Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Cell Rep ; 43(6): 114284, 2024 May 29.
Article En | MEDLINE | ID: mdl-38814785

Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we test a prevailing hypothesis that NE ruptures trigger the pathological cGAS-STING cytosolic DNA-sensing pathway using a mouse model of Lamin cardiomyopathy. The reduction of Lamin A/C in cardio-myocyte of adult mice causes pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures are followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remains inactive. Deleting cGas or Sting does not rescue cardiomyopathy in the mouse model. The lack of cGAS-STING activation is likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling is activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin cardiomyopathy.

2.
Circulation ; 149(15): 1205-1230, 2024 Apr 09.
Article En | MEDLINE | ID: mdl-38189150

BACKGROUND: The relationship between heart failure (HF) and atrial fibrillation (AF) is clear, with up to half of patients with HF progressing to AF. The pathophysiological basis of AF in the context of HF is presumed to result from atrial remodeling. Upregulation of the transcription factor FOG2 (friend of GATA2; encoded by ZFPM2) is observed in human ventricles during HF and causes HF in mice. METHODS: FOG2 expression was assessed in human atria. The effect of adult-specific FOG2 overexpression in the mouse heart was evaluated by whole animal electrophysiology, in vivo organ electrophysiology, cellular electrophysiology, calcium flux, mouse genetic interactions, gene expression, and genomic function, including a novel approach for defining functional transcription factor interactions based on overlapping effects on enhancer noncoding transcription. RESULTS: FOG2 is significantly upregulated in the human atria during HF. Adult cardiomyocyte-specific FOG2 overexpression in mice caused primary spontaneous AF before the development of HF or atrial remodeling. FOG2 overexpression generated arrhythmia substrate and trigger in cardiomyocytes, including calcium cycling defects. We found that FOG2 repressed atrial gene expression promoted by TBX5. FOG2 bound a subset of GATA4 and TBX5 co-bound genomic locations, defining a shared atrial gene regulatory network. FOG2 repressed TBX5-dependent transcription from a subset of co-bound enhancers, including a conserved enhancer at the Atp2a2 locus. Atrial rhythm abnormalities in mice caused by Tbx5 haploinsufficiency were rescued by Zfpm2 haploinsufficiency. CONCLUSIONS: Transcriptional changes in the atria observed in human HF directly antagonize the atrial rhythm gene regulatory network, providing a genomic link between HF and AF risk independent of atrial remodeling.


Atrial Fibrillation , Atrial Remodeling , Heart Failure , Humans , Mice , Animals , Atrial Fibrillation/genetics , Gene Regulatory Networks , Calcium/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Heart Atria , Heart Failure/genetics , Genomics , GATA4 Transcription Factor/genetics
3.
bioRxiv ; 2023 Aug 28.
Article En | MEDLINE | ID: mdl-37693381

Mutations in the nuclear Lamin A/C gene (LMNA) cause diverse degenerative disorders, including malignant dilated cardiomyopathy in adults. A prevailing hypothesis postulates that LMNA mutations cause nuclear envelope ruptures that trigger pathogenic inflammatory signaling via the cGAS-STING cytosolic DNA-sensing pathway. Here, we provide evidence against this hypothesis, using a mouse model of LMNA-related cardiomyopathy that mimics Lamin A/C protein reduction observed in patient cardiomyocytes. We observed that pervasive nuclear envelope ruptures preceded the onset of cardiac transcriptional modulation and dilated cardiomyopathy. Nuclear ruptures activated DNA damage response without causing immediate cardiomyocyte death. However, cGAS-STING downstream cytokine genes remained inactive in the mutant cardiomyocytes. Deleting cGas or Sting did not alleviate cardiomyopathy. Instead, extracellular matrix signaling was predicted to emanate from Lamin A/C-reduced cardiomyocytes to communicate with fibroblasts in the heart. These findings suggest that cGAS-STING is not a major pathogenetic contributor to LMNA-related dilated cardiomyopathy in adult humans.

4.
Nat Commun ; 14(1): 4812, 2023 08 09.
Article En | MEDLINE | ID: mdl-37558654

Branched chain amino acid (BCAA) catabolic impairments have been implicated in several diseases. Branched chain ketoacid dehydrogenase (BCKDH) controls the rate limiting step in BCAA degradation, the activity of which is inhibited by BCKDH kinase (BDK)-mediated phosphorylation. Screening efforts to discover BDK inhibitors led to identification of thiophene PF-07208254, which improved cardiometabolic endpoints in mice. Structure-activity relationship studies led to identification of a thiazole series of BDK inhibitors; however, these inhibitors did not improve metabolism in mice upon chronic administration. While the thiophenes demonstrated sustained branched chain ketoacid (BCKA) lowering and reduced BDK protein levels, the thiazoles increased BCKAs and BDK protein levels. Thiazoles increased BDK proximity to BCKDH-E2, whereas thiophenes reduced BDK proximity to BCKDH-E2, which may promote BDK degradation. Thus, we describe two BDK inhibitor series that possess differing attributes regarding BDK degradation or stabilization and provide a mechanistic understanding of the desirable features of an effective BDK inhibitor.


Amino Acids, Branched-Chain , Thiophenes , Mice , Animals , Amino Acids, Branched-Chain/metabolism , Phosphorylation , Thiophenes/pharmacology , Oxidoreductases/metabolism
5.
J Mol Cell Cardiol ; 169: 28-40, 2022 08.
Article En | MEDLINE | ID: mdl-35533732

A premature truncation of MYBPHL in humans and a loss of Mybphl in mice is associated with dilated cardiomyopathy, atrial and ventricular arrhythmias, and atrial enlargement. MYBPHL encodes myosin binding protein H-like (MyBP-HL). Prior work in mice indirectly identified Mybphl expression in the atria and in small puncta throughout the ventricle. Because of its genetic association with human and mouse cardiac conduction system disease, we evaluated the anatomical localization of MyBP-HL and the consequences of loss of MyBP-HL on conduction system function. Immunofluorescence microscopy of normal adult mouse ventricles identified MyBP-HL-positive ventricular cardiomyocytes that co-localized with the ventricular conduction system marker contactin-2 near the atrioventricular node and in a subset of Purkinje fibers. Mybphl heterozygous ventricles had a marked reduction of MyBP-HL-positive cells compared to controls. Lightsheet microscopy of normal perinatal day 5 mouse hearts showed enrichment of MyBP-HL-positive cells within and immediately adjacent to the contactin-2-positive ventricular conduction system, but this association was not apparent in Mybphl heterozygous hearts. Surface telemetry of Mybphl-null mice revealed atrioventricular block and atrial bigeminy, while intracardiac pacing revealed a shorter atrial relative refractory period and atrial tachycardia. Calcium transient analysis of isolated Mybphl-null atrial cardiomyocytes demonstrated an increased heterogeneity of calcium release and faster rates of calcium release compared to wild type controls. Super-resolution microscopy of Mybphl heterozygous and homozygous null atrial cardiomyocytes showed ryanodine receptor disorganization compared to wild type controls. Abnormal calcium release, shorter atrial refractory period, and atrial dilation seen in Mybphl null, but not wild type control hearts, agree with the observed atrial arrhythmias, bigeminy, and atrial tachycardia, whereas the proximity of MyBP-HL-positive cells with the ventricular conduction system provides insight into how a predominantly atrial expressed gene contributes to ventricular arrhythmias and ventricular dysfunction.


Arrhythmias, Cardiac , Calcium , Cardiac Conduction System Disease , Cytoskeletal Proteins , Animals , Humans , Mice , Arrhythmias, Cardiac/genetics , Calcium/metabolism , Cardiac Conduction System Disease/genetics , Contactins/metabolism , Cytoskeletal Proteins/genetics , Heart Atria/metabolism , Myosins/metabolism , Purkinje Fibers , Tachycardia
6.
Elife ; 92020 08 17.
Article En | MEDLINE | ID: mdl-32804075

Perturbation of addition of second heart field (SHF) cardiac progenitor cells to the poles of the heart tube results in congenital heart defects (CHD). The transcriptional programs and upstream regulatory events operating in different subpopulations of the SHF remain unclear. Here, we profile the transcriptome and chromatin accessibility of anterior and posterior SHF sub-populations at genome-wide levels and demonstrate that Hoxb1 negatively regulates differentiation in the posterior SHF. Spatial mis-expression of Hoxb1 in the anterior SHF results in hypoplastic right ventricle. Activation of Hoxb1 in embryonic stem cells arrests cardiac differentiation, whereas Hoxb1-deficient mouse embryos display premature cardiac differentiation. Moreover, ectopic differentiation in the posterior SHF of embryos lacking both Hoxb1 and its paralog Hoxa1 results in atrioventricular septal defects. Our results show that Hoxb1 plays a key role in patterning cardiac progenitor cells that contribute to both cardiac poles and provide new insights into the pathogenesis of CHD.


Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Stem Cells/metabolism , Transcriptome , Animals , Chromatin/metabolism , Genes, Homeobox , Heart Defects, Congenital/embryology , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic
7.
Circ Res ; 127(2): e28-e43, 2020 07 03.
Article En | MEDLINE | ID: mdl-32347164

RATIONALE: ZO-1 (Zona occludens 1), encoded by the tight junction protein 1 (TJP1) gene, is a regulator of paracellular permeability in epithelia and endothelia. ZO-1 interacts with the actin cytoskeleton, gap, and adherens junction proteins and localizes to intercalated discs in cardiomyocytes. However, the contribution of ZO-1 to cardiac physiology remains poorly defined. OBJECTIVE: We aim to determine the role of ZO-1 in cardiac function. METHODS AND RESULTS: Inducible cardiomyocyte-specific Tjp1 deletion mice (Tjp1fl/fl; Myh6Cre/Esr1*) were generated by crossing the Tjp1 floxed mice and Myh6Cre/Esr1* transgenic mice. Tamoxifen-induced loss of ZO-1 led to atrioventricular (AV) block without changes in heart rate, as measured by ECG and ex vivo optical mapping. Mice with tamoxifen-induced conduction system-specific deletion of Tjp1 (Tjp1fl/fl; Hcn4CreERt2) developed AV block while tamoxifen-induced conduction system deletion of Tjp1 distal to the AV node (Tjp1fl/fl; Kcne1CreERt2) did not demonstrate conduction defects. Western blot and immunostaining analyses of AV nodes showed that ZO-1 loss decreased Cx (connexin) 40 expression and intercalated disc localization. Consistent with the mouse model study, immunohistochemical staining showed that ZO-1 is abundantly expressed in the human AV node and colocalizes with Cx40. Ventricular conduction was not altered despite decreased localization of ZO-1 and Cx43 at the ventricular intercalated disc and modestly decreased left ventricular ejection fraction, suggesting ZO-1 is differentially required for AV node and ventricular conduction. CONCLUSIONS: ZO-1 is a key protein responsible for maintaining appropriate AV node conduction through maintaining gap junction protein localization.


Atrioventricular Node/metabolism , Heart Rate , Zonula Occludens-1 Protein/metabolism , Animals , Atrioventricular Node/physiology , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Potassium Channels, Voltage-Gated/metabolism , Zonula Occludens-1 Protein/genetics , Gap Junction alpha-5 Protein
8.
J Clin Invest ; 129(11): 4937-4950, 2019 11 01.
Article En | MEDLINE | ID: mdl-31609246

Atrial fibrillation (AF), defined by disorganized atrial cardiac rhythm, is the most prevalent cardiac arrhythmia worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with AF risk, including the cardiogenic transcription factor genes TBX5, GATA4, and NKX2-5. We report that Tbx5 and Gata4 interact with opposite signs for atrial rhythm controls compared with cardiac development. Using mouse genetics, we found that AF pathophysiology caused by Tbx5 haploinsufficiency, including atrial arrhythmia susceptibility, prolonged action potential duration, and ectopic cardiomyocyte depolarizations, were all rescued by Gata4 haploinsufficiency. In contrast, Nkx2-5 haploinsufficiency showed no combinatorial effect. The molecular basis of the TBX5/GATA4 interaction included normalization of intra-cardiomyocyte calcium flux and expression of calcium channel genes Atp2a2 and Ryr2. Furthermore, GATA4 and TBX5 showed antagonistic interactions on an Ryr2 enhancer. Atrial rhythm instability caused by Tbx5 haploinsufficiency was rescued by a decreased dose of phospholamban, a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, consistent with a role for decreased sarcoplasmic reticulum calcium flux in Tbx5-dependent AF susceptibility. This work defines a link between Tbx5 dose, sarcoplasmic reticulum calcium flux, and AF propensity. The unexpected interactions between Tbx5 and Gata4 in atrial rhythm control suggest that evaluating specific interactions between genetic risk loci will be necessary for ascertaining personalized risk from genetic association data.


Atrial Fibrillation , Calcium Signaling/genetics , Calcium/metabolism , Genetic Loci , Homeostasis/genetics , Sarcoplasmic Reticulum , Transcription Factors , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Genome-Wide Association Study , Heart Atria/metabolism , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Mice , Risk Factors , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Elife ; 82019 03 21.
Article En | MEDLINE | ID: mdl-30896405

Risk for Atrial Fibrillation (AF), the most common human arrhythmia, has a major genetic component. The T-box transcription factor TBX5 influences human AF risk, and adult-specific Tbx5-mutant mice demonstrate spontaneous AF. We report that TBX5 is critical for cellular Ca2+ homeostasis, providing a molecular mechanism underlying the genetic implication of TBX5 in AF. We show that cardiomyocyte action potential (AP) abnormalities in Tbx5-deficient atrial cardiomyocytes are caused by a decreased sarcoplasmic reticulum (SR) Ca2+ ATPase (SERCA2)-mediated SR calcium uptake which was balanced by enhanced trans-sarcolemmal calcium fluxes (calcium current and sodium/calcium exchanger), providing mechanisms for triggered activity. The AP defects, cardiomyocyte ectopy, and AF caused by TBX5 deficiency were rescued by phospholamban removal, which normalized SERCA function. These results directly link transcriptional control of SERCA2 activity, depressed SR Ca2+ sequestration, enhanced trans-sarcolemmal calcium fluxes, and AF, establishing a mechanism underlying the genetic basis for a Ca2+-dependent pathway for AF risk.


Atrial Fibrillation/physiopathology , Calcium/metabolism , Mutant Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , T-Box Domain Proteins/metabolism , Animals , Cations, Divalent/metabolism , Cells, Cultured , Disease Models, Animal , Mice , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , T-Box Domain Proteins/deficiency
10.
Mech Dev ; 143: 1-8, 2017 02.
Article En | MEDLINE | ID: mdl-27956219

Hox transcription factors play critical roles during early vertebrate development. Previous studies have revealed an overlapping function of Hoxa1 and Hoxb1 during specification of the rhombomeres from which neural crest cells emerge. A recent study on Hoxa1 mutant mice documented its function during cardiovascular development, however, the role of Hoxb1 is still unclear. Here we show using single and compound Hoxa1;Hoxb1 mutant embryos that reduction of Hoxa1 gene dosage in Hoxb1-null genetic background is sufficient to result in abnormal pharyngeal aortic arch (PAA) development and subsequently in great artery defects. Endothelial cells in the 4th PAAs of compound mutant differentiate normally whereas vascular smooth muscle cells of the vessels are absent in the defective PAAs. The importance of Hoxa1 and Hoxb1, and their interaction during specification of cardiac NCCs is demonstrated. Together, our data reveal a critical role for anterior Hox genes during PAA development, providing new mechanistic insights into the etiology of congenital heart defects.


Branchial Region/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Neovascularization, Physiologic/genetics , Transcription Factors/genetics , Animals , Branchial Region/cytology , Branchial Region/embryology , Cell Differentiation , Embryo, Mammalian , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Dosage , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/genetics , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Transcription Factors/deficiency
11.
Dev Biol ; 406(2): 247-58, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26284287

Outflow tract (OFT) anomalies are among the most common congenital heart defects found at birth. The embryonic OFT grows by the progressive addition of cardiac progenitors, termed the second heart field (SHF), which originate from splanchnic pharyngeal mesoderm. Development of the SHF is controlled by multiple intercellular signals and transcription factors; however the relationship between different SHF regulators remains unclear. We have recently shown that Hoxa1 and Hoxb1 are expressed in a sub-population of the SHF contributing to the OFT. Here, we report that Hoxb1 deficiency results in a shorter OFT and ventricular septal defects (VSD). Mechanistically, we show that both FGF/ERK and BMP/SMAD signaling, which regulate proliferation and differentiation of cardiac progenitor cells and OFT morphogenesis, are enhanced in the pharyngeal region in Hoxb1 mutants. Absence of Hoxb1 also perturbed SHF development through premature myocardial differentiation. Hence, the positioning and remodeling of the mutant OFT is disrupted. Hoxa1(-/-) embryos, in contrast, have low percentage of VSD and normal SHF development. However, compound Hoxa1(-/-); Hoxb1(+/-) embryos display OFT defects associated with premature SHF differentiation, demonstrating redundant roles of these factors during OFT development. Our findings provide new insights into the gene regulatory network controlling SHF and OFT formation.


Cell Differentiation/physiology , Cell Proliferation/physiology , Heart/embryology , Homeodomain Proteins/metabolism , Mesoderm/cytology , Morphogenesis/physiology , Transcription Factors/metabolism , DNA Primers/genetics , Embryo, Mammalian , Galactosides , Genotype , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Indoles , Real-Time Polymerase Chain Reaction
12.
Cell Mol Life Sci ; 72(10): 2005-22, 2015 May.
Article En | MEDLINE | ID: mdl-25504289

The regulation of cardiac differentiation is critical for maintaining normal cardiac development and function. The precise mechanisms whereby cardiac differentiation is regulated remain uncertain. Here, we have identified a GATA-4 target, EGF, which is essential for cardiogenesis and regulates cardiac differentiation in a dose- and time-dependent manner. Moreover, EGF demonstrates functional interaction with GATA-4 in inducing the cardiac differentiation of P19CL6 cells in a time- and dose-dependent manner. Biochemically, GATA-4 forms a complex with STAT3 to bind to the EGF promoter in response to EGF stimulation and cooperatively activate the EGF promoter. Functionally, the cooperation during EGF activation results in the subsequent activation of cyclin D1 expression, which partly accounts for the lack of additional induction of cardiac differentiation by the GATA-4/STAT3 complex. Thus, we propose a model in which the regulatory cascade of cardiac differentiation involves GATA-4, EGF, and cyclin D1.


Cell Differentiation/physiology , Epidermal Growth Factor/metabolism , GATA4 Transcription Factor/metabolism , Heart/embryology , Models, Biological , Myocardium/cytology , Signal Transduction/physiology , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Histological Techniques , Immunoprecipitation , Mice , Real-Time Polymerase Chain Reaction , Time Factors
13.
Circ Res ; 115(9): 790-9, 2014 Oct 10.
Article En | MEDLINE | ID: mdl-25190705

RATIONALE: Cardiac progenitor cells from the second heart field (SHF) contribute to rapid growth of the embryonic heart, giving rise to right ventricular and outflow tract (OFT) myocardium at the arterial pole of the heart, and atrial myocardium at the venous pole. Recent clonal analysis and cell-tracing experiments indicate that a common progenitor pool in the posterior region of the SHF gives rise to both OFT and atrial myocytes. The mechanisms regulating deployment of this progenitor pool remain unknown. OBJECTIVE: To evaluate the role of TBX1, the major gene implicated in congenital heart defects in 22q11.2 deletion syndrome patients, in posterior SHF development. METHODS AND RESULTS: Using transcriptome analysis, genetic tracing, and fluorescent dye-labeling experiments, we show that Tbx1-dependent OFT myocardium originates in Hox-expressing cells in the posterior SHF. In Tbx1 null embryos, OFT progenitor cells fail to segregate from this progenitor cell pool, leading to failure to expand the dorsal pericardial wall and altered positioning of the cardiac poles. Unexpectedly, addition of SHF cells to the venous pole of the heart is also impaired, resulting in abnormal development of the dorsal mesenchymal protrusion, and partially penetrant atrioventricular septal defects, including ostium primum defects. CONCLUSIONS: Tbx1 is required for inflow as well as OFT morphogenesis by regulating the segregation and deployment of progenitor cells in the posterior SHF. Our results provide new insights into the pathogenesis of congenital heart defects and 22q11.2 deletion syndrome phenotypes.


Cell Movement , Coronary Vessels/metabolism , DiGeorge Syndrome/metabolism , Heart/embryology , Myocardium/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Animals , Cell Lineage , Cell Proliferation , Coronary Vessels/embryology , Coronary Vessels/pathology , DiGeorge Syndrome/genetics , DiGeorge Syndrome/pathology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Gestational Age , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Myocardium/pathology , Phenotype , Signal Transduction , Stem Cells/pathology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
14.
Methods Mol Biol ; 1196: 37-48, 2014.
Article En | MEDLINE | ID: mdl-25151156

Cell lineage studies have been widely used in developmental biology to establish which cells, and how many cells, in the early embryo will give rise to a specific structure and its derivatives. Several methods have been developed to label progenitor cells in the early embryo. Here, we describe the genetic tracing approach that relies on the use of the recombinase to genetically and permanently label progenitor cells as well as their progeny through specific activation of a conditional reporter gene, the ROSA26 reporter mouse. Labeling of progenitor cells is spatially controlled by the use of a tissue-specific promoter driving Cre, the Hoxb1 (IRES-Cre/+) and the Hoxa1-enhIII-cre. ROSA26R mice and Hoxb1 (IRES-Cre/+) or Hoxa1-enhIII-cremice are crossed together to generate embryos at different stages of development. Embryos are collected and dissected at a specific stage of development and fixed in paraformaldehyde. To follow Hoxb1 (+) and Hoxa1 (+) progeny, X-gal staining is performed to detect ß-galactosidase activity in embryos or developing organ such as the heart. Finally, X-gal-positive cells are observed on whole-mount embryos or dissected organ to determine the lineage contribution of Hoxa1 and Hoxb1 during development.


Gene Expression , Genetic Linkage , Homeodomain Proteins/genetics , Animals , Cell Lineage/genetics , Crosses, Genetic , Embryo, Mammalian/metabolism , Genes, Reporter , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Stem Cells/metabolism
15.
Cardiol Res Pract ; 2012: 180297, 2012.
Article En | MEDLINE | ID: mdl-22701807

Bicuspid aortic valve (BAV) is the most common congenital heart defect, affecting 1-2% of the population. It is generally diagnosed late in adulthood when deterioration of the abnormal leaflet becomes clinically evident. BAV patients have an increased risk of developing serious complications, including stenosis, regurgitation, endocarditis, dilation of the aorta, aortic dissection, and aneurysm. BAV is a heritable trait, but the genetic basis underlying this cardiac malformation remains poorly understood. In the last decade, thanks to studies in animal models as well as genetic and biochemical approaches, a large number of genes that play important roles in heart development have been identified. These discoveries provided valuable insight into cardiac morphogenesis and uncovered congenital-heart-disease-causing genes. This paper will summarize the current knowledge of valve morphogenesis as well as our current understanding of the genetic pathways involved in BAV formation. The impact of these advances on human health including diagnosis of BAV and prevention of cardiovascular complications in individuals with BAV or with a family history of BAV is also discussed.

16.
Dev Biol ; 358(2): 368-78, 2011 Oct 15.
Article En | MEDLINE | ID: mdl-21839733

Members of the GATA family of transcription factors are critical regulators of heart development and mutations in 2 of them, GATA4 and GATA6 are associated with outflow tract and septal defects in human. The heart expresses 3 GATA factors, GATA4, 5 and 6 in a partially overlapping pattern. Here, we report that compound Gata4/Gata5 and Gata5/Gata6 mutants die embryonically or perinatally due to severe congenital heart defects. Almost all Gata4(+/-)Gata5(+/-) mutant embryos have double outlet right ventricles (DORV), large ventricular septal defects (VSD) as well as hypertrophied mitral and tricuspid valves. Only 25% of double compound Gata4/Gata5 heterozygotes survive to adulthood and these mice have aortic stenosis. Compound loss of a Gata5 and a Gata6 allele also leads to DORVs associated with subaortic VSDs. Expression of several transcription factors important for endocardial and myocardial cell differentiation, such as Tbx20, Mef2c, Hey1 and Hand2, was reduced in compound heterozygote embryos. These findings suggest the existence of important genetic interactions between Gata5 and the 2 other cardiac GATA factors in endocardial cushion formation and outflow tract morphogenesis. The data identify GATA5 as a potential genetic modifier of congenital heart disease and provide insight for elucidating the genetic basis of an important class of human birth defects.


Fetal Heart/embryology , Fetal Heart/metabolism , GATA4 Transcription Factor/metabolism , GATA5 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Animals , Aortic Valve Stenosis/embryology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Endocardial Cushion Defects/embryology , Endocardial Cushion Defects/genetics , Endocardial Cushion Defects/metabolism , Female , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA5 Transcription Factor/deficiency , GATA5 Transcription Factor/genetics , GATA6 Transcription Factor/deficiency , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Septal Defects, Ventricular/embryology , Heart Septal Defects, Ventricular/genetics , Heart Septal Defects, Ventricular/metabolism , Heterozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Clin Invest ; 121(7): 2876-87, 2011 Jul.
Article En | MEDLINE | ID: mdl-21633169

Bicuspid aortic valve (BAV), the leading congenital heart disease, occurs in 1%-2% of the population. Genetic studies suggest that BAV is an autosomal-dominant disease with reduced penetrance. However, only 1 gene, NOTCH1, has been linked to cases of BAV. Here, we show that targeted deletion of Gata5 in mice leads to hypoplastic hearts and partially penetrant BAV formation. Endocardial cell-specific inactivation of Gata5 led to BAV, similar to that observed in Gata5-/- mice. In all cases, the observed BAVs resulted from fusion of the right-coronary and noncoronary leaflets, the subtype associated with the more severe valve dysfunction in humans. Neither endocardial cell proliferation nor cushion formation was altered in the absence of Gata5. Rather, defective endocardial cell differentiation, resulting from the deregulation of several components of the Notch pathway and other important endocardial cell regulators, may be the underlying mechanism of disease. The results unravel a critical cell-autonomous role for endocardial Gata5 in aortic valve formation and identify GATA5 as a potential gene responsible for congenital heart disease in humans. Mice with mutated Gata5 alleles represent unique models to dissect the mechanisms underlying degenerative aortic valve disease and to develop much-needed preventive and therapeutic interventions.


Aortic Valve/abnormalities , GATA5 Transcription Factor/metabolism , Heart Defects, Congenital/physiopathology , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , GATA5 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Gene Targeting , Heart/anatomy & histology , Heart/growth & development , Humans , Mice , Mice, Knockout , Morphogenesis/physiology , Myocardium/metabolism , Myocardium/pathology , Receptors, Notch/metabolism , Signal Transduction/physiology
19.
Proc Natl Acad Sci U S A ; 107(45): 19356-61, 2010 Nov 09.
Article En | MEDLINE | ID: mdl-20974940

In humans, septal defects are among the most prevalent congenital heart diseases, but their cellular and molecular origins are not fully understood. We report that transcription factor Tbx5 is present in a subpopulation of endocardial cells and that its deletion therein results in fully penetrant, dose-dependent atrial septal defects in mice. Increased apoptosis of endocardial cells lacking Tbx5, as well as neighboring TBX5-positive myocardial cells of the atrial septum through activation of endocardial NOS (Nos3), is the underlying mechanism of disease. Compound Tbx5 and Nos3 haploinsufficiency in mice worsens the cardiac phenotype. The data identify a pathway for endocardial cell survival and unravel a cell-autonomous role for Tbx5 therein. The finding that Nos3, a gene regulated by many congenital heart disease risk factors including stress and diabetes, interacts genetically with Tbx5 provides a molecular framework to understand gene-environment interaction in the setting of human birth defects.


Atrial Septum/cytology , Endocardium/cytology , GATA4 Transcription Factor/physiology , Heart Diseases/congenital , Nitric Oxide Synthase Type III/physiology , T-Box Domain Proteins/physiology , Animals , Atrial Septum/pathology , Cell Survival , Endocardium/pathology , Haploinsufficiency , Heart Defects, Congenital/etiology , Heart Defects, Congenital/genetics , Mice , Phenotype , T-Box Domain Proteins/analysis
...