Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Respir Res ; 24(1): 118, 2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37120511

ABSTRACT

Increased apoptosis of alveolar epithelial cells is a prominent feature of pulmonary fibrosis. Macrophage efferocytosis, phagocytosis of apoptotic cells by macrophages, is crucial for maintaining tissue homeostasis. Expression of Mer tyrosine kinase (MERTK, an important recognition receptor in efferocytosis) in macrophages is thought to be associated with fibrosis. However, how macrophage MERTK affects pulmonary fibrosis and whether it depends on efferocytosis are not yet clear. Here, we found elevated MERTK expression in lung macrophages from IPF patients and mice with bleomycin-induced pulmonary fibrosis. In vitro experiments showed that macrophages overexpressing MERTK exhibit profibrotic effects and that macrophage efferocytosis abrogates the profibrotic effect of MERTK by downregulating MERTK, forming a negative regulatory loop. In pulmonary fibrosis, this negative regulation is defective, and MERTK mainly exhibits profibrotic effects. Our study reveals a previously unsuspected profibrotic effect of elevated macrophage MERTK in pulmonary fibrosis and defective regulation of efferocytosis function as a result of that elevation, suggesting that targeting MERTK in macrophages may help to attenuate pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , Receptor Protein-Tyrosine Kinases , Animals , Mice , Apoptosis , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Macrophages/metabolism , Phagocytosis , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Receptor Protein-Tyrosine Kinases/genetics
3.
Curr Microbiol ; 78(10): 3656-3666, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34338833

ABSTRACT

Influenza A, influenza B, severe acute respiratory syndrome coronavirus 2, adenovirus, respiratory syncytial virus, Mycoplasma pneumoniae, and Chlamydophila pneumoniae are common pathogens that can cause severe pneumonia and other symptoms, resulting in acute lower respiratory tract infections. The objective of this study was to design and evaluate a sensitive and specific multiplex one-step reverse transcription PCR (RT-PCR)-dipstick chromatography method for simultaneous rapid detection of these seven pathogens. Streptavidin-coated blue latex particles were used to read out a positive signal. Based on the DNA-DNA hybridization of oligonucleotide sequences (Tag) for forward primer with the complementary oligonucleotide sequence (cTag) on the dipstick and biotin-streptavidin interactions, PCR products were able to be illuminated visually on the dipstick. The specificity and the limit of detection (LOD) were also evaluated. Moreover, the clinical performance of this method was compared with Sanger sequencing for 896 samples. No cross reaction with other pathogens was found, confirming the high specificity of this method. The LOD was 10 copies/µL for each of the tested pathogens, and the whole procedure took less than 40 min. Using 896 samples, the sensitivity and specificity were shown to be no lower than 94.5%. The positive predictive value was higher than 82.1%, and the negative predictive value was higher than 99.5%. The kappa value between the PCR-dipstick chromatography method and Sanger sequencing ranged from 0.869 to 0.940. In summary, our one-step RT-PCR-dipstick chromatography method is a sensitive and specific tool for rapidly detecting multiplex respiratory pathogens.


Subject(s)
COVID-19 , Reverse Transcription , Chromatography , Humans , Multiplex Polymerase Chain Reaction , SARS-CoV-2 , Sensitivity and Specificity
4.
Sci Rep ; 11(1): 3138, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542447

ABSTRACT

Liquid biopsy testing utilising Next Generation Sequencing (NGS) is rapidly moving towards clinical adoption for personalised oncology. However, before NGS can fulfil its potential any novel testing approach must identify ways of reducing errors, allowing separation of true low-frequency mutations from procedural artefacts, and be designed to improve upon current technologies. Popular NGS technologies typically utilise two DNA capture approaches; PCR and ligation, which have known limitations and seem to have reached a development plateau with only small, stepwise improvements being made. To maximise the ultimate utility of liquid biopsy testing we have developed a highly versatile approach to NGS: Adaptor Template Oligo Mediated Sequencing (ATOM-Seq). ATOM-Seq's strengths and versatility avoid the major limitations of both PCR- and ligation-based approaches. This technology is ligation free, simple, efficient, flexible, and streamlined, and it offers novel advantages that make it perfectly suited for use on highly challenging clinical material. Using reference and clinical materials, we demonstrate detection of known SNVs down to allele frequencies of 0.1% using as little as 20-25 ng of cfDNA, as well as the ability to detect fusions from RNA. We illustrate ATOM-Seq's suitability for clinical testing by showing high concordance rates between paired cfDNA and FFPE clinical samples.


Subject(s)
Circulating Tumor DNA/genetics , Colonic Neoplasms/diagnosis , High-Throughput Nucleotide Sequencing/methods , Lung Neoplasms/diagnosis , RNA, Neoplasm/genetics , Alleles , Base Sequence , Circulating Tumor DNA/blood , Colonic Neoplasms/blood , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Primers/chemical synthesis , DNA Primers/metabolism , Gene Frequency , Gene Library , Humans , Liquid Biopsy , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Polymorphism, Single Nucleotide , RNA, Neoplasm/blood , Sensitivity and Specificity
5.
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
7.
Oncotarget ; 7(50): 82411-82423, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27705942

ABSTRACT

Activation of Snail1 signaling promotes the migration and invasion of multiple tumors, including glioblastoma multiforme (GBM). However, the molecular mechanism that augments Snail1 signaling during GBM cell migration and invasion remains largely unknown. Identification of the factors that regulate Snail1 signaling is critical to block tumor cell migration and invasion. By screening human GBM specimens, we found that the expression levels of small GTPase RND3 positively correlated with the expression levels of E-cadherin and claudin, the glioblastoma migration biomarkers negatively regulated by Snail1. Downregulation of E-cadherin and claudin has been associated with the migration and invasion of GBM cells. We demonstrated that RND3 functioned as an endogenous inhibitor of the Snail-directed transcriptional regulation. RND3 physically interacted with Snail1 protein, enhanced Snail1 ubiquitination, and facilitated the protein degradation. Forced expression of RND3 inhibited Snail1 activity, which in turn blocked glioblastoma cell migration and invasion in vitro in cell culture and in vivo in GBM xenograft mice. In contrast, downregulation of RND3 augmented Snail1 activity, and subsequently decreased E-cadherin expression, eventually promoted glioblastoma cell migration and invasion. The pro-migration induced by RND3 downregulation was attenuated by Snail1 knockdown. The findings partially explain why Snail1 activity is augmented in GBM, and defines a new function of RND3 in GBM cell migration and invasion.


Subject(s)
Brain Neoplasms/enzymology , Cell Movement , Glioblastoma/enzymology , Snail Family Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Antigens, CD , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Claudins/genetics , Claudins/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice, Nude , Neoplasm Invasiveness , Protein Binding , Proteolysis , RNA Interference , Signal Transduction , Snail Family Transcription Factors/genetics , Transfection , Ubiquitination , rho GTP-Binding Proteins/genetics
8.
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
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 35(9): 1283-6, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26403739

ABSTRACT

OBJECTIVE: To investigate the mechanism of miR-133a in reversing neonatal rat cardiomyocyte hypertrophy induced by phenylephrine. METHODS: A miR-133a precursor cDNA was used to construct an adenovirus vector, which was transfected into 293 cells to harvest miR-133a-containing virus. Neonatal rat cardiac myocytes treated by phenylephrine were exposed to miR-133a adenovirus, and the changes in cell area was measured; the expression levels of miR-133a and Acta1, Actc1, Actb, Myh6, Myh7, and BNP mRNAs were detected by quantitative RT-PCR. RESULTS: Phenylephrine treatment increased the area of cardiomyocytes by more than 3 folds and significantly enhanced the expression levels of Acta1, Actc1, Actb, Myh6, Myh7 and BNP mRNAs. All these changes were obviously reverse by miR-133a treatment. CONCLUSION: miR-133a is an important regulator of phenylephrine-induced cardiomyocyte hypertrophy and negatively regulates this process.


Subject(s)
MicroRNAs/genetics , Myocytes, Cardiac/pathology , Phenylephrine/adverse effects , Adenoviridae , Animals , Cells, Cultured , Genetic Vectors , Hypertrophy , Myocytes, Cardiac/cytology , RNA, Messenger , Rats , Transfection
10.
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
11.
Circ Res ; 116(1): e1-e10, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25348166

ABSTRACT

RATIONALE: Rnd3, a small Rho GTPase, is involved in the regulation of cell actin cytoskeleton dynamics, cell migration, and proliferation. The biological function of Rnd3 in the heart remains unexplored. OBJECTIVE: To define the functional role of the Rnd3 gene in the animal heart and investigate the associated molecular mechanism. METHODS AND RESULTS: By loss-of-function approaches, we discovered that Rnd3 is involved in calcium regulation in cardiomyocytes. Rnd3-null mice died at the embryonic stage with fetal arrhythmias. The deletion of Rnd3 resulted in severe Ca(2+) leakage through destabilized ryanodine receptor type 2 Ca(2+) release channels. We further found that downregulation of Rnd3 attenuated ß2-adrenergic receptor lysosomal targeting and ubiquitination, which in turn resulted in the elevation of ß2-adrenergic receptor protein levels leading to the hyperactivation of protein kinase A (PKA) signaling. The PKA activation destabilized ryanodine receptor type 2 channels. This irregular spontaneous Ca(2+) release can be curtailed by PKA inhibitor treatment. Increases in the PKA activity along with elevated cAMP levels were detected in Rnd3-null embryos, in neonatal rat cardiomyocytes, and noncardiac cell lines with Rnd3 knockdown, suggesting a general mechanism for Rnd3-mediated PKA signaling activation. ß2-Adrenergic receptor blocker treatment reduced arrhythmia and improved cardiac function. CONCLUSIONS: Rnd3 is a novel factor involved in intracellular Ca(2+) homeostasis regulation in the heart. Deficiency of the protein induces ryanodine receptor type 2 dysfunction by a mechanism that attenuates Rnd3-mediated ß2-adrenergic receptor ubiquitination, which leads to the activation of PKA signaling. Increased PKA signaling in turn promotes ryanodine receptor type 2 hyperphosphorylation, which contributes to arrhythmogenesis and heart failure.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Gene Deletion , Myocytes, Cardiac/metabolism , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics , Animals , Animals, Newborn , Cells, Cultured , Female , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Signal Transduction/physiology , Up-Regulation/physiology
12.
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
13.
PLoS One ; 8(9): e75470, 2013.
Article in English | MEDLINE | ID: mdl-24058688

ABSTRACT

Transcriptional regulation is essential for any gene expression including microRNA expression. MiR-1-1 and miR-133a-2 are essential microRNAs (miRs) involved in cardiac and skeletal muscle development and diseases. Early studies reveal two regulatory enhancers, an upstream and an intragenic, that direct the miR-1-1 and miR-133a-2 transcripts. In this study, we identify a unique serum response factor (SRF) binding motif within the enhancer through bioinformatic approaches. This motif is evolutionarily conserved and is present in a range of organisms from yeast, flies, to humans. We provide evidence to demonstrate that this regulatory motif is SRF-dependent in vitro by electrophoretic mobility shift assay, luciferase activity assay, and endogenous chromatin immunoprecipitation assay followed by DNA sequence confirmation, and in vivo by transgenic lacZ reporter mouse studies. Importantly, our transgenic mice indicate that this motif is indispensable for the expression of miR1-1/133a-2 in the heart, but not necessary in skeletal muscle, while the enhancer is sufficient for miR1-1/133a-2 gene expression in both tissues. The mutation of the motif alone completely abolishes miR-1-1/133a-2 gene expression in the animal heart, but not in the skeletal muscle. Our findings reveal an additional architecture of regulatory complex directing miR-1-1/133a-1 gene expression, and demonstrate how this intragenic enhancer differentially manages the expression of the two miRs in the heart and skeletal muscle, respectively.


Subject(s)
Gene Expression Regulation/physiology , MicroRNAs/biosynthesis , Myoblasts, Cardiac/metabolism , Myocardium/metabolism , Serum Response Element/physiology , Animals , Cell Line , Mice , Mice, Transgenic , MicroRNAs/genetics , Myoblasts, Cardiac/cytology , Myocardium/cytology , Organ Specificity/physiology
14.
Proc Natl Acad Sci U S A ; 110(20): 8236-41, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23630292

ABSTRACT

Rho family guanosine triphosphatase (GTPase) 3 (Rnd3), a member of the small Rho GTPase family, is involved in the regulation of cell actin cytoskeleton dynamics, cell migration, and proliferation through the Rho kinase-dependent signaling pathway. We report a role of Rnd3 in the pathogenesis of hydrocephalus disorder. Mice with Rnd3 genetic deletion developed severe obstructive hydrocephalus with enlargement of the lateral and third ventricles, but not of the fourth ventricles. The cerebral aqueducts in Rnd3-null mice were partially or completely blocked by the overgrowth of ependymal epithelia. We examined the molecular mechanism contributing to this Rnd3-deficiency-mediated hydrocephalus and found that Rnd3 is a regulator of Notch signaling. Rnd3 deficiency, through either gene deletion or siRNA knockdown, resulted in a decrease in Notch intracellular domain (NICD) protein degradation. However, there was no correlated change in mRNA change, which in turn led to an increase in NICD protein levels. Immunoprecipitation analysis demonstrated that Rnd3 and NICD physically interacted, and that down-regulation of Rnd3 attenuated NICD protein ubiquitination. This eventually enhanced Notch signaling activity and promoted aberrant growth of aqueduct ependymal cells, resulting in aqueduct stenosis and the development of congenital hydrocephalus. Inhibition of Notch activity rescued the hydrocephalus disorder in the mutant animals.


Subject(s)
Ependyma/cytology , Gene Deletion , Hydrocephalus/metabolism , Receptors, Notch/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Brain/abnormalities , Ependyma/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutation , Oligonucleotide Array Sequence Analysis , Protein Processing, Post-Translational , RNA, Small Interfering/metabolism , Signal Transduction , Up-Regulation , rho GTP-Binding Proteins/genetics
15.
FASEB J ; 26(5): 2105-16, 2012 May.
Article in English | MEDLINE | ID: mdl-22278938

ABSTRACT

We have previously found that in failing human hearts, Rho-associated coiled-coil protein kinase 1 (ROCK1) is processed by caspase-3 into an active isoform, ROCKΔ1. The purpose of the current investigation was to elucidate the pathological consequences of truncated ROCK1 accumulation in the heart, the associated molecular mechanism of ROCKΔ1-mediated cardiac phenotype, and the molecular signaling between Rho kinase activation in cardiomyocytes and extracellular matrix response. We generated transgenic mice expressing ROCKΔ1 in cardiomyocytes to mimic the situation observed in human heart disease, whereas an additional kinase-deficient mouse was generated as a control. The ROCKΔ1 transgenic mice developed fibrotic cardiomyopathy with diastolic dysfunction. Transgenic hearts displayed activated TGFß1 and NF-κB signaling and a release of a subset of cytokines and were susceptible to angiotensin II stress. Treatment with a Rho kinase inhibitor attenuated the fibrotic phenotype. Cardiac fibroblasts differentiated into myofibroblasts when cocultured with transgenic cardiomyocytes but not with wild-type cardiomyocytes. Inhibitors of Rho kinase as well as TGFßR1 and NF-κB decreased these effects. The serum response factor-dependent TGFß1 regulation was shown to be responsible for the Rho kinase-mediated activation of TGFß1 signaling. We conclude that ROCKΔ1 is a novel fibrotic factor. Activation of TGFß1 and NF-κB signaling contributes to the Rho kinase-mediated pathological fibrosis.


Subject(s)
Cardiomyopathies/enzymology , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Fibrosis , Mice , Mice, Transgenic , Polymerase Chain Reaction , Rats , Transforming Growth Factor beta1/metabolism , rho-Associated Kinases/metabolism
16.
Mol Cell Biol ; 32(2): 297-308, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22106411

ABSTRACT

Protein tyrosine phosphatase-like A (PTPLa) has been implicated in skeletal myogenesis and cardiogenesis. Mutations in PTPLa correlated with arrhythmogenic right ventricular dysplasia in humans and congenital centronuclear myopathy with severe hypotonia in dogs. The molecular mechanisms of PTPLa in myogenesis are unknown. In this report, we demonstrate that PTPLa is required for myoblast growth and differentiation. The cells lacking PTPLa remained immature and failed to differentiate into mature myotubes. The repressed MyoG expression was responsible for the impaired myoblast differentiation. Meanwhile, impeded cell growth, with an obvious S-phase arrest and compromised G(2)/M transition, was observed in PTPLa-deficient myoblasts. Further study demonstrated that the upregulation of cyclin D1 and cyclin E2 complexes, along with a compromised G(2)/M transition due to the decreased CDK1 (cyclin-dependent kinase 1) activity and upregulated p21, contributed to the mutant cell S-phase arrest and eventually led to the retarded cell growth. Finally, the transcriptional regulation of the PTPLa gene was explored. We identified PTPLa as a new target gene of the serum response factor (SRF). Skeletal- and cardiac-muscle-specific SRF knockouts resulted in significant decreases in PTPLa expression, suggesting a conserved transcriptional regulation of the PTPLa gene in mice.


Subject(s)
Myoblasts/cytology , Myogenin/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Animals , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Myocardium/metabolism , Myogenin/genetics , Protein Tyrosine Phosphatases/genetics , Serum Response Factor/genetics , Serum Response Factor/metabolism
17.
Cell Res ; 22(2): 360-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21769134

ABSTRACT

Enteroviral infection can lead to dilated cardiomyopathy (DCM), which is a major cause of cardiovascular mortality worldwide. However, the pathogenetic mechanisms have not been fully elucidated. Serum response factor (SRF) is a cardiac-enriched transcription regulator controlling the expression of a variety of target genes, including those involved in the contractile apparatus and immediate early response, as well as microRNAs that silence the expression of cardiac regulatory factors. Knockout of SRF in the heart results in downregulation of cardiac contractile gene expression and development of DCM. The goal of this study is to understand the role of SRF in enterovirus-induced cardiac dysfunction and progression to DCM. Here we report that SRF is cleaved following enteroviral infection of mouse heart and cultured cardiomyocytes. This cleavage is accompanied by impaired cardiac function and downregulation of cardiac-specific contractile and regulatory genes. Further investigation by antibody epitope mapping and site-directed mutagenesis demonstrates that SRF cleavage occurs at the region of its transactivation domain through the action of virus-encoded protease 2A. Moreover, we demonstrate that cleavage of SRF dissociates its transactivation domain from DNA-binding domain, resulting in the disruption of SRF-mediated gene transactivation. In addition to loss of functional SRF, finally we report that the N-terminal fragment of SRF cleavage products can also act as a dominant-negative transcription factor, which likely competes with the native SRF for DNA binding. Our results suggest a mechanism by which virus infection impairs heart function and may offer a new therapeutic strategy to ameliorate myocardial damage and progression to DCM.


Subject(s)
Myocytes, Cardiac/metabolism , Peptide Hydrolases/metabolism , Serum Response Factor/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/metabolism , Caspase Inhibitors , Caspases/metabolism , Cell Line , Enterovirus/enzymology , Gene Expression , HeLa Cells , Heart/physiopathology , Humans , Male , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Serum Response Factor/chemistry , Serum Response Factor/genetics , Virus Replication
18.
Am J Physiol Heart Circ Physiol ; 298(5): H1340-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20173049

ABSTRACT

Activation of NFAT (nuclear factor of activated T cells)-mediated hypertrophic signaling is a major regulatory response to hypertrophic stimuli. A recent study unveiled potential regulatory roles for microRNA-133a (miR-133a) in cardiac hypertrophy. To date, however, no connection has been made between miR-133a and NFAT signaling. In this study, we determined that NFATc4, a hypertrophy-associated mediator, is negatively regulated by miR-133a. Two conserved base-pairing sites between the NFATc4 3'-untranslated region (UTR) and miR-133a were verified. Mutation of these sites in the NFATc4 3'-UTR completely blocked the negative effect of miR-133a on NFATc4, suggesting that NFATc4 is a direct target for miR-133a regulation. Using a gain-of-function approach, we demonstrate that miR-133 significantly reduces the endogenous level of, as well as the hypertrophic stimulus-mediated increase in, NFATc4 gene expression. This latter effect of miR-133a on NFATc4 gene expression was coincided with an attenuated cardiomyocyte hypertrophy induced by an alpha-adrenergic receptor agonist. Conversely, cells treated with miR-133a inhibitor resulted in an increase in NFATc4 expression level. Application of miR-133a had no apparent effect on NFATc4 nuclear localization. We conclude that the negative regulation of NFATc4 expression contributes to miR-133a-mediated hypertrophic repression.


Subject(s)
Cardiomegaly/metabolism , MicroRNAs/physiology , Myocytes, Cardiac/drug effects , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , 3' Untranslated Regions/genetics , Adenoviridae/genetics , Animals , Animals, Newborn , Base Sequence , Blotting, Western , Cardiomegaly/pathology , Cell Size/drug effects , Cells, Cultured , Computational Biology , Genetic Vectors , Immunohistochemistry , Luciferases/metabolism , Male , Myocytes, Cardiac/ultrastructure , Plasmids/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 23(4): 410-4, 2006 Aug.
Article in Chinese | MEDLINE | ID: mdl-16883528

ABSTRACT

OBJECTIVE: To investigate the association that the polymorphisms of interleukin 10 gene (IL10) promoter region are related to the susceptibility and clinical phenotypes of hepatitis B virus (HBV) in Chinese Han population. METHODS: With polymerase chain reaction with restriction fragment length polymorphism (PCR-RFLP) method, the single nucleotide polymorphisms (SNP) of the promoter region of IL10 gene at position -1082G/A, -819T/C, -592A/C were detected in 231 patients with chronic hepatitis B, 165 individuals spontaneously recovered from HBV infection and 135 normal controls. RESULTS: No significant difference was found in frequencies of genotypes and alleles of IL10 gene promoter region at position -1082G/A, -819T/C, -592A/C among normal controls, individuals spontaneously recovering from HBV infection and patients with chronic hepatitis B (P>0.05), also between patients with HBV infection with HBV-DNA<1x10(3)copies/mL and those with HBV-DNA> or =1 x 10(3)copies/mL (P>0.05). However, frequencies of TT genotype at position -819T/C and AA genotype at position -592A/C in chronic hepatitis B were significantly higher than that in asymptomatic HBV carriers (P<0.05). CONCLUSION: It is possible that genetic polymorphisms of IL10 promoter region are not associated with both susceptibility of HBV infection and HBV-DNA replication after infected HBV in Chinese Han population. However, the polymorphisms of the promoter region IL10 at position -819T/C and -592A/C are related to inflammatory reaction to liver of the patients with HBV infection.


Subject(s)
Hepatitis B/genetics , Interleukin-10/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Asian People/genetics , China , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Polymerase Chain Reaction
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 22(5): 528-32, 2005 Oct.
Article in Chinese | MEDLINE | ID: mdl-16215941

ABSTRACT

OBJECTIVE: To investigate the polymorphisms of interleukin-18(IL-18) gene promoters, and to disclose whether such polymorphisms are associated with susceptibility to chronic hepatitis B in Chinese Han population. METHODS: Using polymerase chain reaction with sequence specific primers method, the authors detected the single nucleotide polymorphisms of the promoter region of IL-18 gene at position -607C/A and -137G/C in 231 patients with chronic hepatitis B and 300 normal controls. RESULTS: The frequency of CC genotype in IL-18 gene promoter region at position -607 was 0.22(66/300) in normal controls and 0.27(62/231) in patients. The frequency of CA genotype was 0.53(160/300) in normal controls and 0.50(116/231) in patients. The frequency of AA genotype was 0.25(74/300) in normal controls and 0.23(53/231) in patients. The frequencies of -137GG, GC and CC genotype were 0.67, 0.30 and 0.03 in normal controls respectively; whereas in chronic hepatitis B patients the frequencies were 0.79, 0.19 and 0.02. The genotype frequency of -137GG in chronic hepatitis B groups was significantly higher than that in normal controls(chi2: 8.55, P=0.003), but the frequencies of -607C/-137C and -607A/-137C haplotypes in chronic hepatitis B groups were significantly lower than those in normal controls. The association between genotype of IL-18 promoter region polymorphisms and hepatitis B virus(HBV) copies showed that the frequency of -607AA genotype in high HBV-DNA copies groups was lower than that in low HBV-DNA copies groups(chi2: 6.03, P=0.014). CONCLUSION: The polymorphisms of the promoter region of IL-18 gene at position -607C/A and -137G/C are correlated with chronic hepatitis B in Chinese Han population. The people with -137C allele in the promoter region of IL-18 gene may be protected against HBV infection, and the IL-18 -607AA genotype may be linked to HBV-DNA copy.


Subject(s)
Hepatitis B, Chronic/genetics , Interleukin-18/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Child , China , DNA Mutational Analysis , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Polymerase Chain Reaction , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...