Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Circ Res ; 133(2): 138-157, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37232184

ABSTRACT

BACKGROUND: Cyclic nucleotides play critical roles in cardiovascular biology and disease. PDE10A (phosphodiesterase 10A) is able to hydrolyze both cAMP and cGMP. PDE10A expression is induced in various human tumor cell lines, and PDE10A inhibition suppresses tumor cell growth. Chemotherapy drug such as doxorubicin (DOX) is widely used in chemotherapy. However, cardiotoxicity of DOX remains to be a serious clinical complication. In the current study, we aim to determine the role of PDE10A and the effect of PDE10A inhibition on cancer growth and cardiotoxicity induced by DOX. METHODS: We used global PDE10A knockout (KO) mice and PDE10A inhibitor TP-10 to block PDE10A function. DOX-induced cardiotoxicity was evaluated in C57Bl/6J mice and nude mice with implanted ovarian cancer xenografts. Isolated adult mouse cardiomyocytes and a human ovarian cancer cell line were used for in vitro functional and mechanistic studies. RESULTS: We found that PDE10A deficiency or inhibition alleviated DOX-induced myocardial atrophy, apoptosis, and dysfunction in C57Bl/6J mice. RNA sequencing study revealed a number of PDE10A-regulated signaling pathways involved in DOX-induced cardiotoxicity. PDE10A inhibition increased the death, decreased the proliferation, and potentiated the effect of DOX on various human cancer cells. Importantly, in nude mice with implanted ovarian cancer xenografts, PDE10A inhibition attenuated tumor growth while protecting DOX-induced cardiotoxicity. In isolated cardiomyocytes, PDE10A contributed to DOX-induced cardiomyocyte death via increasing Top2ß (topoisomerase 2ß) expression, mitochondrial dysfunction, and DNA damage by antagonizing cGMP/PKG (protein kinase G) signaling. PDE10A contributed to cardiomyocyte atrophy via potentiating FoxO3 (forkhead box O3) signaling via both cAMP/PKA (protein kinase A)- and cGMP/PKG-dependent signaling. CONCLUSIONS: Taken together, our study elucidates a novel role for PDE10A in cardiotoxicity induced by DOX and cancer growth. Given that PDE10A has been already proven to be a safe drug target, PDE10A inhibition may represent a novel therapeutic strategy in cancer therapy, with effects preventing DOX-induced cardiotoxicity and simultaneously antagonizing cancer growth.


Subject(s)
Cardiotoxicity , Ovarian Neoplasms , Animals , Female , Humans , Mice , Apoptosis , Atrophy/complications , Atrophy/metabolism , Atrophy/pathology , Cardiotoxicity/metabolism , Doxorubicin/adverse effects , Doxorubicin/toxicity , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Myocytes, Cardiac/metabolism , Ovarian Neoplasms/metabolism , Oxidative Stress , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism
2.
Circ Res ; 133(3): 271-287, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37409456

ABSTRACT

BACKGROUND: Cardiomyopathy is characterized by the pathological accumulation of resident cardiac fibroblasts that deposit ECM (extracellular matrix) and generate a fibrotic scar. However, the mechanisms that control the timing and extent of cardiac fibroblast proliferation and ECM production are not known, hampering the development of antifibrotic strategies to prevent heart failure. METHODS: We used the Tcf21 (transcription factor 21)MerCreMer mouse line for fibroblast-specific lineage tracing and p53 (tumor protein p53) gene deletion. We characterized cardiac physiology and used single-cell RNA-sequencing and in vitro studies to investigate the p53-dependent mechanisms regulating cardiac fibroblast cell cycle and fibrosis in left ventricular pressure overload induced by transaortic constriction. RESULTS: Cardiac fibroblast proliferation occurs primarily between days 7 and 14 following transaortic constriction in mice, correlating with alterations in p53-dependent gene expression. p53 deletion in fibroblasts led to a striking accumulation of Tcf21-lineage cardiac fibroblasts within the normal proliferative window and precipitated a robust fibrotic response to left ventricular pressure overload. However, excessive interstitial and perivascular fibrosis does not develop until after cardiac fibroblasts exit the cell cycle. Single-cell RNA sequencing revealed p53 null fibroblasts unexpectedly express lower levels of genes encoding important ECM proteins while they exhibit an inappropriately proliferative phenotype. in vitro studies establish a role for p53 in suppressing the proliferative fibroblast phenotype, which facilitates the expression and secretion of ECM proteins. Importantly, Cdkn2a (cyclin-dependent kinase inhibitor 2a) expression and the p16Ink4a-retinoblastoma cell cycle control pathway is induced in p53 null cardiac fibroblasts, which may eventually contribute to cell cycle exit and fulminant scar formation. CONCLUSIONS: This study reveals a mechanism regulating cardiac fibroblast accumulation and ECM secretion, orchestrated in part by p53-dependent cell cycle control that governs the timing and extent of fibrosis in left ventricular pressure overload.


Subject(s)
Cicatrix , Heart Ventricles , Mice , Animals , Heart Ventricles/pathology , Cicatrix/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Fibrosis , Fibroblasts/metabolism , Cell Proliferation , Myocardium/metabolism
3.
Circ Res ; 131(11): 926-943, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36278398

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is a growing health problem without effective therapies. Epidemiological studies indicate that diabetes is a strong risk factor for HFpEF, and about 45% of patients with HFpEF are suffering from diabetes, yet the underlying mechanisms remain elusive. METHODS: Using a combination of echocardiography, hemodynamics, RNA-sequencing, molecular biology, in vitro and in vivo approaches, we investigated the roles of SIRT6 (sirtuin 6) in regulation of endothelial fatty acid (FA) transport and HFpEF in diabetes. RESULTS: We first observed that endothelial SIRT6 expression was markedly diminished in cardiac tissues from heart failure patients with diabetes. We then established an experimental mouse model of HFpEF in diabetes induced by a combination of the long-term high-fat diet feeding and a low-dose streptozocin challenge. We also generated a unique humanized SIRT6 transgenic mouse model, in which a single copy of human SIRT6 transgene was engineered at mouse Rosa26 locus and conditionally induced with the Cre-loxP technology. We found that genetically restoring endothelial SIRT6 expression in the diabetic mice ameliorated diastolic dysfunction concurrently with decreased cardiac lipid accumulation. SIRT6 gain- or loss-of-function studies showed that SIRT6 downregulated endothelial FA uptake. Mechanistically, SIRT6 suppressed endothelial expression of PPARγ through SIRT6-dependent deacetylation of histone H3 lysine 9 around PPARγ promoter region; and PPARγ reduction mediated SIRT6-dependent inhibition of endothelial FA uptake. Importantly, oral administration of small molecule SIRT6 activator MDL-800 to diabetic mice mitigated cardiac lipid accumulation and diastolic dysfunction. CONCLUSIONS: The impairment of endothelial SIRT6 expression links diabetes to HFpEF through the alteration of FA transport across the endothelial barrier. Genetic and pharmacological strategies that restored endothelial SIRT6 function in mice with diabetes alleviated experimental HFpEF by limiting FA uptake and improving cardiac metabolism, thus warranting further clinical evaluation.


Subject(s)
Diabetes Mellitus, Experimental , Heart Failure , Sirtuins , Humans , Mice , Animals , Stroke Volume/physiology , Heart Failure/metabolism , PPAR gamma , Disease Models, Animal , Sirtuins/genetics , Lipids
4.
Circulation ; 141(3): 217-233, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31801360

ABSTRACT

BACKGROUND: Heart failure is a leading cause of death worldwide. Cyclic nucleotide phosphodiesterases (PDEs), through degradation of cyclic nucleotides, play critical roles in cardiovascular biology and disease. Our preliminary screening studies have revealed PDE10A upregulation in the diseased heart. However, the roles of PDE10A in cardiovascular biology and disease are largely uncharacterized. The current study is aimed to investigate the regulation and function of PDE10A in cardiac cells and in the progression of cardiac remodeling and dysfunction. METHODS: We used isolated adult mouse cardiac myocytes and fibroblasts, as well as preclinical mouse models of hypertrophy and heart failure. The PDE10A selective inhibitor TP-10, and global PDE10A knock out mice were used. RESULTS: We found that PDE10A expression remains relatively low in normal and exercised heart tissues. However, PDE10A is significantly upregulated in mouse and human failing hearts. In vitro, PDE10A deficiency or inhibiting PDE10A with selective inhibitor TP-10, attenuated cardiac myocyte pathological hypertrophy induced by Angiotensin II, phenylephrine, and isoproterenol, but did not affect cardiac myocyte physiological hypertrophy induced by IGF-1 (insulin-like growth factor 1). TP-10 also reduced TGF-ß (transforming growth factor-ß)-stimulated cardiac fibroblast activation, proliferation, migration and extracellular matrix synthesis. TP-10 treatment elevated both cAMP and cGMP levels in cardiac myocytes and cardiac fibroblasts, consistent with PDE10A as a cAMP/cGMP dual-specific PDE. In vivo, global PDE10A deficiency significantly attenuated myocardial hypertrophy, cardiac fibrosis, and dysfunction induced by chronic pressure overload via transverse aorta constriction or chronic neurohormonal stimulation via Angiotensin II infusion. Importantly, we demonstrated that the pharmacological effect of TP-10 is specifically through PDE10A inhibition. In addition, TP-10 is able to reverse pre-established cardiac hypertrophy and dysfunction. RNA-Sequencing and bioinformatics analysis further identified a PDE10A-regualted transcriptome involved in cardiac hypertrophy, fibrosis, and cardiomyopathy. CONCLUSIONS: Taken together, our study elucidates a novel role for PDE10A in the regulation of pathological cardiac remodeling and development of heart failure. Given that PDE10A has been proven to be a safe drug target, PDE10A inhibition may represent a novel therapeutic strategy for preventing and treating cardiac diseases associated with cardiac remodeling.


Subject(s)
Cardiomegaly/enzymology , Fibroblasts/enzymology , Myocytes, Cardiac/enzymology , Phosphoric Diester Hydrolases/metabolism , Ventricular Remodeling , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Fibroblasts/pathology , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Phosphoric Diester Hydrolases/genetics , Transcriptome
5.
Am J Pathol ; 189(9): 1721-1731, 2019 09.
Article in English | MEDLINE | ID: mdl-31220449

ABSTRACT

Mutations in natriuretic peptide receptor 2 (Npr2) gene cause a rare form of short-limbed dwarfism, but its physiological effects have not been well studied. Human and mouse genetic data suggest that Npr2 in the kidney plays a role in salt homeostasis. Herein, we described anatomic changes within renal papilla of Npr2 knockout (Npr2-/-) mice. Dramatic reduction was found in diuresis, and albuminuria was evident after administration of 1% NaCl in drinking water in Npr2-/- and heterozygous (Npr2+/-) mice compared with their wild-type (Npr2+/+) littermates. There was indication of renal epithelial damage accompanied by high numbers of red blood cells and inflammatory cells (macrophage surface glycoproteins binding to galectin-3) and an increase of renal epithelial damage marker (T-cell Ig and mucin domain 1) in Npr2-/- mice. Addition of 1% NaCl tended to increase apoptotic cells (cleaved caspase 3) in the renal papilla of Npr2-/- mice. In vitro, genetic silencing of the Npr2 abolished protective effects of C-type natriuretic peptide, a ligand for Npr2, against death of M-1 kidney epithelial cells exposed to 360 mmol/L NaCl. Finally, significantly lower levels of expression of the NPR2 protein were detected in renal samples of hypertensive compared with normotensive human subjects. Taken together, these findings suggest that Npr2 is essential to protect renal epithelial cells from high concentrations of salt and prevent kidney injury.


Subject(s)
Acute Kidney Injury/prevention & control , Hypertension/pathology , Kidney Medulla/drug effects , Receptors, Atrial Natriuretic Factor/physiology , Sodium Chloride/toxicity , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Female , Humans , Hypertension/genetics , Hypertension/metabolism , Kidney Medulla/metabolism , Kidney Medulla/pathology , Male , Mice , Mice, Knockout
6.
J Mol Cell Cardiol ; 129: 92-104, 2019 04.
Article in English | MEDLINE | ID: mdl-30771308

ABSTRACT

Serum response factor (SRF) and the SRF co-activators myocardin-related transcription factors (MRTFs) are essential for epicardium-derived progenitor cell (EPDC)-mobilization during heart development; however, the impact of developmental EPDC deficiencies on adult cardiac physiology has not been evaluated. Here, we utilize the Wilms Tumor-1 (Wt1)-Cre to delete Mrtfs or Srf in the epicardium, which reduced the number of EPDCs in the adult cardiac interstitium. Deficiencies in Wt1-lineage EPDCs prevented the development of cardiac fibrosis and diastolic dysfunction in aged mice. Mice lacking MRTF or SRF in EPDCs also displayed preservation of cardiac function following myocardial infarction partially due to the depletion of Wt1 lineage-derived cells in the infarct. Interestingly, depletion of Wt1-lineage EPDCs allows for the population of the infarct with a Wt1-negative cell lineage with a reduced fibrotic profile. Taken together, our study conclusively demonstrates the contribution of EPDCs to both ischemic cardiac remodeling and the development of diastolic dysfunction in old age, and reveals the existence of an alternative Wt1-negative source of resident fibroblasts that can populate the infarct.


Subject(s)
Aging/pathology , Fibroblasts/pathology , Myocardial Ischemia/pathology , Pericardium/pathology , Animals , Cell Lineage , Diastole , Fibrosis , Heart/physiopathology , Mice, Knockout , Myocardial Ischemia/physiopathology , Serum Response Factor/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism , Ventricular Remodeling , WT1 Proteins/metabolism
7.
Circulation ; 138(17): 1864-1878, 2018 10 23.
Article in English | MEDLINE | ID: mdl-29716942

ABSTRACT

BACKGROUND: Hypertrophic cardiomyocyte growth and dysfunction accompany various forms of heart disease. The mechanisms responsible for transcriptional changes that affect cardiac physiology and the transition to heart failure are not well understood. The intercalated disc (ID) is a specialized intercellular junction coupling cardiomyocyte force transmission and propagation of electrical activity. The ID is gaining attention as a mechanosensitive signaling hub and hotspot for causative mutations in cardiomyopathy. METHODS: Transmission electron microscopy, confocal microscopy, and single-molecule localization microscopy were used to examine changes in ID structure and protein localization in the murine and human heart. We conducted detailed cardiac functional assessment and transcriptional profiling of mice lacking myocardin-related transcription factor (MRTF)-A and MRTF-B specifically in adult cardiomyocytes to evaluate the role of mechanosensitive regulation of gene expression in load-induced ventricular remodeling. RESULTS: We found that MRTFs localize to IDs in the healthy human heart and accumulate in the nucleus in heart failure. Although mice lacking MRTFs in adult cardiomyocytes display normal cardiac physiology at baseline, pressure overload leads to rapid heart failure characterized by sarcomere disarray, ID disintegration, chamber dilation and wall thinning, cardiac functional decline, and partially penetrant acute lethality. Transcriptional profiling reveals a program of actin cytoskeleton and cardiomyocyte adhesion genes driven by MRTFs during pressure overload. Indeed, conspicuous remodeling of gap junctions at IDs identified by single-molecule localization microscopy may partially stem from a reduction in Mapre1 expression, which we show is a direct mechanosensitive MRTF target. CONCLUSIONS: Our study describes a novel paradigm in which MRTFs control an acute mechanosensitive signaling circuit that coordinates cross-talk between the actin and microtubule cytoskeleton and maintains ID integrity and cardiomyocyte homeostasis in heart disease.


Subject(s)
Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Mechanotransduction, Cellular , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Aged , Animals , Animals, Newborn , COS Cells , Case-Control Studies , Chlorocebus aethiops , Connexin 43/genetics , Connexin 43/metabolism , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Myocytes, Cardiac/ultrastructure , NIH 3T3 Cells , Single Molecule Imaging , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Ventricular Function, Left , Ventricular Remodeling
8.
Am J Pathol ; 188(8): 1794-1806, 2018 08.
Article in English | MEDLINE | ID: mdl-30033030

ABSTRACT

The balance between adaptive and innate immunity in kidney damage in salt-dependent hypertension is unclear. We investigated early renal dysfunction and the influence of Axl, a receptor tyrosine kinase, on innate immune response in hypertensive kidney in mice with lymphocyte deficiency (Rag1-/-). The data suggest that increased presence of CD11b+ myeloid cells in the medulla might explain intensified salt and water retention as well as initial hypertensive response in Rag1-/- mice. Global deletion of Axl on Rag1-/- background reversed kidney dysfunction and accumulation of myeloid cells in the kidney medulla. Chimeric mice that lack Axl in innate immune cells (in the absence of lymphocytes) significantly improved kidney function and abolished early hypertensive response. The bioinformatics analyses of Axl-related gene-gene interaction networks established tissue-specific variation in regulatory pathways. It was confirmed that complement C3 is important for Axl-mediated interactions between myeloid and vascular cells in hypertensive kidney. In summary, innate immunity is crucial for renal dysfunction in early hypertension, and is highly influenced by the presence of Axl.


Subject(s)
Hypertension/immunology , Immunity, Innate/immunology , Kidney Diseases/immunology , Lymphocytes/immunology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Cells, Cultured , Complement C3/metabolism , Homeodomain Proteins/physiology , Hypertension/metabolism , Hypertension/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Axl Receptor Tyrosine Kinase
9.
Proc Natl Acad Sci U S A ; 113(45): E7116-E7125, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27791092

ABSTRACT

Cyclic nucleotide phosphodiesterase 1C (PDE1C) represents a major phosphodiesterase activity in human myocardium, but its function in the heart remains unknown. Using genetic and pharmacological approaches, we studied the expression, regulation, function, and underlying mechanisms of PDE1C in the pathogenesis of cardiac remodeling and dysfunction. PDE1C expression is up-regulated in mouse and human failing hearts and is highly expressed in cardiac myocytes but not in fibroblasts. In adult mouse cardiac myocytes, PDE1C deficiency or inhibition attenuated myocyte death and apoptosis, which was largely dependent on cyclic AMP/PKA and PI3K/AKT signaling. PDE1C deficiency also attenuated cardiac myocyte hypertrophy in a PKA-dependent manner. Conditioned medium taken from PDE1C-deficient cardiac myocytes attenuated TGF-ß-stimulated cardiac fibroblast activation through a mechanism involving the crosstalk between cardiac myocytes and fibroblasts. In vivo, cardiac remodeling and dysfunction induced by transverse aortic constriction, including myocardial hypertrophy, apoptosis, cardiac fibrosis, and loss of contractile function, were significantly attenuated in PDE1C-knockout mice relative to wild-type mice. These results indicate that PDE1C activation plays a causative role in pathological cardiac remodeling and dysfunction. Given the continued development of highly specific PDE1 inhibitors and the high expression level of PDE1C in the human heart, our findings could have considerable therapeutic significance.

10.
Am J Physiol Heart Circ Physiol ; 305(6): H875-84, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23832697

ABSTRACT

Mammalian enabled (Mena) is a key regulator of cytoskeletal actin dynamics, which has been implicated in heart failure (HF). We have previously demonstrated that cardiac Mena deletion produced cardiac dysfunction with conduction abnormalities and hypertrophy. Moreover, elevated Mena expression correlates with HF in human and animal models, yet the precise role of Mena in cardiac pathophysiology is unclear. In these studies, we evaluated mice with cardiac myocyte-specific Mena overexpression (TTA/TgTetMena) comparable to that observed in cardiac pathology. We found that the hearts of TTA/TgTetMena mice were functionally and morphologically comparable to wild-type littermates, except for mildly increased heart mass in the transgenic mice. Interestingly, TTA/TgTetMena mice were particularly susceptible to cardiac injury, as these animals experienced pronounced decreases in ejection fraction and fractional shortening as well as heart dilatation and hypertrophy after transverse aortic constriction (TAC). By "turning off" Mena overexpression in TTA/TgTetMena mice either immediately prior to or immediately after TAC surgery, we discovered that normalizing Mena levels eliminated cardiac hypertrophy in TTA/TgTetMena animals but did not preclude post-TAC cardiac functional deterioration. These findings indicate that hearts with increased levels of Mena fare worse when subjected to cardiac injury and suggest that Mena contributes to HF pathophysiology.


Subject(s)
Cytoskeletal Proteins/metabolism , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Myocardium/metabolism , Ventricular Dysfunction, Left/physiopathology , Animals , Heart Failure/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Organ Size , Up-Regulation , Ventricular Dysfunction, Left/etiology
11.
Am J Physiol Heart Circ Physiol ; 301(5): H1765-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873501

ABSTRACT

Systolic and diastolic dysfunction of the left ventricle (LV) is a hallmark of most cardiac diseases. In vivo assessment of heart function in animal models, particularly mice, is essential to refining our understanding of cardiovascular disease processes. Ultrasound echocardiography has emerged as a powerful, noninvasive tool to serially monitor cardiac performance and map the progression of heart dysfunction in murine injury models. This review covers current applications of small animal echocardiography, as well as emerging technologies that improve evaluation of LV function. In particular, we describe speckle-tracking imaging-based regional LV analysis, a recent advancement in murine echocardiography with proven clinical utility. This sensitive measure enables an early detection of subtle myocardial defects before global dysfunction in genetically engineered and rodent surgical injury models. Novel visualization technologies that allow in-depth phenotypic assessment of small animal models, including perfusion imaging and fetal echocardiography, are also discussed. As imaging capabilities continue to improve, murine echocardiography will remain a critical component of the investigator's armamentarium in translating animal data to enhanced clinical treatment of cardiovascular diseases.


Subject(s)
Echocardiography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Function, Left , Animals , Body Size , Disease Models, Animal , Echocardiography/methods , Image Interpretation, Computer-Assisted , Mice , Predictive Value of Tests , Ventricular Dysfunction, Left/physiopathology
12.
FASEB J ; 24(12): 4917-28, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20724525

ABSTRACT

Growing evidence indicates a critical role of ubiquitin-proteosome system in apoptosis regulation. A cardioprotective effect of ubiquitin (Ub) ligase of the C terminus of Hsc70-interacting protein (CHIP) on myocytes has been reported. In the current study, we found that the cardioprotective effect of insulin growth factor-1 (IGF-1) was mediated by ERK5-CHIP signal module via inducible cAMP early repressor (ICER) destabilization. In vitro runoff assay and Ub assay showed ICER as a substrate of CHIP Ub ligase. Both disruption of ERK5-CHIP binding with inhibitory helical linker domain fragment (aa 101-200) of CHIP and the depletion of ERK5 by siRNA inhibited CHIP Ub ligase activity, which suggests an obligatory role of ERK5 on CHIP activation. Depletion of CHIP, using siRNA, inhibited IGF-1-mediated reduction of isoproterenol-mediated ICER induction and apoptosis. In diabetic mice subjected to myocardial infarction, the CHIP Ub ligase activity was decreased, with an increase in ICER expression. These changes were attenuated significantly in a cardiac-specific constitutively active form of MEK5α transgenic mice (CA-MEK5α-Tg) previously shown to have greater functional recovery. Furthermore, pressure overload-mediated ICER induction was enhanced in heterozygous CHIP(+/-) mice. We identified ICER as a novel CHIP substrate and that the ERK5-CHIP complex plays an obligatory role in inhibition of ICER expression, cardiomyocyte apoptosis, and cardiac dysfunction.


Subject(s)
Cyclic AMP/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Newborn , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cells, Cultured , Cyclic AMP Response Element Modulator/metabolism , Echocardiography , Immunoprecipitation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Binding/genetics , Protein Binding/physiology , Protein Stability , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/genetics
13.
J Am Heart Assoc ; 9(10): e014257, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32394795

ABSTRACT

Background Carotid artery intima/media thickness (IMT) is a hallmark trait associated with future cardiovascular events. The goal of this study was to map new genes that regulate carotid IMT by genome-wide association. Methods and Results We induced IMT by ligation procedure of the left carotid artery in 30 inbred mouse strains. Histologic reconstruction revealed significant variation in left carotid artery intima, media, adventitia, external elastic lamina volumes, intima-to-media ratio, and (intima+media)/external elastic lamina percent ratio in inbred mice. The carotid remodeling trait was regulated by distinct genomic signatures with a dozen common single-nucleotide polymorphisms associated with left carotid artery intima volume, intima-to-media ratio, and (intima+media)/external elastic lamina percent ratio. Among genetic loci on mouse chromosomes 1, 4, and 12, there was natriuretic peptide receptor 2 (Npr2), a strong candidate gene. We observed that only male, not female, mice heterozygous for a targeted Npr2 deletion (Npr2+/-) exhibited defective carotid artery remodeling compared with Npr2 wild-type (Npr2+/+) littermates. Fibrosis in carotid IMT was significantly increased in Npr2+/- males compared with Npr2+/- females or Npr2+/+ mice. We also detected decreased Npr2 expression in human atherosclerotic plaques, similar to that seen in studies in Npr2+/- mice. Conclusions We found that components of carotid IMT were regulated by distinct genetic factors. We also showed a critical role for Npr2 in genetic regulation of vascular fibrosis associated with defective carotid remodeling.


Subject(s)
Carotid Arteries/pathology , Carotid Stenosis/genetics , Genetic Loci , Receptors, Atrial Natriuretic Factor/genetics , Vascular Remodeling/genetics , Animals , Carotid Arteries/diagnostic imaging , Carotid Arteries/metabolism , Carotid Arteries/surgery , Carotid Intima-Media Thickness , Carotid Stenosis/diagnostic imaging , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Disease Models, Animal , Female , Fibrosis , Genome-Wide Association Study , Humans , Ligation , Male , Mice, Inbred Strains , Mice, Knockout , Receptors, Atrial Natriuretic Factor/metabolism , Sex Factors , Species Specificity
14.
Circ Heart Fail ; 12(4): e005565, 2019 04.
Article in English | MEDLINE | ID: mdl-30998392

ABSTRACT

Background Heart failure (HF) is invariably accompanied by development of cardiac fibrosis, a form of scarring that increases muscular tissue rigidity and decreases cardiac contractility. Cardiac fibrosis arises from a pathological attempt to repair tissue damaged during maladaptive remodeling. Treatment options to block or reverse fibrosis have proven elusive. Neprilysin is an endopeptidase that degrades vasoactive peptides, including atrial natriuretic peptide. Thus, neprilysin inhibition reduces hypertension, ultimately limiting maladaptive cardiac remodeling. LCZ696, which consists of an angiotensin receptor blocker (valsartan [VAL]) and a neprilysin inhibitor (sacubitril [SAC]), was shown to be well tolerated and significantly reduced the risk of death and hospitalization in HF patients with reduced ejection fraction. We hypothesized that SAC/VAL directly inhibits fibroblast activation and development of pathological fibrosis. Methods and Results We used a mouse model of left ventricle pressure overload coupled to in vitro studies in primary mouse and human cardiac fibroblasts (CFs) to study the impact of SAC/VAL on CF activation and cardiac fibrosis. SAC/VAL significantly ameliorated pressure overload-induced cardiac fibrosis by blocking CF activation and proliferation, leading to functional improvement. Mechanistically, the beneficial impact of SAC/VAL at least partially stemmed from restoration of PKG (protein kinase G) signaling in HF patient-derived CF, which inhibited Rho activation associated with myofibroblast transition. Conclusions This study reveals that SAC/VAL acts directly on CF to prevent maladaptive cardiac fibrosis and dysfunction during pressure overload-induced hypertrophy and suggests that SAC/VAL should be evaluated as a direct antifibrotic therapeutic for conditions such as HF with preserved ejection fraction.


Subject(s)
Aminobutyrates/pharmacology , Cyclic GMP-Dependent Protein Kinases/drug effects , Fibroblasts/drug effects , Heart Failure/drug therapy , Heart Ventricles/drug effects , Tetrazoles/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Animals , Biphenyl Compounds , Drug Combinations , Fibroblasts/metabolism , Fibrosis/drug therapy , Heart/drug effects , Heart/physiopathology , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Male , Mice, Inbred C57BL , Neprilysin/antagonists & inhibitors , Valsartan
15.
Sci Rep ; 7(1): 12081, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935983

ABSTRACT

Increased arterial thickness measured with ultrasound correlates with future cardiovascular events, but conventional ultrasound imaging techniques cannot distinguish between intima, media, or atherosclerotic plaque in the carotid artery. In this work, we evaluated how well vascular elastography can detect intimal changes in a mouse model of carotid remodeling. We ligated the left external and internal branches of the carotid artery of male FVB mice and performed sham operations for 2 weeks. High-resolution ultrasound imaging accurately detected lower blood velocities and low blood volume flow in the carotid arteries after ligation in FVB mice. However, ultrasound could not detect differences in the carotid wall even at 2 weeks post-surgery. The Young's modulus was measured based on displacements of the carotid artery wall, and Young's modulus was 2-fold greater in shams at 1 week post ligation, and 3-fold greater 2 weeks after ligation. Finally, the higher Young's modulus was most associated with higher intimal thickness but not medial or adventitial thickness as measured by histology. In conclusion, we developed a robust ultrasound-based elastography method for early detection of intimal changes in small animals.


Subject(s)
Carotid Arteries/physiology , Elastic Modulus/physiology , Elasticity Imaging Techniques/methods , Vascular Remodeling/physiology , Algorithms , Animals , Blood Flow Velocity/physiology , Carotid Arteries/diagnostic imaging , Carotid Arteries/surgery , Carotid Intima-Media Thickness , Ligation , Male , Mice, Inbred Strains , Models, Cardiovascular , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/physiopathology , Ultrasonography/methods
16.
J Am Coll Cardiol ; 63(23): 2549-2557, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24703913

ABSTRACT

OBJECTIVES: The authors propose simultaneous inhibition of Gßγ signaling in the heart and the adrenal gland as a novel therapeutic approach for heart failure (HF). BACKGROUND: Elevated sympathetic nervous system activity is a salient characteristic of HF progression. It causes pathologic desensitization of ß-adrenergic receptors (ß-AR), facilitated predominantly through Gßγ-mediated signaling. The adrenal glands are key contributors to the chronically elevated plasma catecholamine levels observed in HF, where adrenal α2-AR feedback inhibitory function is impaired also through Gßγ-mediated signaling. METHODS: We investigated the efficacy of a small molecule Gßγ inhibitor, gallein, in a clinically relevant, pressure-overload model of HF. RESULTS: Daily gallein treatment (10 mg/kg/day), initiated 4 weeks after transverse aortic constriction, improved survival and cardiac function and attenuated cardiac remodeling. Mechanistically, gallein restored ß-AR membrane density in cardiomyocytes, attenuated Gßγ-mediated G-protein-coupled receptor kinase 2-phosphoinositide 3-kinase γ membrane recruitment, and reduced Akt (protein kinase B) and glycogen synthase kinase 3ß phosphorylation. Gallein also reduced circulating plasma catecholamine levels and catecholamine production in isolated mouse adrenal glands by restoring adrenal α2-AR feedback inhibition. In human adrenal endocrine tumors (pheochromocytoma), gallein attenuated catecholamine secretion, as well as G-protein-coupled receptor kinase 2 expression and membrane translocation. CONCLUSIONS: These data suggest small molecule Gßγ inhibition as a systemic pharmacologic therapy for HF by simultaneously normalizing pathologic adrenergic/Gßγ signaling in both the heart and the adrenal gland. Our data also suggest important endocrine/cardiovascular interactions and a possible role for small molecule Gßγ inhibition in treating endocrine tumors such as pheochromocytoma, in addition to HF.


Subject(s)
Adrenal Glands/metabolism , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Heart Failure/drug therapy , Heart/drug effects , Myocardium/metabolism , Xanthenes/therapeutic use , Adrenal Glands/drug effects , Adrenal Glands/pathology , Animals , Cells, Cultured , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction , Myocardium/pathology , Stroke Volume , Treatment Outcome , Xanthenes/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL