Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
2.
Exp Mol Med ; 55(5): 965-973, 2023 05.
Article in English | MEDLINE | ID: mdl-37121973

ABSTRACT

Insulin and insulin-like growth factor 1 (IGF-1) signaling regulate cellular growth and glucose metabolism in the myocardium. However, their physiological role in the cells of the cardiac conduction system has never been explored. Therefore, we sought to determine the spatiotemporal function of insulin/IGF-1 receptors in the sinoatrial node (SAN). We generated cardiac conduction cell-specific inducible IGF-1 receptor (IGF-1R) knockout (KO) (CSIGF1RKO), insulin receptor (IR) KO (CSIRKO), and IR/IGF-1R double-KO (CSDIRKO) mice and evaluated their phenotypes. Telemetric electrocardiography revealed regular sinus rhythm in CSIGF1RKO mice, indicating that IGF-1R is dispensable for normal pacemaking. In contrast, CSIRKO and CSDIRKO mice exhibited profound sinus bradycardia. CSDIRKO mice showed typical sinus node dysfunction characterized by junctional rhythm and sinus pauses on electrocardiography. Interestingly, the lack of an insulin receptor in the SAN cells of CSIRKO and CSDIRKO mice caused sinus nodal fibrosis. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) protein expression significantly decreased in the CSIRKO and CSDIRKO mice relative to the controls. A patch-clamp study of the SAN cells of CSIRKO mice revealed a significant decrease in the funny current, which is responsible for spontaneous diastolic depolarization in the SAN. This result suggested that insulin receptor loss reduces the heart rate via downregulation of the HCN4 channel. Additionally, HCN1 expression was decreased in CSDIRKO mice, explaining their sinus node dysfunction. Our results reveal a previously unrecognized role of insulin/IGF-1 signaling in sinus node structural maintenance and pacemaker function.


Subject(s)
Sick Sinus Syndrome , Sinoatrial Node , Mice , Animals , Sinoatrial Node/metabolism , Sick Sinus Syndrome/metabolism , Receptor, Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism
3.
Cell Death Dis ; 12(7): 688, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244467

ABSTRACT

The insulin-like growth factor 1 receptor (IGF-1R) signaling in cardiomyocytes is implicated in physiological hypertrophy and myocardial aging. Although fibroblasts account for a small amount of the heart, they are activated when the heart is damaged to promote cardiac remodeling. However, the role of IGF-1R signaling in cardiac fibroblasts is still unknown. In this study, we investigated the roles of IGF-1 signaling during agonist-induced cardiac fibrosis and evaluated the molecular mechanisms in cultured cardiac fibroblasts. Using an experimental model of cardiac fibrosis with angiotensin II/phenylephrine (AngII/PE) infusion, we found severe interstitial fibrosis in the AngII/PE infused myofibroblast-specific IGF-1R knockout mice compared to the wild-type mice. In contrast, low-dose IGF-1 infusion markedly attenuated AngII-induced cardiac fibrosis by inhibiting fibroblast proliferation and differentiation. Mechanistically, we demonstrated that IGF-1-attenuated AngII-induced cardiac fibrosis through the Akt pathway and through suppression of rho-associated coiled-coil containing kinases (ROCK)2-mediated α-smooth muscle actin (αSMA) expression. Our study highlights a novel function of the IGF-1/IGF-1R signaling in agonist-induced cardiac fibrosis. We propose that low-dose IGF-1 may be an efficacious therapeutic avenue against cardiac fibrosis.


Subject(s)
Actins/metabolism , Cardiomyopathies/prevention & control , Fibroblasts/drug effects , Insulin-Like Growth Factor I/administration & dosage , Myocytes, Cardiac/drug effects , Angiotensin II , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Infusions, Intravenous , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenylephrine , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction , rho-Associated Kinases/metabolism
4.
Biochem Biophys Res Commun ; 516(2): 350-356, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31208720

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) is a member of the TNF superfamily. RANKL increases endothelial permeability and induces angiogenesis, suggesting its critical roles in the vasculature. Despite the evidence implicating RANKL in vascular pathology, its role in ischemic retinopathy has not been previously reported. In this study, neonatal mice were exposed to 75% oxygen from postnatal day (P)7 to P12 to induce vaso-obliteration, and then returned to room air from P12 to P17, causing the retina to become hypoxic and inducing vascular endothelial growth factor (VEGF) signaling, which produces pathological neovascularization. On P12, the mice received a single intravitreal injection of control IgG1 or RANK-Fc, and retinas were obtained at P17. On P17, RANKL was expressed strongly and selectively in the neovascular tufts (NVT) area. RANKL colocalized with αSMA or PDGFRß in NVT. However, co-immunostaining revealed that CD31-positive areas were not the same as RANKL, which indicates that RANKL might be produced by retinal pericytes, not endothelial cells. Consistent with this finding, chemical hypoxia upregulated RANKL expression in cultured human retinal pericytes but not in endothelial cells. Treatment with RANK-Fc markedly reduced the NVT area compared to that in mice administered the IgG1 injection. In contrast, the central avascular region of RANKL-Fc retina was comparable to the controls. In addition, we assessed retinal vascular permeability using FITC-labeled dextran. RANK-Fc treated mice displayed decreased vascular leakages compared to those injected with IgG1. Our work supports the use of an RANKL blockade as a potential therapeutic approach against ischemic retinopathies.


Subject(s)
Ischemia/pathology , Neovascularization, Pathologic/pathology , RANK Ligand/antagonists & inhibitors , Retinal Diseases/pathology , Animals , Animals, Newborn , Capillary Permeability , Cell Hypoxia , Disease Models, Animal , Endothelial Cells/metabolism , Ischemia/complications , Mice, Inbred C57BL , Neovascularization, Pathologic/complications , Pericytes/metabolism , RANK Ligand/metabolism , Retinal Diseases/complications , Retinal Ganglion Cells/metabolism
5.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30070361

ABSTRACT

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Subject(s)
ErbB Receptors/metabolism , Receptor, IGF Type 1/deficiency , Receptor, Insulin/deficiency , Thyroid Cancer, Papillary/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Neoplasms/metabolism , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Thyroid Cancer, Papillary/pathology , Thyroid Epithelial Cells/pathology , Thyroid Neoplasms/pathology , Thyrotropin/biosynthesis , Thyrotropin/metabolism
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1183-1191, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29378301

ABSTRACT

While deletion of Akt1 results in a smaller heart size and Akt2-/- mice are mildly insulin resistant, Akt1-/-/Akt2-/- mice exhibit perinatal lethality, indicating a large degree of functional overlap between the isoforms of the serine/threonine kinase Akt. The present study aimed to determine the cooperative contribution of Akt1 and Akt2 on the structure and contractile function of adult hearts. To generate an inducible, cardiomyocyte-restricted Akt2 knockout (KO) model, Akt2flox/flox mice were crossed with tamoxifen-inducible MerCreMer transgenic (MCM) mice and germline Akt1-/- mice to generate the following genotypes:Akt1+/+; Akt2flox/flox (WT), Akt2flox/flox; α-MHC-MCM (iAkt2 KO), Akt1-/-, and Akt1-/-; Akt2flox/flox; α-MHC-MCM mice (Akt1-/-/iAkt2 KO). At 28 days after the first tamoxifen injection, Akt1-/-/iAkt2 KO mice developed contractile dysfunction paralleling increased atrial and brain natriuretic peptide (ANP and BNP) levels, and repressed mitochondrial gene expression. Neither cardiac fibrosis nor apoptosis were detected in Akt1-/-/iAkt2 KO hearts. To explore potential molecular mechanisms for contractile dysfunction, we investigated myocardial microstructure before the onset of heart failure. At 3 days after the first tamoxifen injection, Akt1-/-/iAkt2 KO hearts showed decreased expression of connexin43 (Cx43) and connexin-interacting protein zonula occludens-1 (ZO-1). Furthermore, Akt1/2 silencing significantly decreased both Cx43 and ZO-1 expression in cultured neonatal rat cardiomyocytes in concert with reduced beating frequency. Akt1 and Akt2 are required to maintain cardiac contraction. Loss of Akt signaling disrupts gap junction protein, which might precipitate early contractile dysfunction prior to heart failure in the absence of myocardial remodeling, such as hypertrophy, fibrosis, or cell death.


Subject(s)
Cardiomyopathies/metabolism , Connexin 43/metabolism , Myocardial Contraction , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/deficiency , Zonula Occludens-1 Protein/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Connexin 43/genetics , Fibrosis , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Rats , Zonula Occludens-1 Protein/genetics
7.
Endocrinology ; 157(1): 336-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26469138

ABSTRACT

IGF-1 receptor (IGF-1R) signaling is implicated in cardiac hypertrophy and longevity. However, the role of IGF-1R in age-related cardiac remodeling is only partially understood. We therefore sought to determine whether the deletion of the IGF-1R in cardiomyocytes might delay the development of aging-associated myocardial pathologies by examining 2-year-old male cardiomyocyte-specific IGF-1R knockout (CIGF1RKO) mice. Aging was associated with the induction of IGF-1R expression in hearts. Cardiomyocytes hypertrophied with age in wild-type (WT) mice. In contrast, the cardiac hypertrophic response associated with aging was blunted in CIGF1RKO mice. Concomitantly, fibrosis was reduced in aged CIGF1RKO compared with aged WT hearts. Expression of proinflammatory cytokines such as IL-1α, IL-1ß, IL-6, and receptor activator of nuclear factor-κB ligand was increased in aged WT hearts, but this increase was attenuated in aged CIGF1RKO hearts. Phosphorylation of Akt was increased in aged WT, but not in aged CIGF1RKO, hearts. In cultured cardiomyocytes, IGF-1 induced senescence as demonstrated by increased senescence-associated ß-galactosidase staining, and a phosphoinositide 3-kinase inhibitor inhibited this effect. Furthermore, inhibition of phosphoinositide 3-kinase significantly prevented the increase in IL-1α, IL-1ß, receptor activator of nuclear factor-κB ligand, and p21 protein expression by IGF-1. These data reveal an essential role for the IGF-1-IGF-1R-Akt pathway in mediating cardiomyocyte senescence.


Subject(s)
Aging , Cardiomegaly/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Receptor, IGF Type 1/metabolism , Ventricular Remodeling , Animals , Biomarkers/metabolism , Cardiomegaly/immunology , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cells, Cultured , Cellular Senescence/drug effects , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Fibrosis , Gene Expression Regulation, Developmental/drug effects , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Insulin-Like Growth Factor I/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Ventricular Remodeling/drug effects
8.
J Biol Chem ; 289(8): 4839-52, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24398673

ABSTRACT

Doxorubicin is one of the most widely used anti-cancer drugs, but its clinical application is compromised by severe adverse effects in different organs including cardiotoxicity. In the present study we explored mechanisms of doxorubicin-induced cytotoxicity by revealing a novel role for the AMP-activated protein kinase α2 (AMPKα2) in mouse embryonic fibroblasts (MEFs). Doxorubicin robustly induced the expression of AMPKα2 in MEFs but slightly reduced AMPKα1 expression. Our data support the previous notion that AMPKα1 harbors survival properties under doxorubicin treatment. In contrast, analyses of Ampkα2(-/-) MEFs, gene knockdown of AMPKα2 by shRNA, and inhibition of AMPKα2 activity with an AMPK inhibitor indicated that AMPKα2 functions as a pro-apoptotic molecule under doxorubicin treatment. Doxorubicin induced AMPKα2 at the transcription level via E2F1, a transcription factor that regulates apoptosis in response to DNA damage. E2F1 directly transactivated the Ampkα2 gene promoter. In turn, AMPKα2 significantly contributed to stabilization and activation of E2F1 by doxorubicin, forming a positive signal amplification loop. AMPKα2 directly interacted with and phosphorylated E2F1. This signal loop was also detected in H9c2, C2C12, and ECV (human epithelial cells) cells as well as mouse liver under doxorubicin treatment. Resveratrol, which has been suggested to attenuate doxorubicin-induced cytotoxicity, significantly blocked induction of AMPKα2 and E2F1 by doxorubicin, leading to protection of these cells. This signal loop appears to be non-carcinoma-specific because AMPKα2 was not induced by doxorubicin in five different tested cancer cell lines. These results suggest that AMPKα2 may serve as a novel target for alleviating the cytotoxicity of doxorubicin.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Doxorubicin/pharmacology , E2F1 Transcription Factor/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Cell Death/drug effects , Cell Line , Cytoprotection/drug effects , DNA Damage , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Stability/drug effects , Resveratrol , Stilbenes/pharmacology
9.
FASEB J ; 27(12): 4899-908, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23982142

ABSTRACT

Although thyroid-stimulating hormone (TSH) is known to be a major regulator of thyroid hormone biosynthesis and thyroid growth, insulin-like growth factor 1 (IGF-1) is required for mediating thyrocyte growth in concert with TSH in vitro. We generated mice with thyrocyte-selective ablation of IGF-1 receptor (TIGF1RKO) to explore the role of IGF-1 receptor signaling on thyroid function and growth. In 5-wk-old TIGF1RKO mice, serum thyroxine (T4) concentrations were decreased by 30% in concert with a 43% down-regulation of the monocarboxylate transporter 8 (MCT8), which is involved in T4 secretion. Despite a 3.5-fold increase in circulating concentrations of TSH, thyroid architecture and size were normal. Furthermore, thyrocyte area was increased by 40% in WT thyroids after 10 d TSH injection, but this effect was absent in TSH-injected TIGF1RKO mice. WT mice treated with methimazole and sodium perchlorate for 2 or 6 wk exhibited pronounced goiter development (2.0 and 5.4-fold, respectively), but in TIGF1RKO mice, goiter development was completely abrogated. These data reveal an essential role for IGF-1 receptor signaling in the regulation of thyroid function and TSH-stimulated goitrogenesis.


Subject(s)
Goiter/metabolism , Receptor, IGF Type 1/genetics , Thyrotropin/metabolism , Thyroxine/metabolism , Animals , Antithyroid Agents/pharmacology , Down-Regulation , Goiter/chemically induced , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methimazole/pharmacology , Mice , Mice, Knockout , Monocarboxylic Acid Transporters , Perchlorates/toxicity , Receptor, IGF Type 1/deficiency , Sodium Compounds/toxicity , Symporters , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology
10.
Cardiovasc Res ; 94(1): 105-14, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22298642

ABSTRACT

AIMS: Although increased levels of myocardial receptor activator of nuclear factor (NF)-κB ligand (RANKL) have been reported in heart failure, the role of this pathway in mediating activation of inflammatory pathways during myocardial remodelling is less well understood. This study sought to determine the role of myocardial RANKL in regulating cytokine expression. METHODS AND RESULTS: A marked increase in RANKL expression occurred as early as 6h following transverse aortic constriction (TAC) in mouse hearts and persisted at 3 and 17 days. An increase in tumour necrosis factor-α (TNF-α), interleukin (IL)-1α, and IL-1ß was observed in the hypertrophied hearts only at 3 or 17 days after TAC. Treatment with losartan significantly attenuated TAC-induced cardiac hypertrophy, in parallel with decreased expression of RANKL, TNF-α, IL-1α, and IL-1ß. Furthermore, injection of a RANKL-neutralizing monoclonal antibody attenuated RANKL-induced cytokine expression. RANKL stimulated expression of TNF-α, IL-1α, and IL-1ß in neonatal rat cardiomyocytes via activation of NF-κB. RANKL-induced NF-κB activation and expression of these cytokines were both attenuated when RANK, receptor for RANKL, or TRAF2 or TRAF6, adaptors for RANK, was silenced by siRNA. Furthermore, inhibitors of phospholipase C (PLC), protein kinase C (PKC), and inhibitor of κB kinase also significantly inhibited RANKL-induced cellular activities, but inhibitors of phosphatidylinositol 3-kinase, extracellular signal-regulated kinase, or p38 mitogen-activated protein kinase were without effect. CONCLUSION: Our data demonstrate for the first time that the pressure-overloaded myocardium generates RANKL, which induces TNF-α, IL-1α, and IL-1ß production via a RANK-TRAF2/TRAF6-PLC-PKC-NF-κB-mediated autocrine mechanism.


Subject(s)
Cardiomegaly/immunology , Cytokines/metabolism , Inflammation Mediators/metabolism , Myocarditis/immunology , Myocytes, Cardiac/immunology , RANK Ligand/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Autocrine Communication , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured , Cytokines/genetics , Disease Models, Animal , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Myocarditis/genetics , Myocarditis/prevention & control , RANK Ligand/antagonists & inhibitors , RNA Interference , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
11.
Endocr J ; 58(11): 1013-9, 2011.
Article in English | MEDLINE | ID: mdl-21908931

ABSTRACT

Thyroid-stimulating hormone (TSH) is the primary regulator of thyroid growth and function acting via cyclic AMP signaling cascades. In cultured thyrocytes, insulin and/or insulin-like growth factor-1 (IGF-1) are required for mediating thyrocyte proliferation in concert with TSH. To determine the role of insulin signaling in thyroid, growth in vivo, mice with thyrocyte-selective ablation of the insulin receptor (IR) were generated by crossing mice homozygous for a floxed IR allele with transgenic mice in which thyrocyte-specific expression of Cre recombinase was driven by the human thyroid peroxidase (TPO) gene promoter. Immunohistochemistry and Western blot analysis confirmed near complete loss of IR expression in the thyroid of thyrocyte IR knockout mice. These mice are viable and have no obvious thyroid dysfunction and macro- and microscopic thyroid morphology was normal. Thus, insulin signaling in thyrocytes does not play an essential role in the architecture and function of the thyroid in vivo.


Subject(s)
Receptor, Insulin/deficiency , Thyroid Gland/physiology , Animals , Immunohistochemistry , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/physiology , Integrases/genetics , Integrases/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA/chemistry , RNA/genetics , Receptor, Insulin/genetics , Receptor, Insulin/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Specific Pathogen-Free Organisms , Thyroid Gland/cytology , Thyrotropin/blood , Thyroxine/blood
SELECTION OF CITATIONS
SEARCH DETAIL