Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Circulation ; 139(9): 1185-1198, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30586715

ABSTRACT

BACKGROUND: Inflammatory response after myocardial infarction (MI) is essential for cardiac healing, whereas excessive and prolonged inflammation extends the infarction and promotes adverse cardiac remodeling. Understanding the mechanistic insight of these tightly controlled inflammatory processes has a significant impact on post-MI recovery and therapy. Here, we uncover the critical role of small GTPase RhoE in post-MI recovery and its clinical implication. METHODS: Three genetic mouse lines are used: global RhoE knockout, cardiomyocyte-specific RhoE heterozygous, and cardiomyocyte-specific RhoE overexpression mice. A set of molecular signaling experiments, including bimolecular fluorescence complementation, immunoprecipitation, electrophoretic mobility shift assay, and mRNA microarray analysis, were conducted. Permanent ligation of the left anterior descending artery was performed, followed by the assessments of cardiac function, inflammation, and survival in the first week after MI. Finally, we examined the correlation of the expression levels of RhoE in MI patient heart and patient prognosis. RESULTS: RhoE deficiency turns on a group of proinflammatory gene expressions in mouse heart. Mice with cardiomyocyte-specific haploinsufficiency exhibit excessive inflammatory response with deleterious cardiac function after MI. A profound increase in nuclear factor-κB activity is detected in the mutant heart and the isolated cardiomyocytes. We further find that the expression of RhoE is upregulated in response to MI. Mechanistically, RhoE interacts with p65 and p50 individually in cytosol and blocks their nuclear translocation. RhoE also occupies the dimerization domain of p65 and subsequently disrupts the heterodimerization between p65 and p50. Cardiac RhoE overexpression inhibits nuclear factor-κB activity, restrains post-MI inflammation, and improves cardiac function and survival. Consistently, we find that the expression level of RhoE is elevated in the heart of patients with MI and that the patients with a higher expression level of RhoE exhibit a better prognosis in cardiac function recovery. CONCLUSIONS: The study uncovers RhoE as a new fine-tuning factor modulating MI-induced inflammation and promoting injured heart recovery. RhoE may serve as a new potential biomarker for the assessment of MI patient prognosis. Manipulation of RhoE could be as a potential therapeutic approach for MI and other inflammatory diseases.


Subject(s)
Gene Expression Regulation , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , rho GTP-Binding Proteins/genetics
2.
Biochim Biophys Acta Mol Cell Res ; 1864(4): 697-707, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28130125

ABSTRACT

Skeletal muscle cell proliferation and differentiation are tightly regulated. Epigenetic regulation is a major component of the regulatory mechanism governing these processes. Histone modification is part of the epigenetic code used for transcriptional regulation of chromatin through the establishment of an active or repressive state for genes involved in myogenesis in a temporal manner. Here, we uncovered the function of SET domain containing 2 (Setd2), an essential histone 3 lysine 36 trimethyltransferase, in regulating the proliferation and differentiation of myoblasts. Setd2 was silenced in the skeletal muscle myoblast cell line, C2C12, using the CRISPR/CAS9 system. The mutant cells exhibited defect in myotube formation. The myotube formation marker, myosin heavy chain (MHC), was downregulated earlier in Setd2 silenced cells compared to wild-type myoblasts during differentiation. The deficiency in Setd2 also resulted in repression of Myogenin (MyoG) expression, a key myogenic regulator during differentiation. In addition to the myoblast differentiation defect, decreased proliferation rate with significantly reduced levels of histone 3 phosphorylation, indicative of cell proliferation defect, were observed in the Setd2 silenced cells; suggesting an impaired proliferation phenotype. Furthermore, compromised G1/S- and G2/M-phase transition and decreased expression levels of major regulators of cell cycle G1/S checkpoints, cyclin D1, CDK4, CDK6, and cyclin E2 were detected in Setd2 silenced cells. Consistent with the cell cycle arrested phenotype, cyclin-dependent kinase inhibitor p21 was upregulated in Setd2 silenced cells. Together, this study demonstrates an essential role of Setd2 in myoblast proliferation and differentiation, and uncovers Setd2-mediated molecular mechanism through regulating MyoG and p21.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Myogenin/genetics , Animals , Base Sequence , CRISPR-Cas Systems , Cell Cycle Checkpoints , Cell Differentiation , Cell Line , Cell Proliferation , Chromatin/chemistry , Chromatin/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/genetics , Cyclins/metabolism , Gene Editing , Gene Silencing , Histone-Lysine N-Methyltransferase/deficiency , Histones/metabolism , Mice , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Myogenin/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phosphorylation
3.
Hypertension ; 67(3): 597-605, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26781283

ABSTRACT

The insufficiency of compensatory angiogenesis in the heart of patients with hypertension contributes to heart failure transition. The hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling cascade controls responsive angiogenesis. One of the challenges in reprograming the insufficient angiogenesis is to achieve a sustainable tissue exposure to the proangiogenic factors, such as HIF1α stabilization. In this study, we identified Rnd3, a small Rho GTPase, as a proangiogenic factor participating in the regulation of the HIF1α-VEGF signaling cascade. Rnd3 physically interacted with and stabilized HIF1α, and consequently promoted VEGFA expression and endothelial cell tube formation. To demonstrate this proangiogenic role of Rnd3 in vivo, we generated Rnd3 knockout mice. Rnd3 haploinsufficient (Rnd3(+/-)) mice were viable, yet developed dilated cardiomyopathy with heart failure after transverse aortic constriction stress. The poststress Rnd3(+/-) hearts showed significantly impaired angiogenesis and decreased HIF1α and VEGFA expression. The angiogenesis defect and heart failure phenotype were partially rescued by cobalt chloride treatment, a HIF1α stabilizer, confirming a critical role of Rnd3 in stress-responsive angiogenesis. Furthermore, we generated Rnd3 transgenic mice and demonstrated that Rnd3 overexpression in heart had a cardioprotective effect through reserved cardiac function and preserved responsive angiogenesis after pressure overload. Finally, we assessed the expression levels of Rnd3 in the human heart and detected significant downregulation of Rnd3 in patients with end-stage heart failure. We concluded that Rnd3 acted as a novel proangiogenic factor involved in cardiac responsive angiogenesis through HIF1α-VEGFA signaling promotion. Rnd3 downregulation observed in patients with heart failure may explain the insufficient compensatory angiogenesis involved in the transition to heart failure.


Subject(s)
Coronary Vessels/pathology , Gene Expression Regulation , Hypertension/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/genetics , rho GTP-Binding Proteins/genetics , Animals , Blotting, Western , Coronary Vessels/metabolism , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hypertension/metabolism , Hypertension/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , RNA/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/biosynthesis , rho GTP-Binding Proteins/biosynthesis
4.
Cancer Med ; 4(9): 1404-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26108681

ABSTRACT

Activation of Notch signaling contributes to glioblastoma multiform (GBM) tumorigenesis. However, the molecular mechanism that promotes the Notch signaling augmentation during GBM genesis remains largely unknown. Identification of new factors that regulate Notch signaling is critical for tumor treatment. The expression levels of RND3 and its clinical implication were analyzed in GBM patients. Identification of RND3 as a novel factor in GBM genesis was demonstrated in vitro by cell experiments and in vivo by a GBM xenograft model. We found that RND3 expression was significantly decreased in human glioblastoma. The levels of RND3 expression were inversely correlated with Notch activity, tumor size, and tumor cell proliferation, and positively correlated with patient survival time. We demonstrated that RND3 functioned as an endogenous repressor of the Notch transcriptional complex. RND3 physically interacted with NICD, CSL, and MAML1, the Notch transcriptional complex factors, promoted NICD ubiquitination, and facilitated the degradation of these cofactor proteins. We further revealed that RND3 facilitated the binding of NICD to FBW7, a ubiquitin ligase, and consequently enhanced NICD protein degradation. Therefore, Notch transcriptional activity was inhibited. Forced expression of RND3 repressed Notch signaling, which led to the inhibition of glioblastoma cell proliferation in vitro and tumor growth in the xenograft mice in vivo. Downregulation of RND3, however, enhanced Notch signaling activity, and subsequently promoted glioma cell proliferation. Inhibition of Notch activity abolished RND3 deficiency-mediated GBM cell proliferation. We conclude that downregulation of RND3 is responsible for the enhancement of Notch activity that promotes glioblastoma genesis.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Receptors, Notch/metabolism , rho GTP-Binding Proteins/genetics , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Heterografts , Humans , Male , Mice , Multiprotein Complexes/metabolism , Protein Binding , Proteolysis , Signal Transduction , Transcriptional Activation , Tumor Burden , Ubiquitination , rho GTP-Binding Proteins/metabolism
5.
Compr Physiol ; 6(1): 169-86, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26756630

ABSTRACT

Rnd3, also known as RhoE, belongs to the Rnd subclass of the Rho family of small guanosine triphosphate (GTP)-binding proteins. Rnd proteins are unique due to their inability to switch from a GTP-bound to GDP-bound conformation. Even though studies of the biological function of Rnd3 are far from being concluded, information is available regarding its expression pattern, cellular localization, and its activity, which can be altered depending on the conditions. The compiled data from these studies implies that Rnd3 may not be a traditional small GTPase. The basic role of Rnd3 is to report as an endogenous antagonist of RhoA signaling-mediated actin cytoskeleton dynamics, which specifically contributes to cell migration and neuron polarity. In addition, Rnd3 also plays a critical role in arresting cell cycle distribution, inhibiting cell growth, and inducing apoptosis and differentiation. Increasing data have shown that aberrant Rnd3 expression may be the leading cause of some systemic diseases; particularly highlighted in apoptotic cardiomyopathy, developmental arrhythmogenesis and heart failure, hydrocephalus, as well as tumor metastasis and chemotherapy resistance. Therefore, a better understanding of the function of Rnd3 under different physiological and pathological conditions, through the use of suitable models, would provide a novel insight into the origin and treatment of multiple human diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Neoplasms/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Cardiovascular Diseases/genetics , Humans , Neoplasms/genetics , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...