Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 320(5): H1873-H1886, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33739154

RESUMO

Compared with acyanotic congenital heart disease (CHD), cyanotic CHD has an increased risk of lifelong mortality and morbidity. These adverse outcomes may be attributed to delayed cardiomyocyte maturation, since the transition from a hypoxic fetal milieu to oxygen-rich postnatal environment is disrupted. We established a rodent model to replicate hypoxic myocardial conditions spanning perinatal development, and tested the hypothesis that chronic hypoxia impairs cardiac development. Pregnant mice were housed in hypoxia beginning at embryonic day 16. Pups stayed in hypoxia until postnatal day (P)8 when cardiac development is nearly complete. Global gene expression was quantified at P8 and at P30, after recovering in normoxia. Phenotypic testing included electrocardiogram, echocardiogram, and ex vivo electrophysiology study. Hypoxic P8 animals were 47% smaller than controls with preserved heart size. Gene expression was grossly altered by hypoxia at P8 (1,427 genes affected), but normalized after recovery (P30). Electrocardiograms revealed bradycardia and slowed conduction velocity in hypoxic animals at P8, with noticeable resolution after recovery (P30). Notable differences that persisted after recovery (P30) included a 65% prolongation in ventricular effective refractory period, sinus node dysfunction, 23% reduction in ejection fraction, and 16% reduction in fractional shortening in animals exposed to hypoxia. We investigated the impact of chronic hypoxia on the developing heart. Perinatal hypoxia was associated with changes in gene expression and cardiac function. Persistent changes to the electrophysiological substrate and contractile function warrant further investigation and may contribute to adverse outcomes observed in the cyanotic CHD population.NEW & NOTEWORTHY We utilized a new mouse model of chronic perinatal hypoxia to simulate the hypoxic myocardial conditions present in cyanotic congenital heart disease. Hypoxia caused numerous abnormalities in cardiomyocyte gene expression, the electrophysiologic substrate of the heart, and contractile function. Taken together, alterations observed in the neonatal period suggest delayed cardiac development immediately following hypoxia.


Assuntos
Cianose/etiologia , Coração Fetal/crescimento & desenvolvimento , Cardiopatias Congênitas/etiologia , Hipóxia/complicações , Fatores Etários , Animais , Animais Recém-Nascidos , Doença Crônica , Cianose/genética , Cianose/metabolismo , Cianose/fisiopatologia , Modelos Animais de Doenças , Feminino , Coração Fetal/metabolismo , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Frequência Cardíaca , Hipóxia/genética , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Organogênese , Gravidez , Efeitos Tardios da Exposição Pré-Natal
2.
J Am Heart Assoc ; 10(2): e017839, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33399005

RESUMO

The failure of adult cardiomyocytes to reproduce themselves to repair an injury results in the development of severe cardiac disability leading to death in many cases. The quest for an understanding of the inability of cardiac myocytes to repair an injury has been ongoing for decades with the identification of various factors which have a temporary effect on cell-cycle activity. Fetal cardiac myocytes are continuously replicating until the time that the developing fetus reaches a stage of maturity sufficient for postnatal life around the time of birth. Recent reports of the ability for early neonatal mice and pigs to completely repair after the severe injury has stimulated further study of the regulators of the cardiomyocyte cell cycle to promote replication for the remuscularization of injured heart. In all mammals just before or after birth, single-nucleated hyperplastically growing cardiomyocytes, 1X2N, undergo ≥1 additional DNA replications not followed by cytokinesis, resulting in cells with ≥2 nuclei or as in primates, multiple DNA replications (polyploidy) of 1 nucleus, 2X2(+)N or 1X4(+)N. All further growth of the heart is attributable to hypertrophy of cardiomyocytes. Animal studies ranging from zebrafish with 100% 1X2N cells in the adult to some strains of mice with up to 98% 2X2N cells in the adult and other species with variable ratios of 1X2N and 2X2N cells are reviewed relative to the time of conversion. Various structural, physiologic, metabolic, genetic, hormonal, oxygenation, and other factors that play a key role in the inability of post-neonatal and adult myocytes to undergo additional cytokinesis are also reviewed.


Assuntos
Ciclo Celular/fisiologia , Coração Fetal , Miócitos Cardíacos , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/fisiologia , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências , Coração Fetal/citologia , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/metabolismo , Mamíferos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia
3.
J Perinat Med ; 49(3): 371-376, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33085638

RESUMO

OBJECTIVES: To evaluate 24-segment fractional shortening (FS) of the fetal heart using FetalHQ by speckle-tracking regarding reproducibility and the change with advancing gestation. METHODS: Eighty-one pregnant women at 18-21+6 and 28-31+6 weeks of gestation were studied using FetalHQ with the speckle-tracking technique to calculate 24-segment FS of left and right ventricles. Intra- and inter-class correlation coefficients and intra- and inter-observer agreements of measurements for FS were assessed in each segment. RESULTS: With respect to intra-observer reproducibility, all FS values showed correlations between 0.575 and 0.862 for the left ventricle, with good intra-observer agreements except for left ventricular segments 14-24. Right ventricular FS values showed correlations between 0.334 and 0.685, with good intra-observer agreements. With respect to inter-observer reproducibility, all FS values showed correlations between 0.491 and 0.801 for the left ventricle, with good intra-observer agreements except for left ventricular segments 16-22. Right ventricular FS values showed correlations between 0.375 and 0.575, with good inter-observer agreements. There were significant differences in the mean FS values in the basal segment (segments 1-5) of the left ventricle between 18 and 21+6 and 28-31+6 weeks of gestation (p<0.05), whereas there were significant differences in all mean FS values in the right ventricle between both gestational ages (p<0.05). CONCLUSIONS: These results suggest that the reproducibility of the 24-segment FS of the fetal heart using FetalHQ is fair. However, there may be significant differences in FS values with advancing gestational age, especially for the right ventricle.


Assuntos
Coração Fetal , Ventrículos do Coração , Ultrassonografia Pré-Natal/métodos , Adulto , Feminino , Coração Fetal/diagnóstico por imagem , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/fisiologia , Maturidade dos Órgãos Fetais , Idade Gestacional , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/crescimento & desenvolvimento , Humanos , Variações Dependentes do Observador , Tamanho do Órgão , Gravidez , Reprodutibilidade dos Testes
4.
BMC Cardiovasc Disord ; 20(1): 369, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32795258

RESUMO

BACKGROUND: It is unclear whether the offspring of subclinical hypothyroidism (SCH) pregnant rats still have abnormal cardiac development, and whether early intervention with L-T4 can improve the abnormality of these offspring. Therefore, the aim of this study was to investigate the effect of early L-T4 intervention on the heart development of offspring of SCH pregnant rats and its possible molecular mechanism. METHODS: Eighty female Wistar rats were randomly divided into Sham group (placebo control), SCH group, LT4-E10 group (L-T4 treatment started on the 10th day of gestation), and LT4-E13 group (L-T4 treatment started on the 13th day of gestation). Each group was further divided into E16 (16th day of gestation), E18 (18th day of gestation), P5 (5th day postnatal day), and P10 (10th day postnatal day) subgroups. The levels of serum TT4 and TSH, the ratio of heart weight to body weight of offspring rats, the expression of metabolic enzymes, and the histopathology of cardiomyocytes were determined. To elucidate the effects of L-T4 on cardiac development of offspring of SCH pregnant rats, the expression levels of GATA4, Nkx2-5 and proteins involved in BMP4/Smad4 signaling pathway were detected by immunohistochemistry, real time quantitative polymerase chain reaction and Western blotting to elucidate the molecular mechanism of L-T4 regulating the heart development of the offspring of SCH pregnant rats. RESULTS: Compared with Sham group, serum TSH was significantly increased in SCH pregnant rats. Moreover, early L-T4 intervention significantly reduced the levels of serum TSH. Compared with the offspring in the SCH group, early L-T4 intervention significantly increased the heart weight, heart weight to body weight ratio, the activities of succinate dehydrogenase (SDH), Na+/K+-ATPase and Ca2+-ATPase, but reduced myocardial cell shrinkage and nuclear staining, hyperemia/congestion and vacuolar degeneration. In addition, early L-T4 intervention not only significantly increased the mRNA and protein expression of Gata4 and Nkx2-5, but also increased the protein expression involved in BMP4/Smad4 signal pathway in myocardium of the offspring of SCH pregnant rats. CONCLUSIONS: Early L-T4 intervention can regulate the cardiac development of the offspring of SCH pregnant rats by activating BMP4/Smad4 signaling pathway and increasing the expression of Gata4 and Nkx2-5 proteins.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Coração Fetal/efeitos dos fármacos , Hipotireoidismo/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Proteína Smad4/metabolismo , Tiroxina/farmacologia , Animais , Doenças Assintomáticas , Modelos Animais de Doenças , Feminino , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/metabolismo , Fator de Transcrição GATA4/metabolismo , Idade Gestacional , Proteína Homeobox Nkx-2.5/metabolismo , Hipotireoidismo/metabolismo , Hipotireoidismo/fisiopatologia , Miócitos Cardíacos/metabolismo , Gravidez , Ratos Wistar , Transdução de Sinais
6.
Ultrasound Obstet Gynecol ; 55(4): 516-522, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30989734

RESUMO

OBJECTIVE: EXTrauterine Environment for Neonatal Development (EXTEND) is a system to support ongoing fetal growth and organ development in an extrauterine environment, utilizing a pumpless low-resistance oxygenator circuit. The aim of this study was to evaluate hemodynamics and cardiac function in fetal sheep sustained on the EXTEND system. METHODS: This was a prospective study of fetal sheep supported for a minimum of 3 weeks on EXTEND. Hemodynamic parameters were assessed weekly and included heart rate, mean arterial pressure (MAP), Doppler-echocardiography-derived cardiac output (CO), pulsatility indices (PIs) of the fetal middle cerebral artery (MCA), umbilical artery (UA) and ductus venosus and cardiac function, as assessed by speckle-tracking-derived global longitudinal strain and strain rate in the right (RV) and left (LV) ventricles. Parameters were compared at 0 days and 1, 2 and 3 weeks following placement on EXTEND. RESULTS: Of 10 fetal sheep enrolled, seven survived for 3 weeks and were included in the analysis. Median gestational age at cannulation was 107 (range, 95-109) days. Heart rate decreased and MAP increased significantly, but within acceptable ranges, during the study period. The quantities and relative ratios of right and left CO remained stable within the anticipated physiological range throughout the study period. Vascular tracings and PIs appeared to be similar to those seen normally in the natural in-utero state, with MCA-PI being higher than UA-PI. UA tracings demonstrated maintained abundant diastolic flow despite the absence of placental circulation. In both the RV and LV, strain decreased significantly at 1 and 2 weeks relative to baseline but returned to baseline values by week 3. CONCLUSIONS: The EXTEND mechanical support system replicates natural physiology and creates a stable and sustainable cardiovascular construct that supports growth over a 3-week period. However, there is a period of depressed contractility within the first week with subsequent improvement by week 3. This may reflect a period of physiological accommodation that warrants further investigation. This study lays the foundation for further exploration as the EXTEND system moves towards human application. © 2019 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of the International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Cardiotocografia/métodos , Ecocardiografia Doppler/métodos , Oxigenação por Membrana Extracorpórea , Coração Fetal/diagnóstico por imagem , Feto/diagnóstico por imagem , Animais , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/crescimento & desenvolvimento , Débito Cardíaco , Feminino , Desenvolvimento Fetal/fisiologia , Coração Fetal/embriologia , Coração Fetal/crescimento & desenvolvimento , Feto/embriologia , Feto/fisiopatologia , Ventrículos do Coração/diagnóstico por imagem , Hemodinâmica , Artéria Cerebral Média/embriologia , Gravidez , Estudos Prospectivos , Fluxo Pulsátil , Ovinos , Fatores de Tempo , Artérias Umbilicais/embriologia
7.
Eur J Prev Cardiol ; 27(1): 63-74, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31357885

RESUMO

OBJECTIVES: Early life is critical for cardiac development. We examined the associations of longitudinal fetal and childhood growth patterns with childhood right and left ventricular structures measured by cardiac magnetic resonance imaging. METHODS: In a population-based prospective cohort study among 2827 children, we measured growth at 20 and 30 weeks of pregnancy, at birth, 0.5, 1, 2, 6 and 10 years. At 10 years, we measured right ventricular end-diastolic volume, left ventricular end-diastolic volume, left ventricular mass and left ventricular mass-to-volume ratio by cardiac magnetic resonance imaging. RESULTS: Small size for gestational age at birth was associated with smaller right and left ventricular end-diastolic volume relative to current body surface area, but with larger left ventricular mass-to-volume ratio (P < 0.05). Children in the upper 25% of right and left ventricular end-diastolic volume and left ventricular mass at age 10 years were larger at birth and became taller and leaner in childhood (P < 0.05). In contrast, children in the lower 25% of right and left ventricular end-diastolic volume and left ventricular mass were smaller at birth and became shorter and heavier in childhood (P < 0.05). Both fetal and childhood growth were independently of each other associated with childhood right and left ventricular end-diastolic volume and left ventricular mass. CONCLUSION: Children who are larger at birth and grow taller and leaner in childhood have larger hearts relative to body surface area. Small size at birth children, who grow shorter and heavier in childhood, have relatively smaller hearts with larger left ventricular mass-to-volume ratio. Both fetal and childhood growth are important for the development of cardiac dimensions.


Assuntos
Desenvolvimento Infantil , Coração Fetal/diagnóstico por imagem , Ventrículos do Coração/diagnóstico por imagem , Imagem Cinética por Ressonância Magnética , Fatores Etários , Peso ao Nascer , Estatura , Criança , Pré-Escolar , Feminino , Coração Fetal/crescimento & desenvolvimento , Idade Gestacional , Ventrículos do Coração/crescimento & desenvolvimento , Humanos , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Valor Preditivo dos Testes , Gravidez , Estudos Prospectivos , Aumento de Peso
8.
Cells ; 8(12)2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31835778

RESUMO

Antenatal hypoxia caused epigenetic reprogramming of methylome and transcriptome in the developing heart and increased the risk of heart disease later in life. Herein, we investigated the impact of gestational hypoxia in proteome and metabolome in the hearts of fetus and adult offspring. Pregnant rats were treated with normoxia or hypoxia (10.5% O2) from day 15 to 21 of gestation. Hearts were isolated from near-term fetuses and 5 month-old offspring, and proteomics and metabolomics profiling was determined. The data demonstrated that antenatal hypoxia altered proteomics and metabolomics profiling in the heart, impacting energy metabolism, lipid metabolism, oxidative stress, and inflammation-related pathways in a developmental and sex dependent manner. Of importance, integrating multi-omics data of transcriptomics, proteomics, and metabolomics profiling revealed reprogramming of the mitochondrion, especially in two clusters: (a) the cluster associated with "mitochondrial translation"/"aminoacyl t-RNA biosynthesis"/"one-carbon pool of folate"/"DNA methylation"; and (b) the cluster with "mitochondrion"/"TCA cycle and respiratory electron transfer"/"acyl-CoA dehydrogenase"/"oxidative phosphorylation"/"complex I"/"troponin myosin cardiac complex". Our study provides a powerful means of multi-omics data integration and reveals new insights into phenotypic reprogramming of the mitochondrion in the developing heart by fetal hypoxia, contributing to an increase in the heart vulnerability to disease later in life.


Assuntos
Coração Fetal/crescimento & desenvolvimento , Hipóxia Fetal/metabolismo , Metabolômica/métodos , Proteômica/métodos , Animais , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Coração Fetal/metabolismo , Metabolismo dos Lipídeos , Masculino , Estresse Oxidativo , Gravidez , Ratos , Caracteres Sexuais
9.
Card Electrophysiol Clin ; 11(3): 409-420, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31400866

RESUMO

Embryogenesis of the heart involves the complex cellular differentiation of slow-conducting primary myocardium into the rapidly conducting chamber myocardium of the adult. However, small areas of relatively undifferentiated cells remain to form components of the adult cardiac conduction system (CCS) and nodal tissues. Further investigation has revealed additional areas of nodal-like tissues outside of the established CCS. The embryologic origins of these areas are similar to those of the adult CCS. Under pathologic conditions, these areas can give rise to important clinical arrhythmias. Here, we review the embryologic basis for these proarrhythmic structures within the heart.


Assuntos
Arritmias Cardíacas , Coração Fetal , Sistema de Condução Cardíaco , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Coração Fetal/embriologia , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/fisiologia , Sistema de Condução Cardíaco/embriologia , Sistema de Condução Cardíaco/crescimento & desenvolvimento , Sistema de Condução Cardíaco/fisiologia , Humanos
10.
Early Hum Dev ; 129: 23-32, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30616038

RESUMO

AIM: To test the hypothesis that right ventricular (RV) function has age-specific patterns of development, we tracked the evolution of RV strain mechanics by 2D-speckle tracking echocardiography (2DSTE) in healthy subjects from mid-gestation through one year of age. METHODS: We conducted a prospective longitudinal echocardiography study in 50 healthy subjects at five time periods across gestation (16-20 weeks, 21-25 weeks, 26-30 weeks, 31-35 weeks, and 36-40 weeks) and four time periods following delivery (1 week, 1 month, 6 months, and 1 year of age). We characterized RV function by measuring RV global and free wall longitudinal strain and systolic strain rate, and segmental longitudinal strain at the apical-, mid-, and basal- ventricular levels of the free wall. Possible associations of gestational age, postnatal age, estimated fetal weight, body surface area, gender, and heart rate on strain were investigated. RESULTS: The magnitudes of RV global and free wall longitudinal strain and global strain rate were decreased throughout gestation (p < 0.05 for all). Following birth, the magnitudes of all measures increased from one week through one year (p < 0.001 for all). RV segmental longitudinal strain maintained a base-to-apex gradient (highest-to-lowest) from mid-gestation through one year (p < 0.001). There was no significant difference in strain patterns based on gender or hear rate. CONCLUSION: The maturational patterns of RV strain are gestational- and postnatal age- specific. With accepted physiological maturation patterns in healthy subjects, these myocardial deformation parameters can provide a valid basis that allows comparison between health and disease.


Assuntos
Coração Fetal/crescimento & desenvolvimento , Ventrículos do Coração/crescimento & desenvolvimento , Adulto , Fenômenos Biomecânicos , Feminino , Coração Fetal/diagnóstico por imagem , Coração Fetal/fisiologia , Ventrículos do Coração/diagnóstico por imagem , Hemodinâmica , Humanos , Recém-Nascido , Masculino , Gravidez , Função Ventricular
11.
Nutr Metab Cardiovasc Dis ; 29(2): 170-176, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30579777

RESUMO

BACKGROUND AND AIMS: Intra-uterine metabolic environment predicts newborns' cardiac morphology, metabolism and future health. In adults, gut microbiota composition relates to altered cardiac structure and metabolism. We investigated the relationship between gut microbiota colonization and fetal cardiac growth. METHODS AND RESULTS: Bacterial composition in meconium samples of 26 healthy, full-term newborns was assessed by 16S rDNA gene sequencing. Its relationship with birth echocardiographic parameters, and the interaction with cord blood levels of inflammatory markers were investigated. Correlative and cluster analysis, linear discriminant analysis effect size and predictive functional analysis based on Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were applied. Fetal left ventricle growth was related to gut microbiota composition at birth. Specifically, left ventricle posterior wall thickness (LVPW) greater than 4 mm was associated with lower microbiota beta and alpha diversity, depletion (LDA score > 3) of several bacteria at each taxonomic level, including Lactobacillales, and enrichment (LDA score > 5) in Enterobacteriales and Enterobacteriaceae. The latter was significantly related to cord blood gamma-glutamyltransferase levels (r = 0.58, p = 0.0057). Functionally, a thicker LVPW was related to up-regulation of pathways involved in lipopolysaccharide biosynthesis (+50%, p = 0.045 in correlative analysis) and energy metabolism (+12%, p = 0.028), and down-regulation of pathways involved in xenobiotic biodegradation (-21 to -53%, p = 0.0063-0.039), PPAR signaling (-24%, p = 0.021) and cardiac muscle contraction (-100%, p = 0.049). CONCLUSION: Fetal cardiac growth and gut colonization are associated. Greater neonatal LVPW thickness is related to lower diversity of the gut microbiota community, depletion of bacteria having anti-remodeling effects, and enrichment in bacteria functionally linked to inflammation.


Assuntos
Bactérias/crescimento & desenvolvimento , Coração Fetal/crescimento & desenvolvimento , Microbioma Gastrointestinal , Ventrículos do Coração/crescimento & desenvolvimento , Intestinos/microbiologia , Bactérias/classificação , Bactérias/genética , Biomarcadores/sangue , Ecocardiografia , Sangue Fetal/química , Coração Fetal/diagnóstico por imagem , Trato Gastrointestinal , Ventrículos do Coração/diagnóstico por imagem , Interações Hospedeiro-Patógeno , Humanos , Recém-Nascido , Mediadores da Inflamação/sangue , Mecônio/microbiologia , Ribotipagem
12.
Nat Rev Cardiol ; 15(11): 685-704, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30287945

RESUMO

Cardiogenesis is a complex developmental process involving multiple overlapping stages of cell fate specification, proliferation, differentiation, and morphogenesis. Precise spatiotemporal coordination between the different cardiogenic processes is ensured by intercellular signalling crosstalk and tissue-tissue interactions. Notch is an intercellular signalling pathway crucial for cell fate decisions during multicellular organismal development and is aptly positioned to coordinate the complex signalling crosstalk required for progressive cell lineage restriction during cardiogenesis. In this Review, we describe the role of Notch signalling and the crosstalk with other signalling pathways during the differentiation and patterning of the different cardiac tissues and in cardiac valve and ventricular chamber development. We examine how perturbation of Notch signalling activity is linked to congenital heart diseases affecting the neonate and adult, and discuss studies that shed light on the role of Notch signalling in heart regeneration and repair after injury.


Assuntos
Cardiopatias/metabolismo , Valvas Cardíacas/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Cross-Talk , Receptores Notch/metabolismo , Regeneração , Transdução de Sinais , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Valvas Cardíacas/patologia , Valvas Cardíacas/fisiopatologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Modelos Animais , Miócitos Cardíacos/patologia , Organogênese , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
13.
Nat Rev Cardiol ; 15(11): 672-684, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30111784

RESUMO

The Hippo-YAP (Yes-associated protein) pathway is an evolutionarily and functionally conserved regulator of organ size and growth with crucial roles in cell proliferation, apoptosis, and differentiation. This pathway has great potential for therapeutic manipulation in different disease states and to promote organ regeneration. In this Review, we summarize findings from the past decade revealing the function and regulation of the Hippo-YAP pathway in cardiac development, growth, homeostasis, disease, and regeneration. In particular, we highlight the roles of the Hippo-YAP pathway in endogenous heart muscle renewal, including the pivotal role of the Hippo-YAP pathway in regulating cardiomyocyte proliferation and differentiation, stress response, and mechanical signalling. The human heart lacks the capacity to self-repair; therefore, the loss of cardiomyocytes after injury such as myocardial infarction can result in heart failure and death. Despite substantial advances in the treatment of heart failure, an enormous unmet clinical need exists for alternative treatment options. Targeting the Hippo-YAP pathway has tremendous potential for developing therapeutic strategies for cardiac repair and regeneration for currently intractable cardiovascular diseases such as heart failure. The lessons learned from cardiac repair and regeneration studies will also bring new insights into the regeneration of other tissues with limited regenerative capacity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cardiopatias/metabolismo , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Via de Sinalização Hippo , Humanos , Miócitos Cardíacos/patologia , Organogênese , Fatores de Transcrição , Proteínas de Sinalização YAP
14.
J Mol Cell Cardiol ; 121: 60-68, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29969579

RESUMO

G-protein receptor kinases (GRKs) regulate adult hearts by modulating inotropic, chronotropic and hypertrophic signaling of 7-transmembrane spanning neurohormone receptors. GRK-mediated desensitization and downregulation of ß-adrenergic receptors has been implicated in adult heart failure; GRKs are therefore a promising therapeutic target. However, germ-line (but not cardiomyocyte-specific) GRK2 deletion provoked lethal fetal heart defects, suggesting an unexplained role for GRKs in heart development. Here we undertook to better understand the consequences of GRK deficiency on fetal heart development by creating mice and cultured murine embryonic fibroblasts (MEFs) having floxed GRK2 and GRK5 alleles on the GRK6 null background; simultaneous conditional deletion of these 3 GRK genes was achieved using Nkx2-5 Cre or adenoviral Cre, respectively. Phenotypes were related to GRK-modulated gene expression using whole-transcriptome RNA sequencing, RT-qPCR, and luciferase reporter assays. In cultured MEFs the atypical 7-transmembrane spanning protein and GRK2 substrate Smoothened (Smo) stimulated Gli-mediated transcriptional activity, which was interrupted by deleting GRK2/5/6. Mice with Nkx2-5 Cre mediated GRK2/5/6 ablation died between E15.5 and E16.5, whereas mice expressing any one of these 3 GRKs (i.e. GRK2/5, GRK2/6 or GRK5/6 deleted) were developmentally normal. GRK2/5/6 triple null mice at E14.5 exhibited left and right heart blood intermixing through single atrioventricular valves or large membranous ventricular septal defects. Hedgehog and GATA pathway gene expression promoted by Smo/Gli was suppressed in GRK2/5/6 deficient fetal hearts and MEFs. These data indicate that GRK2, GRK5 and GRK6 redundantly modulate Smo-GATA crosstalk in fetal mouse hearts, orchestrating transcriptional pathways previously linked to clinical and experimental atrioventricular canal defects. GRK modulation of Smo reflects convergence of conventional neurohormonal signaling and transcriptional regulation pathways, comprising an unanticipated mechanism for spatiotemporal orchestration of developmental gene expression in the heart.


Assuntos
Coração Fetal/crescimento & desenvolvimento , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/genética , Receptor Smoothened/genética , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Coração Fetal/fisiopatologia , Fibroblastos/metabolismo , Fator de Transcrição GATA1/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Análise de Sequência de RNA , Transcriptoma/genética
15.
Circ Cardiovasc Imaging ; 11(7): e007575, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30006353

RESUMO

BACKGROUND: Assessment of the outflow tract views is an integral part of routine fetal cardiac scanning. For some congenital heart defects, notably coarctation of the aorta, pulmonary valve stenosis, and aortic valve stenosis, the size of vessels is important both for diagnosis and prognosis. Existing reference ranges of fetal outflow tracts are derived from a small number of cases. METHODS AND RESULTS: The study population comprised 7945 fetuses at 13 to 36 weeks' gestation with no detectable abnormalities from pregnancies resulting in normal live births. Prospective measurements were taken of (1) the aortic and pulmonary valves in diastole at the largest diameter with the valve closed, (2) the distal transverse aortic arch on the 3 vessel and trachea view beyond the trachea at the distal point at its widest systolic diameter, and (3) the arterial duct on the 3 vessel and trachea view at its widest systolic diameter. Regression analysis, with polynomial terms to assess for linear and nonlinear contributors, was used to establish the relationship between each measurement and gestational age. The measurement for each cardiac diameter was expressed as a z score (difference between observed and expected value divided by the fitted SD corrected for gestational age) and percentile. Analysis included calculation of gestation-specific SDs. Regression equations are provided for the cardiac outflow tracts and for the distal transverse aortic arch:arterial duct ratio. CONCLUSIONS: The study established reference ranges for fetal outflow tract measurements at 13 to 36 weeks' gestation that are useful in clinical practice.


Assuntos
Ecocardiografia , Coração Fetal/diagnóstico por imagem , Ultrassonografia Pré-Natal/métodos , Pontos de Referência Anatômicos , Aorta Torácica/diagnóstico por imagem , Aorta Torácica/embriologia , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/embriologia , Canal Arterial/diagnóstico por imagem , Canal Arterial/embriologia , Ecocardiografia/normas , Feminino , Coração Fetal/crescimento & desenvolvimento , Idade Gestacional , Humanos , Morfogênese , Valor Preditivo dos Testes , Gravidez , Estudos Prospectivos , Valva Pulmonar/diagnóstico por imagem , Valva Pulmonar/embriologia , Valores de Referência , Ultrassonografia Pré-Natal/normas
16.
Adv Exp Med Biol ; 1065: 347-360, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30051395

RESUMO

The size, hemodynamics, and function of cardiovascular structures change dramatically from the early fetal life to late adolescence. The principal determinants of cardiovascular dimensions are related to the blood flow needed to meet metabolic demands. This demand is in turn tightly related to body size and body composition, keeping in mind that various tissues may have different metabolic rates. There is no simple model that links cardiac dimensions with a single body size measurement. Consequently, despite abundant scientific literature, few studies have proposed pediatric reference values that efficiently and completely account for the effect of body size. Other factors influence cardiovascular size and function in children, including sex. The influence of sex is multifactorial and not fully understood, but differences in body size and body composition play an important role. We will first review the determinants of cardiovascular size and function in children. We then explore the evaluation and normalization of cardiovascular size and function in pediatric cardiology in relation to the growth of cardiovascular structures during childhood, with a particular focus on sex differences.


Assuntos
Desenvolvimento do Adolescente , Sistema Cardiovascular/crescimento & desenvolvimento , Desenvolvimento Infantil , Coração Fetal/crescimento & desenvolvimento , Hemodinâmica , Função Ventricular , Adolescente , Fatores Etários , Animais , Tamanho Corporal , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/embriologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Morfogênese , Fatores de Risco , Caracteres Sexuais , Fatores Sexuais
17.
Physiol Rev ; 98(3): 1241-1334, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717932

RESUMO

Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.


Assuntos
Desenvolvimento Fetal , Hipóxia Fetal/metabolismo , Adaptação Fisiológica , Tecido Adiposo/embriologia , Animais , Epigênese Genética , Feminino , Coração Fetal/crescimento & desenvolvimento , Cardiopatias/etiologia , Humanos , Hipertensão Pulmonar/congênito , Sistema Hipotálamo-Hipofisário , Saúde Materna , Sistema Hipófise-Suprarrenal , Circulação Placentária , Gravidez
18.
Nat Commun ; 9(1): 754, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467410

RESUMO

The cellular mechanisms driving cardiac tissue formation remain poorly understood, largely due to the structural and functional complexity of the heart. It is unclear whether newly generated myocytes originate from cardiac stem/progenitor cells or from pre-existing cardiomyocytes that re-enter the cell cycle. Here, we identify the source of new cardiomyocytes during mouse development and after injury. Our findings suggest that cardiac progenitors maintain proliferative potential and are the main source of cardiomyocytes during development; however, the onset of αMHC expression leads to reduced cycling capacity. Single-cell RNA sequencing reveals a proliferative, "progenitor-like" population abundant in early embryonic stages that decreases to minimal levels postnatally. Furthermore, cardiac injury by ligation of the left anterior descending artery was found to activate cardiomyocyte proliferation in neonatal but not adult mice. Our data suggest that clonal dominance of differentiating progenitors mediates cardiac development, while a distinct subpopulation of cardiomyocytes may have the potential for limited proliferation during late embryonic development and shortly after birth.


Assuntos
Traumatismos Cardíacos/patologia , Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células-Tronco Embrionárias/citologia , Feminino , Coração Fetal/citologia , Coração Fetal/crescimento & desenvolvimento , Traumatismos Cardíacos/genética , Masculino , Camundongos , Camundongos Transgênicos , Mioblastos Cardíacos/citologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Pericárdio/citologia , Pericárdio/embriologia , Pericárdio/crescimento & desenvolvimento , Gravidez , Análise de Sequência de RNA
19.
J Appl Toxicol ; 38(6): 834-842, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29377175

RESUMO

Accumulating evidence has suggested a link between maternal di-(2-ethylhexyl)-phthalate (DEHP) exposure and various developmental abnormalities. However, the evidence regarding the effect of maternal DEHP exposure on fetal cardiac development is scarce. The present study aimed to determine the effect of maternal DEHP exposure on fetal cardiac development in mice and explore the possible involved mechanism preliminarily. The C57BL mice were randomly divided into four groups: the vehicle group (corn oil, n = 10), 250 mg kg-1 DEHP group (n = 15), 500 mg kg-1 DEHP group (n = 20) and 1 g kg-1 DEHP group (n = 20). Pregnant dams in different group received respective intervention by gavage once daily from embryonic day (E)6.5 to E14.5. Maternal weights were monitored every day and samples were collected at E15.5. Hematoxylin and eosin staining was used to examine fetal cardiac malformations. Real-time quantitative polymerase chain reaction and western blot were applied to detect peroxisome proliferator-activated receptor (PPAR)α/PPARγ/Nkx2.5/Gata4/Tbx5/Mef2c/Chf1 mRNA and protein expression, respectively. Maternal DEHP exposure significantly decreased maternal body weight, fetal weight and placental weight, and remarkably elevated fetal cardiac malformations rate. The phenotypes of cardiac anomalies mainly include septal defects, ventricular myocardium noncompaction and cardiac hypoplasia. Higher doses DEHP (500 mg kg-1 and 1 g kg-1 ) could significantly decreased fetal cardiac Gata4/Mef2c/Chf1 expression, while PPARγ expression was upregulated. Maternal exposure to higher doses of DEHP could result in fetal cardiac development malformations in mice and it might have resulted from the inhibition of cardiac GATA4/Mef2c/Chf1 expression via PPARγ activation.


Assuntos
Dietilexilftalato/toxicidade , Coração Fetal/efeitos dos fármacos , Cardiopatias Congênitas/induzido quimicamente , Plastificantes/toxicidade , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Feminino , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Exposição Materna , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , PPAR gama/agonistas , PPAR gama/genética , PPAR gama/metabolismo , Gravidez , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
J Am Heart Assoc ; 6(8)2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28778941

RESUMO

BACKGROUND: Fetal growth impacts cardiovascular health throughout postnatal life in humans. Various animal models of intrauterine growth restriction exhibit reduced heart size at birth, which negatively influences cardiac function in adulthood. The mechanistic target of rapamycin complex 1 (mTORC1) integrates nutrient and growth factor availability with cell growth, thereby regulating organ size. This study aimed at elucidating a possible involvement of mTORC1 in intrauterine growth restriction and prenatal heart growth. METHODS AND RESULTS: We inhibited mTORC1 in fetal mice by rapamycin treatment of pregnant dams in late gestation. Prenatal rapamycin treatment reduces mTORC1 activity in various organs at birth, which is fully restored by postnatal day 3. Rapamycin-treated neonates exhibit a 16% reduction in body weight compared with vehicle-treated controls. Heart weight decreases by 35%, resulting in a significantly reduced heart weight/body weight ratio, smaller left ventricular dimensions, and reduced cardiac output in rapamycin- versus vehicle-treated mice at birth. Although proliferation rates in neonatal rapamycin-treated hearts are unaffected, cardiomyocyte size is reduced, and apoptosis increased compared with vehicle-treated neonates. Rapamycin-treated mice exhibit postnatal catch-up growth, but body weight and left ventricular mass remain reduced in adulthood. Prenatal mTORC1 inhibition causes a reduction in cardiomyocyte number in adult hearts compared with controls, which is partially compensated for by an increased cardiomyocyte volume, resulting in normal cardiac function without maladaptive left ventricular remodeling. CONCLUSIONS: Prenatal rapamycin treatment of pregnant dams represents a new mouse model of intrauterine growth restriction and identifies an important role of mTORC1 in perinatal cardiac growth.


Assuntos
Retardo do Crescimento Fetal/induzido quimicamente , Coração Fetal/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Efeitos Tardios da Exposição Pré-Natal , Sirolimo/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/patologia , Idade Gestacional , Proteína Homeobox Nkx-2.5/genética , Liases/deficiência , Liases/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Tamanho do Órgão , Organogênese/efeitos dos fármacos , Gravidez , Regiões Promotoras Genéticas , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...