cycling defects in advanced heart failure toward molecular therapy. J Am Coll Cardiol 2007; 48:A15-23. [PMID: 17084280 DOI: 10.1016/j.jacc.2006.06.070] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 05/22/2006] [Accepted: 06/22/2006] [Indexed: 02/04/2023]
Abstract
Heart failure is a growing major cause of human morbidity and mortality worldwide. A wave of new insights from diverse laboratories has begun to uncover new therapeutic strategies that affect the molecular pathways within cardiomyocytes that drive heart failure progression. Using an integrative approach that employs insights from genetic-based studies in mouse and humans and in vivo somatic gene transfer studies, we have uncovered a new link between stress signals mediated by mechanical stretch and defects in sarcoplasmic reticulum (SR) calcium cycling. An intrinsic mechanical stress sensing system is embedded in the Z disc of cardiomyocytes, and defects in stretch responses can lead to heart failure progression and associated increases in wall stress. Reversal of the chronic increases in wall stress by promoting SR calcium cycling can prevent and partially reverse heart failure progression in multiple genetic and acquired model systems of heart failure in both small and large animals. We propose that reversal of advanced heart failure is possible by targeting the defects in SR calcium cycling, which may be a final common pathway for the progression of many forms of heart failure.
Collapse
Affiliation(s)
- Masahiko Hoshijima
- Institute of Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | | | | | | |
Collapse
|
98
|
Laing JG, Saffitz JE, Steinberg TH, Yamada KA. Diminished zonula occludens-1 expression in the failing human heart. Cardiovasc Pathol 2007; 16:159-64. [PMID: 17502245 DOI: 10.1016/j.carpath.2007.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2006] [Revised: 10/16/2006] [Accepted: 01/08/2007] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Reduced expression of the major gap junction protein connexin 43 (Cx43) in the failing human heart may lead to arrhythmias and sudden cardiac death. Cx43 interacts with the actin binding protein, zonula occludens-1 (ZO-1), and it has recently been demonstrated that ZO-1 regulates the formation and function of Cx43 gap junctions. We hypothesize that normal expression of ZO-1 and its interaction with Cx43 are required for appropriate assembly and function of Cx43 gap junctions in the heart. Here, we determined whether expression of ZO-1 is altered in patients with heart failure. METHODS We examined ventricular myocardium from hearts of patients in end-stage heart failure, obtained at transplant, for ZO-1 expression by immunohistochemistry. We also subjected lysates made from this tissue to immunoblotting to determine the level of ZO-1 expression. RESULTS AND CONCLUSIONS ZO-1 was found at 96% of the intercalated discs in nonfailing control human hearts, where it colocalized with Cx43. In contrast, there was ZO-1 immunostaining at 5% of intercalated discs in failing hearts, coincident with a reduction in Cx43 staining in intercalated discs. Immunoblotting analysis showed that there was a 95% reduction in ZO-1 expression in human heart failure. Loss of ZO-1 at intercalated discs in heart failure may play a critical role in remodeling of Cx43 gap junctions, which may contribute to abnormal impulse propagation and arrhythmogenesis, thereby predisposing patients in heart failure to sudden cardiac death.
Collapse
Affiliation(s)
- James G Laing
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
99
|
Kuramochi Y, Guo X, Sawyer DB. Neuregulin activates erbB2-dependent src/FAK signaling and cytoskeletal remodeling in isolated adult rat cardiac myocytes. J Mol Cell Cardiol 2006; 41:228-35. [PMID: 16769082 PMCID: PMC1847613 DOI: 10.1016/j.yjmcc.2006.04.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 03/14/2006] [Accepted: 04/11/2006] [Indexed: 02/01/2023]
Abstract
Cardiac myocyte erbB2 expression is required for maintenance of normal cardiac structure and function, though its role in cardiac cellular physiology is incompletely understood. We tested the hypothesis that erbB2 signaling modulates focal adhesion formation via activation of a src/FAK pathway using adult rat ventricular myocytes in primary culture. The erbB ligand neuregulin-1Beta (NRG-1Beta) induced phosphorylation of Src at Y416 and Y215, and FAK at Y861. Using antibody and pharmacological inhibitor strategies, we found that FAK activation was erbB2- and Src-dependent, but independent of PI3-kinase/Akt pathway. Furthermore, NRG-1Beta stimulated the formation of a multiprotein complex between erbB2, FAK, p130(CAS) and paxillin within 30 min, and induced lamellipodia with longitudinal elongation of the myocytes within days. The extension of lamellipodia resulted in restoration of cell-to-cell contact between isolated myocytes, allowing for synchronous beating. These effects of NRG-1Beta were prevented by a src inhibitor as well as an antibody to erbB2. These results suggest the potential role of NRG-1Beta/erbB2/Src/FAK signaling in the maintenance and repair of electrical and mechanical coupling in cardiomyocytes.
Collapse
Affiliation(s)
- Yukio Kuramochi
- Center for Molecular Stress Response, Whitaker Cardiovascular Institute, Department of Medicine, Boston University Medical Center, EBRC Room 329, 650 Albany Street, Boston, MA 02118, USA
| | - Xinxin Guo
- Center for Molecular Stress Response, Whitaker Cardiovascular Institute, Department of Medicine, Boston University Medical Center, EBRC Room 329, 650 Albany Street, Boston, MA 02118, USA
| | - Douglas B. Sawyer
- Center for Molecular Stress Response, Whitaker Cardiovascular Institute, Department of Medicine, Boston University Medical Center, EBRC Room 329, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
100
|
|