Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
1.
Clin Exp Hypertens ; 44(6): 567-572, 2022 Aug 18.
Article in English | MEDLINE | ID: mdl-35699093

ABSTRACT

OBJECTIVE: To investigate whether endothelial nitric oxide synthase (eNOS) rs1799983, rs2070744, and rs61722009 gene polymorphisms are associated with pulmonary arterial hypertension (PAH) in South Fujian newborns with congenital heart disease (CHD). METHODS: Genotyping for the eNOS rs1799983, rs2070744, and rs61722009 polymorphisms was performed using Sanger sequencing in 50 newborns with PAH secondary to CHD [CHD PAH (+)], 52 newborns with CHD without PAH [CHD PAH (-)], and 60 healthy controls. RESULTS: The genotype and allele frequency distributions of eNOS rs1799983, rs2070744, and rs61722009 were similar between CHD and healthy controls (P > .05). The frequencies of the eNOS rs1799983 G/T allele were 85% and 15% in the CHD PAH (+) group and 96.15% and 3.85% in the CHD PAH (-) group, the frequency of the T allele was higher in the CHD PAH (+) group than in the CHD PAH (-) group(P< .05), and patients with the GT/TT genotypes of eNOS rs1799983 may present higher PAH (OR = 4.412, 95%CI:1.411-13.797, P= .011). Newborns with the GT/TT genotypes had decreased plasma NO production compared to newborns with the GG genotype (P< .01), and NO levels in the CHD PAH (+) group were significantly lower than those in the CHD PAH (-) group (P < .05). CONCLUSION: The T allele could be a risk factor for PAH in newborns with CHD in South Fujian through decreased levels of nitric oxide production by the endothelium.


Subject(s)
Heart Defects, Congenital , Pulmonary Arterial Hypertension , Case-Control Studies , Gene Frequency , Genetic Predisposition to Disease , Genotype , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Humans , Infant, Newborn , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Pulmonary Arterial Hypertension/enzymology , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/pathology
2.
Clin Res Cardiol ; 111(2): 154-162, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33280062

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme (ACE) 2 is known to be a functional receptor for SARS-CoV-2 in the current pandemic. Soluble ACE2 (sACE2) concentrations are elevated in patients with various cardiovascular disorders including heart failure. METHODS: In a total of 182 consecutive adult patients with complex congenital heart disease (CHD) and 63 healthy controls, sACE2 concentrations were measured in serum using the Human ACE2® assay by Cloud-Clone Corporation and associated with clinical, laboratory and echocardiographic parameters. RESULTS: Median sACE2 levels were increased in patients with complex CHD as compared to healthy controls (761.9 pg/ml vs 365.2 pg/ml, p < 0.001). Moreover, sACE2 concentrations were significantly elevated in patients with a higher NYHA class ≥ III (1856.2 pg/ml vs 714.5 pg/ml in patients with NYHA class I/II, p < 0.001). Using linear regression analysis, higher sACE2 levels were associated with a higher NYHA class ≥ III, more severe CHD, a morphological left systemic ventricle, higher creatinine and the use of mineralocorticoid receptor antagonists (MRA) in the univariable model. The use of ACE inhibitors or angiotensin receptor blockers (ARB) was associated with lower sACE2 levels. In the multivariable model, higher sACE2 levels were independently associated with a higher NYHA class ≥ III (p = 0.002) and lower sACE2 levels with the use of ACE inhibitors or ARB (p = 0.001). CONCLUSION: Soluble ACE2 concentrations were significantly increased in all types of complex CHD with highest levels found in patients with NYHA class ≥ III. Moreover, a higher NYHA class ≥ III was the most significant determinant that was independently associated with elevated sACE2 concentrations.


Subject(s)
Angiotensin-Converting Enzyme 2/blood , Heart Defects, Congenital/enzymology , Receptors, Virus/blood , Survivors , Adult , Biomarkers/blood , COVID-19/enzymology , COVID-19/virology , Case-Control Studies , Female , Heart Defects, Congenital/blood , Heart Defects, Congenital/diagnosis , Humans , Male , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Up-Regulation , Virus Internalization , Young Adult
3.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33645542

ABSTRACT

Congenital heart disease is the most common type of birth defect, accounting for one-third of all congenital anomalies. Using whole-exome sequencing of 2718 patients with congenital heart disease and a search in GeneMatcher, we identified 30 patients from 21 unrelated families of different ancestries with biallelic phospholipase D1 (PLD1) variants who presented predominantly with congenital cardiac valve defects. We also associated recessive PLD1 variants with isolated neonatal cardiomyopathy. Furthermore, we established that p.I668F is a founder variant among Ashkenazi Jews (allele frequency of ~2%) and describe the phenotypic spectrum of PLD1-associated congenital heart defects. PLD1 missense variants were overrepresented in regions of the protein critical for catalytic activity, and, correspondingly, we observed a strong reduction in enzymatic activity for most of the mutant proteins in an enzymatic assay. Finally, we demonstrate that PLD1 inhibition decreased endothelial-mesenchymal transition, an established pivotal early step in valvulogenesis. In conclusion, our study provides a more detailed understanding of disease mechanisms and phenotypic expression associated with PLD1 loss of function.


Subject(s)
Alleles , Heart Defects, Congenital , Heart Valve Diseases , Loss of Function Mutation , Phospholipase D , Female , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Heart Valve Diseases/enzymology , Heart Valve Diseases/genetics , Humans , Male , Phospholipase D/genetics , Phospholipase D/metabolism
4.
Clin Sci (Lond) ; 132(23): 2483-2491, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30322850

ABSTRACT

Congenital heart disease (CHD) is one of the most common birth defects in humans, but its genetic etiology remains largely unknown despite decades of research. The Notch signaling pathway plays critical roles in embryonic cardiogenesis. Mind bomb 1 (Mib1) is a vital protein that activates the Notch signaling pathway through promoting ubiquitination, endocytosis and subsequent activation of Notch ligands. Previous studies show that Mib1 knockout in mice completely abolishes Notch signaling, leading to cardiac deformity. However, the function of MIB1 and its potential disease-causing mutations are poorly studied in human CHD. In this research, we identified four novel non-synonymous heterozygous rare mutations of MIB1 from 417 Han Chinese CHD patients. The following biochemical analyses revealed that mutations p.T312K fs*55 and p.W271G significantly deplete MIB1's function, resulting in a lower level of JAGGED1 (JAG1) ubiquitination and Notch signaling induction. Our results suggest that pathologic variants in MIB1 may contribute to CHD occurrence, shedding new light on the genetic mechanism of CHD in the context of the Notch signaling pathway.


Subject(s)
Heart Defects, Congenital/genetics , Mutation, Missense , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Animals, Genetically Modified , Asian People/genetics , Case-Control Studies , Child , Child, Preschool , China/epidemiology , Female , Genetic Predisposition to Disease , HEK293 Cells , Heart Defects, Congenital/blood , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/ethnology , Humans , Infant , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Phenotype , Receptors, Notch/genetics , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/genetics , Zebrafish/metabolism
5.
Circ Heart Fail ; 11(9): e004571, 2018 09.
Article in English | MEDLINE | ID: mdl-30354365

ABSTRACT

Background Single ventricle (SV) congenital heart disease is fatal without intervention, and eventual heart failure is a major cause of morbidity and mortality. Although there are no proven medical therapies for the treatment or prevention of heart failure in the SV heart disease population, phosphodiesterase-5 inhibitors (PDE5i), such as sildenafil, are increasingly used. Although the pulmonary vasculature is the primary target of PDE5i therapy in patients with SV heart disease, the effects of PDE5i on the SV heart disease myocardium remain largely unknown. We sought to determine PDE5 expression and activity in the single right ventricle of SV heart disease patients relative to nonfailing controls and to determine whether PDE5 impacts cardiomyocyte remodeling using a novel serum-based in vitro model. Methods and Results PDE5 expression (n=9 nonfailing; n=7 SV heart disease), activity (n=8 nonfailing; n=9 SV heart disease), and localization (n=3 SV heart disease) were determined in explanted human right ventricle myocardium. PDE5 is expressed in SV heart disease cardiomyocytes, and PDE5 protein expression and activity are increased in SV heart disease right ventricle compared with nonfailing right ventricle. Isolated neonatal rat ventricular myocytes were treated for 72 hours with nonfailing or SV heart disease patient serum±sildenafil. Reverse transcription quantitative polymerase chain reaction (n=5 nonfailing; n=12 SV heart disease) and RNA sequencing (n=3 nonfailing; n=3 SV heart disease) were performed on serum-treated neonatal rat ventricular myocytes and demonstrated that treatment with SV heart disease sera results in pathological gene expression changes that are attenuated with PDE5i. Conclusions PDE5 is increased in failing SV heart disease myocardium, and pathological gene expression changes in SV heart disease serum-treated neonatal rat ventricular myocytes are abrogated by PDE5i. These results suggest that PDE5 represents an intriguing myocardial therapeutic target in this population.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Heart Defects, Congenital/enzymology , Heart Failure/enzymology , Heart Ventricles/enzymology , Myocytes, Cardiac/enzymology , Ventricular Function, Right , Ventricular Remodeling , Animals , Animals, Newborn , Case-Control Studies , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Heart Defects, Congenital/physiopathology , Heart Failure/physiopathology , Heart Ventricles/abnormalities , Heart Ventricles/physiopathology , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphodiesterase 5 Inhibitors/pharmacology , Rats, Sprague-Dawley , Up-Regulation
6.
J Biol Regul Homeost Agents ; 32(5): 1255-1260, 2018.
Article in English | MEDLINE | ID: mdl-30334422

ABSTRACT

This article is to investigate the association between C677T polymorphism of 5, 10-methylenetetrahydrofolate (MTHFR) gene and congenital heart defects (CHD). Two hundred thirty-five nuclear families (father, mother and child) with CHD were enrolled in the study (experimental group), and two hundred thirty-five healthy nuclear families were selected as a control group. Under the case-control study, the C677T polymorphism of MTHFR gene was detected with polymerase chain reaction-restriction fragment length polymorphism and DNA sequencing. The distribution of genotype frequency in the CHD group and control group were analyzed. SPSS 13.0 software was used to analyze the data. The distribution of genotype frequency at C677T polymorphism site was significantly different between the CHD group (including ventricular septal defect, atrial septal defect, tetralogy of fallot, double outlet right ventricle, patent ductus arteriosus) (child and mother) and healthy control group (child and mother). There were no differences between CHD group-fathers and healthy control group-fathers. Analyses of the MTHFR genotypes of CHD nuclear family data with transmitted disequilibrium test (TDT) and haplotype-based haplotype relative risk statistical method both revealed significant indications that the parents transmitted more T allele of MTHFR to their CHD children. TT genotype of MTHFR gene is associated with CHD, and a mother or a child with T allele has a much higher risk of CHD.


Subject(s)
Genetic Predisposition to Disease , Heart Defects, Congenital/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic , Tetrahydrofolates/metabolism , Adult , Alleles , Case-Control Studies , Child , Female , Gene Frequency , Genotype , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/metabolism , Humans , Male
7.
Cardiovasc Res ; 114(6): 830-845, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29409032

ABSTRACT

Aims: Phosphodiesterase 2 A (Pde2A), a cAMP-hydrolysing enzyme, is essential for mouse development; however, the cause of Pde2A knockout embryonic lethality is unknown. To understand whether Pde2A plays a role in cardiac development, hearts of Pde2A deficient embryos were analysed at different stage of development. Methods and results: At the stage of four chambers, Pde2A deficient hearts were enlarged compared to the hearts of Pde2A heterozygous and wild-type. Pde2A knockout embryos revealed cardiac defects such as absence of atrial trabeculation, interventricular septum (IVS) defects, hypertrabeculation and thinning of the myocardial wall and in rare cases they had overriding aorta and valves defects. E14.5 Pde2A knockouts showed reduced cardiomyocyte proliferation and increased apoptosis in the IVS and increased proliferation in the ventricular trabeculae. Analyses of E9.5 Pde2A knockout embryos revealed defects in cardiac progenitor and neural crest markers, increase of Islet1 positive and AP2 positive apoptotic cells. The expression of early cTnI and late Mef2c cardiomyocyte differentiation markers was strongly reduced in Pde2A knockout hearts. The master transcription factors of cardiac development, Tbx, were down-regulated in E14.5 Pde2A knockout hearts. Absence of Pde2A caused an increase of intracellular cAMP level, followed by an up-regulation of the inducible cAMP early repressor, Icer in fetal hearts. In vitro experiments on wild-type fetal cardiomyocytes showed that Tbx gene expression is down-regulated by cAMP inducers. Furthermore, Pde2A inhibition in vivo recapitulated the heart defects observed in Pde2A knockout embryos, affecting cardiac progenitor cells. Interestingly, the expression of Pde2A itself was dramatically affected by Pde2A inhibition, suggesting a potential autoregulatory loop. Conclusions: We demonstrated for the first time a direct relationship between Pde2A impairment and the onset of mouse congenital heart defects, highlighting a novel role for cAMP in cardiac development regulation.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/deficiency , Fetal Heart/enzymology , Heart Defects, Congenital/enzymology , Myocytes, Cardiac/enzymology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Fetal Heart/abnormalities , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Gestational Age , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Myocytes, Cardiac/pathology , Phenotype , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Troponin I/genetics , Troponin I/metabolism
8.
Circ Cardiovasc Genet ; 10(5)2017 10.
Article in English | MEDLINE | ID: mdl-28986453

ABSTRACT

BACKGROUND: Hypoxia is often associated with cardiopulmonary diseases, which represent some of the leading causes of mortality worldwide. Long-term hypoxia exposures, whether from disease or environmental condition, can cause cardiomyopathy and lead to heart failure. Indeed, hypoxia-induced heart failure is a hallmark feature of chronic mountain sickness in maladapted populations living at high altitude. In a previously established Drosophila heart model for long-term hypoxia exposure, we found that hypoxia caused heart dysfunction. Calcineurin is known to be critical in cardiac hypertrophy under normoxia, but its role in the heart under hypoxia is poorly understood. METHODS AND RESULTS: In the present study, we explore the function of calcineurin, a gene candidate we found downregulated in the Drosophila heart after lifetime and multigenerational hypoxia exposure. We examined the roles of 2 homologs of Calcineurin A, CanA14F, and Pp2B in the Drosophila cardiac response to long-term hypoxia. We found that knockdown of these calcineurin catalytic subunits caused cardiac restriction under normoxia that are further aggravated under hypoxia. Conversely, cardiac overexpression of Pp2B under hypoxia was lethal, suggesting that a hypertrophic signal in the presence of insufficient oxygen supply is deleterious. CONCLUSIONS: Our results suggest a key role for calcineurin in cardiac remodeling during long-term hypoxia with implications for diseases of chronic hypoxia, and it likely contributes to mechanisms underlying these disease states.


Subject(s)
Calcineurin/biosynthesis , Down-Regulation , Drosophila Proteins/biosynthesis , Gene Expression Regulation, Enzymologic , Heart Defects, Congenital/enzymology , Hypoxia/enzymology , Mutation, Missense , Myocardium/enzymology , Amino Acid Substitution , Animals , Calcineurin/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Hypoxia/genetics , Hypoxia/pathology , Myocardium/pathology
9.
Hum Mol Genet ; 26(23): 4715-4727, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973166

ABSTRACT

Germline mutations in BRAF are a major cause of cardio-facio-cutaneous (CFC) syndrome, which is characterized by heart defects, characteristic craniofacial dysmorphology and dermatologic abnormalities. Patients with CFC syndrome also commonly show gastrointestinal dysfunction, including feeding and swallowing difficulties and gastroesophageal reflux. We have previously found that knock-in mice expressing a Braf Q241R mutation exhibit CFC syndrome-related phenotypes, such as growth retardation, craniofacial dysmorphisms, congenital heart defects and learning deficits. However, it remains unclear whether BrafQ241R/+ mice exhibit gastrointestinal dysfunction. Here, we report that BrafQ241R/+ mice have neonatal feeding difficulties and esophageal dilation. The esophagus tissues from BrafQ241R/+ mice displayed incomplete replacement of smooth muscle with skeletal muscle and decreased contraction. Furthermore, the BrafQ241R/+ mice showed hyperkeratosis and a thickened muscle layer in the forestomach. Treatment with MEK inhibitors ameliorated the growth retardation, esophageal dilation, hyperkeratosis and thickened muscle layer in the forestomach in BrafQ241R/+ mice. The esophageal dilation with aberrant skeletal-smooth muscle boundary in BrafQ241R/+ mice were recovered after treatment with the histone H3K27 demethylase inhibitor GSK-J4. Our results provide clues to elucidate the pathogenesis and possible treatment of gastrointestinal dysfunction and failure to thrive in patients with CFC syndrome.


Subject(s)
Ectodermal Dysplasia/enzymology , Esophageal Stenosis/enzymology , Failure to Thrive/enzymology , Focal Epithelial Hyperplasia/enzymology , Heart Defects, Congenital/enzymology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Stomach Diseases/enzymology , Animals , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Esophageal Stenosis/genetics , Esophageal Stenosis/pathology , Facies , Failure to Thrive/genetics , Failure to Thrive/pathology , Female , Focal Epithelial Hyperplasia/genetics , Germ-Line Mutation , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Stomach Diseases/genetics
10.
J Am Heart Assoc ; 6(11)2017 Oct 27.
Article in English | MEDLINE | ID: mdl-29079565

ABSTRACT

BACKGROUND: Congenital heart defects are the most common birth defects worldwide. Although defective Notch signaling is the major cause of mouse embryonic death from cardiovascular defects, how Notch signaling is regulated during embryonic vasculogenesis and heart development is poorly understood. METHODS AND RESULTS: Regulator of G protein signaling 6 (RGS6)-/-/Ca2+/calmodulin-dependent protein kinase II (CaMKII)VV double mutant mice were developed by crossing RGS6-/- mice with mice expressing an oxidation-resistant CaMKIIδ (CaMKIIVV), and the resulting embryonic defects/lethality were investigated using E7.5 to E15.5 embryos. While loss of either RGS6 or oxidized CaMKIIδ does not alter embryogenesis, their combined loss causes defective Notch signaling, severe cardiovascular defects, and embryonic lethality (≈E10.5-11.5). Embryos lacking RGS6 and expressing oxidation-resistant CaMKIIδ exhibit reduced myocardial wall thickness, abnormal trabeculation, and arterial specification defects. Double mutants show vascular remodeling defects, including reduced neurovascularization, delayed neural tube maturation, and small dorsal aortae. These striking cardiovascular defects were accompanied by placental and yolk sac defects in angiogenesis, hematopoiesis, and vascular remodeling similar to what is seen with defective Notch1 signaling. Double mutant hearts, embryos, and yolk sacs exhibit profound downregulation of Notch1, Jagged 1, and Notch downstream target genes Hey1, Hey2, and Hey1L as well as impaired Notch1 signaling in embryos/hearts. CONCLUSIONS: RGS6 and oxidized CaMKIIδ together function as novel critical upstream modulators of Notch signaling required for normal cardiovascular development and embryo survival. Their combined need indicates that they function in parallel pathways needed for Notch1 signaling in yolk sac, placenta and embryos. Thus, dysregulated embryonic RGS6 expression and oxidative activation of CaMKII may potentially contribute to congenital heart defects.


Subject(s)
Blood Vessels/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Defects, Congenital/enzymology , Heart , RGS Proteins/metabolism , Receptors, Notch/metabolism , Animals , Blood Vessels/abnormalities , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Differentiation , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Genotype , Gestational Age , Heart/embryology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Oxidation-Reduction , Phenotype , RGS Proteins/deficiency , RGS Proteins/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Vascular Remodeling
11.
Cardiovasc Res ; 113(10): 1186-1197, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28899000

ABSTRACT

AIMS: RHOA-ROCK signalling regulates cell migration, proliferation, differentiation, and transcription. RHOA is expressed in the developing cardiac conduction system in chicken and mice. In early development, the entire sinus venosus myocardium, including both the transient left-sided and the definitive sinoatrial node (SAN), has pacemaker potential. Later, pacemaker potential is restricted to the right-sided SAN. Disruption of RHOA expression in adult mice causes arrhythmias including bradycardia and atrial fibrillation, the mechanism of which is unknown but presumed to affect the SAN. The aim of this study is to assess the role of RHOA-ROCK signalling in SAN development in the chicken heart. METHODS AND RESULTS: ROCK signalling was inhibited chemically in embryonic chicken hearts using Y-27632. This prolonged the immature state of the sinus venosus myocardium, evidenced by up-regulation of the transcription factor ISL1, wide distribution of pacemaker potential, and significantly reduced heart rate. Furthermore ROCK inhibition caused aberrant expression of typical SAN genes: ROCK1, ROCK2, SHOX2, TBX3, TBX5, ISL1, HCN4, CX40, CAV3.1, and NKX2.5 and left-right asymmetry genes: PITX2C and NODAL. Anatomical abnormalities in pulmonary vein development were also observed. Patch clamp electrophysiology confirmed the immature phenotype of the SAN cells and a residual left-sided sinus venosus myocardium pacemaker-like potential. CONCLUSIONS: RHOA-ROCK signalling is involved in establishing the right-sided SAN as the definitive pacemaker of the heart and restricts typical pacemaker gene expression to the right side of the sinus venosus myocardium.


Subject(s)
Biological Clocks , Cell Differentiation , Signal Transduction , Sinoatrial Node/enzymology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Action Potentials , Animals , Arrhythmias, Cardiac/enzymology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Biological Clocks/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Chick Embryo , Gene Expression Regulation, Developmental , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Heart Rate , Morphogenesis , Myocytes, Cardiac/enzymology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sinoatrial Node/drug effects , Sinoatrial Node/embryology , Sinoatrial Node/physiopathology , Time Factors , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
12.
Pediatr Res ; 82(4): 642-649, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28549058

ABSTRACT

BackgroundHistone deacetylase (HDAC) inhibitors are promising therapeutics for various forms of cardiac diseases. The purpose of this study was to assess cardiac HDAC catalytic activity and expression in children with single ventricle (SV) heart disease of right ventricular morphology, as well as in a rodent model of right ventricular hypertrophy (RVH).MethodsHomogenates of right ventricle (RV) explants from non-failing controls and children born with a SV were assayed for HDAC catalytic activity and HDAC isoform expression. Postnatal 1-day-old rat pups were placed in hypoxic conditions, and echocardiographic analysis, gene expression, HDAC catalytic activity, and isoform expression studies of the RV were performed.ResultsClass I, IIa, and IIb HDAC catalytic activity and protein expression were elevated in the hearts of children born with a SV. Hypoxic neonatal rats demonstrated RVH, abnormal gene expression, elevated class I and class IIb HDAC catalytic activity, and protein expression in the RV compared with those in the control.ConclusionsThese data suggest that myocardial HDAC adaptations occur in the SV heart and could represent a novel therapeutic target. Although further characterization of the hypoxic neonatal rat is needed, this animal model may be suitable for preclinical investigations of pediatric RV disease and could serve as a useful model for future mechanistic studies.


Subject(s)
Heart Defects, Congenital/enzymology , Heart Ventricles/enzymology , Histone Deacetylases/metabolism , Hypertrophy, Right Ventricular/enzymology , Ventricular Function, Right , Ventricular Remodeling , Adaptation, Physiological , Adolescent , Animals , Animals, Newborn , Case-Control Studies , Child , Female , Gene Expression Regulation, Enzymologic , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Heart Ventricles/abnormalities , Heart Ventricles/physiopathology , Histone Deacetylases/genetics , Humans , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Infant , Isoenzymes , Male , Rats, Sprague-Dawley , Signal Transduction
13.
Circ Cardiovasc Genet ; 10(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-28196902

ABSTRACT

BACKGROUND: Hyaluronan (HA) is required for endothelial-to-mesenchymal transition and normal heart development in the mouse. Heart abnormalities in hyaluronidase 2 (HYAL2)-deficient (Hyal2-/- ) mice and humans suggested removal of HA is also important for normal heart development. We have performed longitudinal studies of heart structure and function in Hyal2-/- mice to determine when, and how, HYAL2 deficiency leads to these abnormalities. METHODS AND RESULTS: Echocardiography revealed atrial enlargement, atrial tissue masses, and valvular thickening at 4 weeks of age, as well as diastolic dysfunction that progressed with age, in Hyal2-/- mice. These abnormalities were associated with increased HA, vimentin-positive cells, and fibrosis in Hyal2-/- compared with control mice. Based on the severity of heart dysfunction, acute and chronic groups of Hyal2-/- mice that died at an average of 12 and 25 weeks respectively, were defined. Increased HA levels and mesenchymal cells, but not vascular endothelial growth factor in Hyal2-/- embryonic hearts, suggest that HYAL2 is important to inhibit endothelial-to-mesenchymal transition. Consistent with this, in wild-type embryos, HYAL2 and HA were readily detected, and HA levels decreased with age. CONCLUSIONS: These data demonstrate that disruption of normal HA catabolism in Hyal2-/- mice causes increased HA, which may promote endothelial-to-mesenchymal transition and proliferation of mesenchymal cells. Excess endothelial-to-mesenchymal transition, resulting in increased mesenchymal cells, is the likely cause of morphological heart abnormalities in both humans and mice. In mice, these abnormalities result in progressive and severe diastolic dysfunction, culminating in heart failure.


Subject(s)
Heart Defects, Congenital/enzymology , Heart Failure/enzymology , Hyaluronoglucosaminidase/deficiency , Mesenchymal Stem Cells/enzymology , Ventricular Dysfunction, Left/enzymology , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Proliferation , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition , Fibrosis , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , Genetic Predisposition to Disease , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Heart Valve Diseases/enzymology , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Heart Valve Diseases/physiopathology , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/genetics , Mesenchymal Stem Cells/pathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Phenotype , Stroke Volume , Time Factors , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
14.
Perfusion ; 32(4): 306-312, 2017 May.
Article in English | MEDLINE | ID: mdl-27913766

ABSTRACT

BACKGROUND: Myocardial tolerance to ischemia is influenced by age and preoperative cyanosis through unknown mechanisms and significantly affects postoperative outcomes. Cytochrome c oxidase (CcOx), the terminal enzyme of the mitochondrial electron transport chain, may play a role in the susceptibility to ischemic-reperfusion (IR) injury. Our study aimed at investigating changes in human myocardial CcOx activity based on age and preoperative oxygen saturation to understand its role in transition from neonatal to mature myocardium and hypoxic conditions. METHODS: The right atrial appendage from patients undergoing first time surgical repair/palliation of congenital heart defects was analyzed for steady state CcOx activity by oxidation of ferrocytochrome c via spectrophotometry and steady state CcOx subunit I protein content by protein immunoblotting. Student's t-test compared CcOx activity and protein levels between patients with preoperative hypoxia and normoxia. Multiple linear regression analysis was used to assess the effects of age and preoperative arterial oxygen saturations (SaO2) on CcOx protein activity and protein content. RESULTS: Thirty-two patients with a median (interquartile range) age of 83 days (8-174) and preoperative oxygen saturation 98% (85-100%) were enrolled. Independent of age, preoperative SaO2 ⩽90% was associated with significantly greater CcOx steady state activity (p=0.004). Additionally, older age itself was associated with increased CcOx steady state activity (p=0.022); the combination of preoperative SaO2 and age account for 33% of the variation in CcOx steady state activity (R2=0.332). There was no increase in the CcOx subunit I protein content with either age or preoperative hypoxia. CONCLUSIONS: In patients with congenital heart disease, an increase in CcOx steady state activity is seen with increasing age. Hypoxia leads to upregulation of CcOx steady state activity without an increase in the amount of enzyme protein itself. Higher CcOx activity in older and cyanotic patients may indicate CcOx-dependent reactive oxygen species as the mechanism for IR injury.


Subject(s)
Electron Transport Complex IV/metabolism , Heart Defects, Congenital/enzymology , Hypoxia/enzymology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Humans , Middle Aged , Young Adult
15.
Aging Cell ; 15(5): 853-60, 2016 10.
Article in English | MEDLINE | ID: mdl-27242197

ABSTRACT

Fibrotic aortic valve disease (FAVD) is an important cause of aortic stenosis, yet currently there is no effective treatment for FAVD due to its unknown etiology. The purpose of this study was to investigate whether deficiency in the anti-aging Klotho gene (KL) promotes high-fat-diet-induced FAVD and to explore the underlying molecular mechanism. Heterozygous Klotho-deficient (KL(+/-) ) mice and WT littermates were fed with a high-fat diet (HFD) or normal diet for 13 weeks, followed by treatment with the AMPKα activator (AICAR) for an additional 2 weeks. A HFD caused a greater increase in collagen levels in the aortic valves of KL(+/-) mice than of WT mice, indicating that Klotho deficiency promotes HFD-induced aortic valve fibrosis (AVF). AMPKα activity (pAMPKα) was decreased, while protein expression of collagen I and RUNX2 was increased in the aortic valves of KL(+/-) mice fed with a HFD. Treatment with AICAR markedly attenuated HFD-induced AVF in KL(+/-) mice. AICAR not only abolished the downregulation of pAMPKα but also eliminated the upregulation of collagen I and RUNX2 in the aortic valves of KL(+/-) mice fed with HFD. In cultured porcine aortic valve interstitial cells, Klotho-deficient serum plus cholesterol increased RUNX2 and collagen I protein expression, which were attenuated by activation of AMPKα by AICAR. Interestingly, silencing of RUNX2 abolished the stimulatory effect of Klotho deficiency on cholesterol-induced upregulation of matrix proteins, including collagen I and osteocalcin. In conclusion, Klotho gene deficiency promotes HFD-induced fibrosis in aortic valves, likely through the AMPKα-RUNX2 pathway.


Subject(s)
Adenylate Kinase/metabolism , Aging/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Glucuronidase/deficiency , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Animals , Aortic Valve/enzymology , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Cholesterol/pharmacology , Collagen Type I/metabolism , Diet, High-Fat , Down-Regulation/drug effects , Fibrosis , Gene Knockdown Techniques , Glucuronidase/genetics , Heart Defects, Congenital/enzymology , Heart Valve Diseases/enzymology , Klotho Proteins , Mice , Osteocalcin/metabolism , Sus scrofa , Up-Regulation/drug effects
16.
Annu Rev Biochem ; 85: 485-514, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145839

ABSTRACT

Radical S-adenosylmethionine (SAM) enzymes catalyze an astonishing array of complex and chemically challenging reactions across all domains of life. Of approximately 114,000 of these enzymes, 8 are known to be present in humans: MOCS1, molybdenum cofactor biosynthesis; LIAS, lipoic acid biosynthesis; CDK5RAP1, 2-methylthio-N(6)-isopentenyladenosine biosynthesis; CDKAL1, methylthio-N(6)-threonylcarbamoyladenosine biosynthesis; TYW1, wybutosine biosynthesis; ELP3, 5-methoxycarbonylmethyl uridine; and RSAD1 and viperin, both of unknown function. Aberrations in the genes encoding these proteins result in a variety of diseases. In this review, we summarize the biochemical characterization of these 8 radical S-adenosylmethionine enzymes and, in the context of human health, describe the deleterious effects that result from such genetic mutations.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Heart Defects, Congenital/genetics , Metal Metabolism, Inborn Errors/genetics , Mutation , Neurodegenerative Diseases/genetics , S-Adenosylmethionine/metabolism , Carbon-Carbon Lyases , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Gene Expression , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/pathology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Metal Metabolism, Inborn Errors/enzymology , Metal Metabolism, Inborn Errors/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Proteins/metabolism , Thioctic Acid/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
17.
Am J Physiol Heart Circ Physiol ; 311(1): H137-45, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27199125

ABSTRACT

Associated abnormalities of the lymphatic circulation are well described in congenital heart disease. However, their mechanisms remain poorly elucidated. Using a clinically relevant ovine model of a congenital cardiac defect with chronically increased pulmonary blood flow (shunt), we previously demonstrated that exposure to chronically elevated pulmonary lymph flow is associated with: 1) decreased bioavailable nitric oxide (NO) in pulmonary lymph; and 2) attenuated endothelium-dependent relaxation of thoracic duct rings, suggesting disrupted lymphatic endothelial NO signaling in shunt lambs. To further elucidate the mechanisms responsible for this altered NO signaling, primary lymphatic endothelial cells (LECs) were isolated from the efferent lymphatic of the caudal mediastinal node in 4-wk-old control and shunt lambs. We found that shunt LECs (n = 3) had decreased bioavailable NO and decreased endothelial nitric oxide synthase (eNOS) mRNA and protein expression compared with control LECs (n = 3). eNOS activity was also low in shunt LECs, but, interestingly, inducible nitric oxide synthase (iNOS) expression and activity were increased in shunt LECs, as were total cellular nitration, including eNOS-specific nitration, and accumulation of reactive oxygen species (ROS). Pharmacological inhibition of iNOS reduced ROS in shunt LECs to levels measured in control LECs. These data support the conclusion that NOS signaling is disrupted in the lymphatic endothelium of lambs exposed to chronically increased pulmonary blood and lymph flow and may contribute to decreased pulmonary lymphatic bioavailable NO.


Subject(s)
Endothelial Cells/enzymology , Heart Defects, Congenital/enzymology , Lymph/metabolism , Lymphatic Diseases/enzymology , Lymphatic Vessels/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Down-Regulation , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Heart Defects, Congenital/complications , Heart Defects, Congenital/physiopathology , Lymphatic Diseases/etiology , Lymphatic Diseases/physiopathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/physiopathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Pulmonary Circulation , Reactive Oxygen Species/metabolism , Sheep , Signal Transduction , Stress, Mechanical
18.
PLoS One ; 11(3): e0151140, 2016.
Article in English | MEDLINE | ID: mdl-26990189

ABSTRACT

BACKGROUND: High resolution melting (HRM) is a simple, flexible and low-cost mutation screening technique. The methylenetetrahydrofolate reductase (MTHFR) gene encoding a critical enzyme, potentially affects susceptibility to some congenital defects like congenital heart disease (CHD). We evaluate the performance of HRM for genotyping of the MTHFR gene C677T locus in CHD cases and healthy controls of Chinese Han population. METHODS: A total of 315 blood samples from 147 CHD patients (male72, female 75) and 168 healthy controls (male 92, female 76) were enrolled in the study. HRM was utilized to genotype MTHFR C677T locus of all the samples. The results were compared to that of PCR-RFLP and Sanger sequencing. The association of the MTHFR C677T genotypes and the risk of CHD was analyzed using odds ratio with their 95% confidence interval (CIs) from unconditional logistic regression. RESULTS: All the samples were successfully genotyped by HRM within 1 hour and 30 minutes while at least 6 hours were needed for PCR-RFLP and sequencing. The genotypes of MTHFR C677T CC, CT, and TT were 9.52%, 49.66%, and 40.82% in CHD group but 29.17%, 50% and 20.83% in control group, which were identical using both methods of HRM and PCR-RFLP, demonstrating the sensitivity and specificity of HRM were all 100%. CONCLUSION: MTHFR C677T is a potential risk factor for CHD in our local residents of Shandong province in China. HRM is a fast, sensitive, specific and reliable method for clinical application of genotyping.


Subject(s)
Genotype , Heart Defects, Congenital/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Restriction Fragment Length , Child, Preschool , Female , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/epidemiology , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Polymerase Chain Reaction
19.
Biomarkers ; 21(5): 466-73, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26983903

ABSTRACT

Context Matrix metalloproteinases (MMPs) are associated with diastolic dysfunction and heart failure in acquired heart disease. Objective To investigate the role of MMPs as novel biomarkers in clinically stable adults with congenital heart disease. Methods We measured serum MMP-2, -3, -9 and tissue inhibitor of matrix metalloproteinase-1 in 425 patients and analysed the association with cardiac function and exercise capacity. Results MMP-2 was significantly associated with exercise capacity, ventilatory efficiency and left ventricular deceleration time, independently of age, sex, body surface area and NT-proBNP. Conclusion MMP-2 may provide new information in the clinical evaluation of adults with congenital heart disease.


Subject(s)
Heart Defects, Congenital/enzymology , Heart Function Tests/methods , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinases/blood , Adult , Biomarkers/blood , Female , Heart Defects, Congenital/blood , Humans , Male , Physical Exertion
20.
Circ Cardiovasc Genet ; 8(5): 665-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26232087

ABSTRACT

BACKGROUND: Oxidative stress is manifested in embryos exposed to maternal diabetes mellitus, yet specific mechanisms for diabetes mellitus-induced heart defects are not defined. Gene deletion of intermediates of Wingless-related integration (Wnt) signaling causes heart defects similar to those observed in embryos from diabetic pregnancies. We tested the hypothesis that diabetes mellitus-induced oxidative stress impairs Wnt signaling, thereby causing heart defects, and that these defects can be rescued by transgenic overexpression of the reactive oxygen species scavenger superoxide dismutase 1 (SOD1). METHODS AND RESULTS: Wild-type (WT) and SOD1-overexpressing embryos from nondiabetic WT control dams and nondiabetic/diabetic WT female mice mated with SOD1 transgenic male mice were analyzed. No heart defects were observed in WT and SOD1 embryos under nondiabetic conditions. WT embryos of diabetic dams had a 26% incidence of cardiac outlet defects that were suppressed by SOD1 overexpression. Insulin treatment reduced blood glucose levels and heart defects. Diabetes mellitus increased superoxide production, canonical Wnt antagonist expression, caspase activation, and apoptosis and suppressed cell proliferation. Diabetes mellitus suppressed Wnt signaling intermediates and Wnt target gene expression in the embryonic heart, each of which were reversed by SOD1 overexpression. Hydrogen peroxide and peroxynitrite mimicked the inhibitory effect of high glucose on Wnt signaling, which was abolished by the SOD1 mimetic, tempol. CONCLUSIONS: The oxidative stress of diabetes mellitus impairs Wnt signaling and causes cardiac outlet defects that are rescued by SOD1 overexpression. This suggests that targeting of components of the Wnt5a signaling pathway may be a viable strategy for suppression of congenital heart defects in fetuses of diabetic pregnancies.


Subject(s)
Heart Defects, Congenital/genetics , Oxidative Stress , Pregnancy in Diabetics , Superoxide Dismutase/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis , Diabetes Complications/genetics , Diabetes Mellitus, Experimental , Female , Gene Expression , Heart/embryology , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/metabolism , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Transgenic , Peroxynitrous Acid/pharmacology , Pregnancy , Signal Transduction/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Wnt Proteins/genetics , Wnt-5a Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...