Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Nucleic Acids Res ; 50(4): 2270-2286, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35137168

ABSTRACT

Human genetic studies identified a strong association between loss of function mutations in RBFOX2 and hypoplastic left heart syndrome (HLHS). There are currently no Rbfox2 mouse models that recapitulate HLHS. Therefore, it is still unknown how RBFOX2 as an RNA binding protein contributes to heart development. To address this, we conditionally deleted Rbfox2 in embryonic mouse hearts and found profound defects in cardiac chamber and yolk sac vasculature formation. Importantly, our Rbfox2 conditional knockout mouse model recapitulated several molecular and phenotypic features of HLHS. To determine the molecular drivers of these cardiac defects, we performed RNA-sequencing in Rbfox2 mutant hearts and identified dysregulated alternative splicing (AS) networks that affect cell adhesion to extracellular matrix (ECM) mediated by Rho GTPases. We identified two Rho GTPase cycling genes as targets of RBFOX2. Modulating AS of these two genes using antisense oligos led to cell cycle and cell-ECM adhesion defects. Consistently, Rbfox2 mutant hearts displayed cell cycle defects and inability to undergo endocardial-mesenchymal transition, processes dependent on cell-ECM adhesion and that are seen in HLHS. Overall, our work not only revealed that loss of Rbfox2 leads to heart development defects resembling HLHS, but also identified RBFOX2-regulated AS networks that influence cell-ECM communication vital for heart development.


Subject(s)
Alternative Splicing , Heart/embryology , RNA Splicing Factors/metabolism , Animals , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Organogenesis , RNA/metabolism , RNA Splicing Factors/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
2.
Biochem Biophys Res Commun ; 509(2): 384-389, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30594394

ABSTRACT

Alternative splicing (AS) is dysregulated in Type 1 diabetic (T1D) hearts but mechanisms responsible are unclear. Here, we provide evidence that the RNA binding protein (RBP) PTBP1 is modulated in adult T1D hearts contributing to AS changes. We show that a spliced variant of PTBP1 that is highly expressed in normal newborn mouse hearts is aberrantly expressed in adult T1D mouse hearts. Comparing known PTBP1-target datasets to our T1D mouse transcriptome datasets, we discovered a group of genes with PTBP1 binding sites in their pre-mRNAs that are differentially spliced in T1D mouse hearts. We demonstrated that inducible expression of diabetes-induced PTBP1 spliced variant has less repressive splicing function. Notably, PTBP1 regulates AS of some of its targets antagonistically to RBFOX2. In sum, our results indicate that diabetic conditions disrupt developmental regulation of PTBP1 leading to differential AS of PTBP1 target genes. Identification of PTBP1 and PTBP1-regulated RNA networks can provide RNA-based therapies for the treatment of diabetes cardiac complications.


Subject(s)
Alternative Splicing , Diabetes Mellitus, Experimental/genetics , Diabetic Cardiomyopathies/genetics , Gene Expression Regulation, Developmental , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Myocardium/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Animals , CELF1 Protein/genetics , CELF1 Protein/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Gene Expression Profiling , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mice , Mice, Inbred NOD , Myocardium/pathology , Polypyrimidine Tract-Binding Protein/metabolism , Protein Binding , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Signal Transduction , Streptozocin , Transcriptional Activation
3.
Biochem Biophys Res Commun ; 503(4): 3205-3211, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30158053

ABSTRACT

Dysregulated alternative splicing (AS) that contributes to diabetes pathogenesis has been identified, but little is known about the RNA binding proteins (RBPs) involved. We have previously found that the RBP CELF1 is upregulated in the diabetic heart; however, it is unclear if CELF1 contributes to diabetes-induced AS changes. Utilizing genome wide approaches, we identified extensive changes in AS patterns in Type 1 diabetic (T1D) mouse hearts. We discovered that many aberrantly spliced genes in T1D hearts have CELF1 binding sites. CELF1-regulated AS affects key genes within signaling pathways relevant to diabetes pathogenesis. Disruption of CELF1 binding sites impairs AS regulation by CELF1. In sum, our results indicate that CELF1 target RNAs are aberrantly spliced in the T1D heart leading to abnormal gene expression. These discoveries pave the way for targeting RBPs and their RNA networks as novel therapies for cardiac complications of diabetes.


Subject(s)
Alternative Splicing , CELF1 Protein/metabolism , Diabetes Complications/genetics , Diabetes Mellitus, Type 1/genetics , Heart Diseases/genetics , Animals , Diabetes Complications/etiology , Diabetes Complications/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Female , Heart Diseases/etiology , Heart Diseases/metabolism , Male , Mice, Inbred C57BL , Myocardium/metabolism , Protein Binding , RNA/genetics , RNA/metabolism
4.
Am J Respir Cell Mol Biol ; 56(4): 506-520, 2017 04.
Article in English | MEDLINE | ID: mdl-27911568

ABSTRACT

Airway remodeling is resultant of a complex multicellular response associated with a progressive decline of pulmonary function in patients with chronic airway disease. Here, repeated infections with respiratory viruses are linked with airway remodeling through largely unknown mechanisms. Although acute activation of the Toll-like receptor (TLR) 3 pathway by extracellular polyinosinic:polycytidylic acid (poly[I:C]) induces innate signaling through the NF-κB transcription factor in normal human small airway epithelial cells, prolonged (repetitive or tonic) poly(I:C) stimulation produces chronic stress fiber formation, mesenchymal transition, and activation of a fibrotic program. Chronic poly(I:C) stimulation enhanced the expression of core mesenchymal regulators Snail family zinc finger 1, zinc finger E-box binding homeobox, mesenchymal intermediate filaments (vimentin), and extracellular matrix proteins (fibronectin-1), and collagen 1A. This mesenchymal transition was prevented by silencing expression of NF-κB/RelA or administration of a small-molecule inhibitor of the IκB kinase, BMS345541. Acute poly(I:C) exposure in vivo induced profound neutrophilic airway inflammation. When administered repetitively, poly(I:C) resulted in enhanced fibrosis observed by lung micro-computed tomography, second harmonic generation microscopy of optically cleared lung tissue, and by immunohistochemistry. Epithelial flattening, expansion of the epithelial mesenchymal trophic unit, and enhanced Snail family zinc finger 1 and fibronectin 1 expression in airway epithelium were also observed. Repetitive poly(I:C)-induced airway remodeling, fibrosis, and epithelial-mesenchymal transition was inhibited by BMS345541 administration. Based on this novel model of viral inflammation-induced remodeling, we conclude that NF-κB is a major controller of epithelial-mesenchymal transition and pulmonary fibrosis, a finding that has potentially important relevance to airway remodeling produced by repetitive viral infections.


Subject(s)
Airway Remodeling , Epithelial-Mesenchymal Transition , Mesoderm/pathology , NF-kappa B/metabolism , Pneumonia/pathology , Pneumonia/physiopathology , Pulmonary Fibrosis/physiopathology , RNA, Viral/pharmacology , Airway Remodeling/drug effects , Animals , Bronchoalveolar Lavage Fluid , Chronic Disease , Collagen/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung/pathology , Mesoderm/drug effects , Mice, Inbred C57BL , Neutrophils/pathology , Pneumonia/complications , Pneumonia/diagnostic imaging , Poly I-C/pharmacology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism , X-Ray Microtomography
5.
Mediators Inflamm ; 2016: 3762561, 2016.
Article in English | MEDLINE | ID: mdl-27524866

ABSTRACT

Asthma is characterized by reversible airway narrowing, shortness of breath, wheezing, coughing, and other symptoms driven by chronic inflammatory processes, commonly triggered by allergens. In 90% of asthmatics, most of these symptoms can also be triggered by intense physical activities and severely exacerbated by environmental factors. This condition is known as exercise-induced asthma (EIA). Current theories explaining EIA pathogenesis involve osmotic and/or thermal alterations in the airways caused by changes in respiratory airflow during exercise. These changes, along with existing airway inflammatory conditions, are associated with increased cellular levels of reactive oxygen species (ROS) affecting important biomolecules including DNA, although the underlying molecular mechanisms have not been completely elucidated. One of the most abundant oxidative DNA lesions is 8-oxoguanine (8-oxoG), which is repaired by 8-oxoguanine DNA glycosylase 1 (OGG1) during the base excision repair (BER) pathway. Whole-genome expression analyses suggest a cellular response to OGG1-BER, involving genes that may have a role in the pathophysiology of EIA leading to mast cell degranulation, airway hyperresponsiveness, and bronchoconstriction. Accordingly, this review discusses a potential new hypothesis in which OGG1-BER-induced gene expression is associated with EIA symptoms.


Subject(s)
Asthma/metabolism , DNA Glycosylases/metabolism , DNA Repair , Exercise , Guanine/analogs & derivatives , Animals , Bronchoconstriction , DNA/analysis , Guanine/chemistry , Humans , Inflammation , Lipid Peroxidation , Mast Cells/cytology , Mice , Oxidative Stress , Physical Conditioning, Animal , Reactive Oxygen Species/metabolism , Signal Transduction
6.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165620, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31778749

ABSTRACT

Aberrations in the cGMP-PKG-Ca2+ pathway are implicated in cardiovascular complications of diverse etiologies, though involved molecular mechanisms are not understood. We performed RNA-Seq analysis to profile global changes in gene expression and exon splicing in Chagas disease (ChD) murine myocardium. Ingenuity-Pathway-Analysis of transcriptome dataset identified 26 differentially expressed genes associated with increased mobilization and cellular levels of Ca2+ in ChD hearts. Mixture-of-isoforms and Enrichr KEGG pathway analyses of the RNA-Seq datasets from ChD (this study) and diabetic (previous study) murine hearts identified alternative splicing (AS) in eleven genes (Arhgef10, Atp2b1, Atp2a3, Cacna1c, Itpr1, Mef2a, Mef2d, Pde2a, Plcb1, Plcb4, and Ppp1r12a) of the cGMP-PKG-Ca2+ pathway in diseased hearts. AS of these genes was validated by an exon exclusion-inclusion assay. Further, Arhgef10, Atp2b1, Mef2a, Mef2d, Plcb1, and Ppp1r12a genes consisted RBFOX2 (RNA-binding protein) binding-site clusters, determined by analyzing the RBFOX2 CLIP-Seq dataset. H9c2 rat heart cells transfected with Rbfox2 (vs. scrambled) siRNA confirmed that expression of Rbfox2 is essential for proper exon splicing of genes of the cGMP-PKG-Ca2+ pathway. We conclude that changes in gene expression may influence the Ca2+ mobilization pathway in ChD, and AS impacts the genes involved in cGMP/PKG/Ca2+ signaling pathway in ChD and diabetes. Our findings suggest that ChD patients with diabetes may be at increased risk of cardiomyopathy and heart failure and provide novel ways to restore cGMP-PKG regulated signaling networks via correcting splicing patterns of key factors using oligonucleotide-based therapies for the treatment of cardiovascular complications.


Subject(s)
Alternative Splicing/genetics , Calcium/metabolism , Cardiomyopathies/genetics , Cyclic GMP/genetics , RNA Splicing Factors/genetics , RNA Splicing/genetics , Signal Transduction/genetics , Animals , Cell Line , Female , Heart/physiopathology , Heart Failure/genetics , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL