Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 454
Filtrar
1.
Dev Cell ; 58(24): 2867-2880.e7, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-37972593

RESUMO

Cardiomyocytes are highly metabolic cells responsible for generating the contractile force in the heart. During fetal development and regeneration, these cells actively divide but lose their proliferative activity in adulthood. The mechanisms that coordinate their metabolism and proliferation are not fully understood. Here, we study the role of the transcription factor NFYa in developing mouse hearts. Loss of NFYa alters cardiomyocyte composition, causing a decrease in immature regenerative cells and an increase in trabecular and mature cardiomyocytes, as identified by spatial and single-cell transcriptome analyses. NFYa-deleted cardiomyocytes exhibited reduced proliferation and impaired mitochondrial metabolism, leading to cardiac growth defects and embryonic death. NFYa, interacting with cofactor SP2, activates genes linking metabolism and proliferation at the transcription level. Our study identifies a nodal role of NFYa in regulating prenatal cardiac growth and a previously unrecognized transcriptional control mechanism of heart metabolism, highlighting the importance of mitochondrial metabolism during heart development and regeneration.


Assuntos
Miócitos Cardíacos , Fatores de Transcrição , Animais , Camundongos , Proliferação de Células/fisiologia , Desenvolvimento Fetal , Coração Fetal/metabolismo , Coração/fisiologia , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo
2.
Am J Physiol Regul Integr Comp Physiol ; 325(6): R809-R819, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37867472

RESUMO

At birth, the fetus experiences a dramatic change in environment that is accompanied by a shift in myocardial fuel preference from lactate and glucose in fetal life to fatty acid oxidation after birth. We hypothesized that fatty acid metabolic machinery would mature during fetal life in preparation for this extreme metabolic transformation at birth. We quantified the pre- (94-day and 135-day gestation, term ∼147 days) and postnatal (5 ± 4 days postnatal) gene expression and protein levels for fatty acid transporters and enzymes in hearts from a precocial species, the sheep. Gene expression of fatty acid translocase (CD36), acyl-CoA synthetase long-chain 1 (ACSL1), carnitine palmitoyltransferase 1 (CPT1), hydroxy-acyl dehydrogenase (HADH), acetyl-CoA acetyltransferase (ACAT1), isocitrate dehydrogenase (IDH), and glycerol phosphate acyltransferase (GPAT) progressively increased through the perinatal period, whereas several genes [fatty acid transport protein 6 (FATP6), acyl-CoA synthetase long chain 3 (ACSL3), long-chain acyl-CoA dehydrogenase (LCAD), very long-chain acyl-CoA dehydrogenase (VLCAD), pyruvate dehydrogenase kinase (PDK4), phosphatidic acid phosphatase (PAP), and diacylglycerol acyltransferase (DGAT)] were stable in fetal hearts and had high expression after birth. Protein expression of CD36 and ACSL1 progressively increased throughout the perinatal period, whereas protein expression of carnitine palmitoyltransferase 1a (fetal isoform) (CPT1a) decreased and carnitine palmitoyltransferase 1b (adult isoform) (CPT1b) remained constitutively expressed. Using fluorescent-tagged long-chain fatty acids (BODIPY-C12), we demonstrated that fetal (125 ± 1 days gestation) cardiomyocytes produce 59% larger lipid droplets (P < 0.05) compared with newborn (8 ± 1 day) cardiomyocytes. These results provide novel insights into the perinatal maturation of cardiac fatty acid metabolism in a precocial species.NEW & NOTEWORTHY This study characterized the previously unknown expression patterns of genes that regulate the metabolism of free fatty acids in the perinatal sheep myocardium. This study shows that the prenatal myocardium prepares for the dramatic switch from carbohydrate metabolism to near complete reliance on free fatty acids postnatally. Fetal and neonatal cardiomyocytes also demonstrate differing lipid storage mechanisms where fetal cardiomyocytes form larger lipid droplets compared with newborn cardiomyocytes.


Assuntos
Carnitina O-Palmitoiltransferase , Ácidos Graxos não Esterificados , Gravidez , Feminino , Animais , Ovinos , Carnitina O-Palmitoiltransferase/metabolismo , Metabolismo dos Lipídeos , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Ácidos Graxos/metabolismo , Coração Fetal/metabolismo , Isoformas de Proteínas/metabolismo , Ligases/metabolismo , Oxirredução
3.
Clin Chim Acta ; 544: 117330, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37037297

RESUMO

AIMS: To explore the maternal metabolic changes of fetal congenital heart disease (FCHD), and screen metabolic markers to establish a practical diagnostic model. METHODS: Maternal peripheral serum from 17 FCHD and 63 non-FCHD pregnant were analyzed by Ultra High-performance Liquid Chromatography-Mass/Mass (UPLC-MS/MS). RESULTS: In the FCHD and the non-FCHD, 132 metabolites were identified, including 35 differential metabolites enriched in the purine, caffeine, primary bile acid, and arachidonic acid metabolism pathway. Finally, the screened (+/-)9,10-dihydroxy-12Z-octadecenoic acid (AUC = 0.888) and 11,12-epoxy-(5Z,8Z,11Z)-icosatrienoic acid (AUC = 0.995) were incorporated into the logistic regression model. The AUC value of the two-metabolite model was 1.0, superior to proline (AUC = 0.867), uric acid (AUC = 0.789), glutamine (AUC = 0.705), and taurine (AUC = 0.923) previously reported. The clinical decision curve analysis (DCA) showed the highest clinical net benefit of the model, and internal validation by bootstrap shows the robustness of the model (Brier Score = 0.005). CONCLUSION: For the prenatal diagnosis of CHD, our findings are of great clinical significance. As an additional screening procedure, the identification model might be used to detect.


Assuntos
Cardiopatias Congênitas , Espectrometria de Massas em Tandem , Gravidez , Feminino , Humanos , Cromatografia Líquida , Espectrometria de Massas em Tandem/métodos , Cardiopatias Congênitas/diagnóstico , Diagnóstico Pré-Natal , Coração Fetal/metabolismo , Lipídeos , Metabolômica/métodos , Biomarcadores
4.
Ecotoxicol Environ Saf ; 255: 114753, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36933485

RESUMO

BACKGROUND: Bisphenol A (BPA), a common endocrine-disrupting chemical (EDC), has been revealed to be closely associated with the induction of abnormal heart development, obesity, prediabetes, and other metabolic disorders. However, the underlying mechanism of maternal BPA exposure on fetal heart development abnormalities is not clear. METHODS: To explore the adverse effects of BPA and its potential mechanism on heart development, C57BL/6 J mice and human cardiac AC-16 cells were used to conduct in vivo and in vitro studies. For the in vivo study, mice were exposed to low-dose BPA (40 mg/(kg·bw)) and high-dose BPA (120 mg/(kg·bw)) for 18 d during pregnancy. In vitro study, human cardiac AC-16 cells were exposed to BPA of various concentrations (0.01, 0.1, 1, 10, and 100 µM) for 24 h. Cell viability and ferroptosis were evaluated using 2,5-diphenyl-2 H-tetrazolium bromide (MTT), immunofluorescence staining, and western blotting. RESULTS: In BPA-treated mice, the alterations of fetal cardiac structure were observed. Increased NK2 homeobox 5(Nkx2.5) was detected in vivo with the induction of ferroptosis, revealing that BPA induced abnormal fetal heart development. Furthermore, the results showed that SLC7A11 and SLC3A2 decreased in low- and high-dose BPA-treated groups, suggesting that system Xc- mediated BPA-induced abnormal fetal heart development via inhibiting the expression of GPX4. Observing AC-16 cells confirmed that cell viability declined significantly with various concentrations of BPA. Moreover, BPA exposure suppressed GPX4 expression by inhibiting System Xc- (the decrease of SLC3A2 and SLC7A11). Collectively, system Xc- modulating cell ferroptosis might play important in abnormal fetal heart development induced by BPA exposure.


Assuntos
Ferroptose , Gravidez , Feminino , Camundongos , Humanos , Animais , Camundongos Endogâmicos C57BL , Exposição Materna/efeitos adversos , Coração Fetal/metabolismo , Compostos Benzidrílicos/metabolismo
5.
Life Sci ; 314: 121308, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36563841

RESUMO

BACKGROUND: Maternal exercise lowers the incidence of congenital heart defects (CHDs) induced by pregestational diabetes. However, the molecular mechanisms underlying the beneficial effects of maternal exercise remain unclear. The present study aimed to identify circular RNA (circRNA), microRNA (miRNA) and mRNA networks that are regulated by maternal exercise in fetal hearts of pregestational diabetes. METHODS: Pregestational diabetes was induced in adult C57BL/6 female mice by streptozotocin. The expression profiles of circRNAs, miRNAs and mRNAs in E10.5 fetal hearts of offspring of control and diabetic mothers with or without exercise were analyzed using next generation sequencing. circRNA-miRNA-mRNA networks in fetal hearts were mapped and key candidate transcripts were verified by qPCR analysis. RESULTS: Pregestational diabetes dysregulated the expression of 206 circRNAs, 66 miRNAs and 391 mRNAs in fetal hearts. Maternal exercise differentially regulated 188 circRNAs, 57 miRNAs and 506 mRNAs in fetal hearts of offspring of pregestational diabetes. A total of 5 circRNAs, 12 miRNAs, and 28 mRNAs were incorporated into a final maternal exercise-associated regulatory network in fetal hearts of offspring of maternal diabetes. Notably, maternal exercise normalized the dysregulated circ_0003226/circ_0015638/miR-351-5p and circ_0002768/miR-3102-3p.2-3p pairs in fetal hearts of pregestational diabetes. CONCLUSION: Maternal exercise reverses the dysregulated circ_0003226/circ_0015638/miR-351-5p and circ_0002768/miR-3102-3p.2-3p pairs, and partially normalizes circRNA, miRNA, and mRNA expression profiles in fetal hearts of pregestational diabetes. These findings shed new light on the potential mechanisms of the beneficial effects of maternal exercise on the developing heart in diabetic pregnancies.


Assuntos
Diabetes Gestacional , MicroRNAs , Humanos , Gravidez , Animais , Camundongos , Feminino , MicroRNAs/genética , RNA Circular/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Camundongos Endogâmicos C57BL , Diabetes Gestacional/genética , Coração Fetal/metabolismo , Redes Reguladoras de Genes
6.
J Pineal Res ; 73(3): e12821, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35941749

RESUMO

Insufficient oxygen supply (hypoxia) during fetal development leads to cardiac remodeling and a predisposition to cardiovascular disease in later life. Previous work has shown hypoxia causes oxidative stress in the fetal heart and alters the activity and expression of mitochondrial proteins in a sex-dependent manner. However, the functional effects of these modifications on mitochondrial respiration remain unknown. Furthermore, while maternal antioxidant treatments are emerging as a promising new strategy to protect the hypoxic fetus, whether these treatments convey similar protection to cardiac mitochondria in the male or female fetus has not been investigated. Therefore, using an established rat model, we measured the sex-dependent effects of gestational hypoxia and maternal melatonin treatment on fetal cardiac mitochondrial respiration, reactive oxygen species (ROS) production, and lipid peroxidation. Pregnant Wistar rats were subjected to normoxia or hypoxia (13% oxygen) during gestational days (GDs) 6-20 (term ~22 days) with or without melatonin treatment (5 µg/ml in maternal drinking water). On GD 20, mitochondrial aerobic respiration and H2 O2 production were measured in fetal heart tissue, together with lipid peroxidation and citrate synthase (CS) activity. Gestational hypoxia reduced maternal body weight gain (p < .01) and increased placental weight (p < .05) but had no effect on fetal weight or litter size. Cardiac mitochondria from male but not female fetuses of hypoxic pregnancy had reduced respiratory capacity at Complex II (CII) (p < .05), and an increase in H2 O2 production/O2 consumption (p < .05) without any changes in lipid peroxidation. CS activity was also unchanged in both sexes. Despite maternal melatonin treatment increasing maternal and fetal plasma melatonin concentration (p < .001), melatonin treatment had no effect on any of the mitochondrial parameters investigated. To conclude, we show that gestational hypoxia leads to ROS generation from the mitochondrial electron transport chain and affects fetal cardiac mitochondrial respiration in a sex-dependent manner. We also show that maternal melatonin treatment had no effect on these relationships, which has implications for the development of future therapies for hypoxic pregnancies.


Assuntos
Melatonina , Animais , Feminino , Coração Fetal/metabolismo , Hipóxia/metabolismo , Masculino , Melatonina/metabolismo , Melatonina/farmacologia , Mitocôndrias Cardíacas/metabolismo , Estresse Oxidativo , Oxigênio/metabolismo , Oxigênio/farmacologia , Placenta , Gravidez , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
7.
Sci Rep ; 12(1): 1111, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35064159

RESUMO

Preeclampsia is a cardiovascular pregnancy complication characterised by new onset hypertension and organ damage or intrauterine growth restriction. It is one of the leading causes of maternal and fetal mortality in pregnancy globally. Short of pre-term delivery of the fetus and placenta, treatment options are limited. Consequently, preeclampsia leads to increased cardiovascular disease risk in both mothers and offspring later in life. Here we aim to examine the impact of the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia on the maternal cardiovascular system, placental and fetal heart metabolism. The surgical RUPP model was induced in pregnant rats by applying silver clips around the aorta and uterine arteries on gestational day 14, resulting in ~ 40% uterine blood flow reduction. The experiment was terminated on gestational day 19 and metabolomic profile of placentae, maternal and fetal hearts analysed using high-resolution 1H NMR spectroscopy. Impairment of uterine perfusion in RUPP rats caused placental and cardiac hypoxia and a series of metabolic adaptations: altered energetics, carbohydrate, lipid and amino acid metabolism of placentae and maternal hearts. Comparatively, the fetal metabolic phenotype was mildly affected. Nevertheless, long-term effects of these changes in both mothers and the offspring should be investigated further in the future.


Assuntos
Hipóxia/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Útero/irrigação sanguínea , Animais , Pressão Sanguínea/fisiologia , Simulação por Computador , Modelos Animais de Doenças , Feminino , Coração Fetal/metabolismo , Humanos , Hipóxia/fisiopatologia , Metabolômica , Modelos Biológicos , Placenta/irrigação sanguínea , Circulação Placentária/fisiologia , Pré-Eclâmpsia/fisiopatologia , Gravidez , Espectroscopia de Prótons por Ressonância Magnética , Ratos , Útero/fisiologia
8.
Am J Physiol Regul Integr Comp Physiol ; 322(1): R83-R98, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34851727

RESUMO

Previous studies in our laboratory have suggested that the increase in stillbirth in pregnancies complicated by chronic maternal stress or hypercortisolemia is associated with cardiac dysfunction in late stages of labor and delivery. Transcriptomics analysis of the overly represented differentially expressed genes in the fetal heart of hypercortisolemic ewes indicated involvement of mitochondrial function. Sodium dichloroacetate (DCA) has been used to improve mitochondrial function in several disease states. We hypothesized that administration of DCA to laboring ewes would improve both cardiac mitochondrial activity and cardiac function in their fetuses. Four groups of ewes and their fetuses were studied: control, cortisol-infused (1 g/kg/day from 115 to term; CORT), DCA-treated (over 24 h), and DCA + CORT-treated; oxytocin was delivered starting 48 h before the DCA treatment. DCA significantly decreased cardiac lactate, alanine, and glucose/glucose-6-phosphate and increased acetylcarnitine/isobutyryl-carnitine. DCA increased mitochondrial activity, increasing oxidative phosphorylation (PCI, PCI + II) per tissue weight or per unit of citrate synthase. DCA also decreased the duration of the QRS, attenuating the prolongation of the QRS observed in CORT fetuses. The effect to reduce QRS duration with DCA treatment correlated with increased glycerophosphocholine and serine and decreased phosphorylcholine after DCA treatment. There were negative correlations of acetylcarnitine/isobutyryl-carnitine to both heart rate (HR) and mean arterial pressure (MAP). These results suggest that improvements in mitochondrial respiration with DCA produced changes in the cardiac lipid metabolism that favor improved conduction in the heart. DCA may therefore be an effective treatment of fetal cardiac metabolic disturbances in labor that can contribute to impairments of fetal cardiac conduction.


Assuntos
Síndrome de Cushing/tratamento farmacológico , Ácido Dicloroacético/farmacologia , Metabolismo Energético/efeitos dos fármacos , Sofrimento Fetal/prevenção & controle , Coração Fetal/efeitos dos fármacos , Frequência Cardíaca Fetal/efeitos dos fármacos , Metaboloma , Mitocôndrias Cardíacas/efeitos dos fármacos , Animais , Síndrome de Cushing/induzido quimicamente , Síndrome de Cushing/metabolismo , Síndrome de Cushing/fisiopatologia , Modelos Animais de Doenças , Feminino , Sofrimento Fetal/induzido quimicamente , Sofrimento Fetal/metabolismo , Sofrimento Fetal/fisiopatologia , Coração Fetal/metabolismo , Coração Fetal/fisiopatologia , Hidrocortisona , Trabalho de Parto , Metabolismo dos Lipídeos/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Gravidez , Carneiro Doméstico
9.
J Clin Endocrinol Metab ; 106(11): 3295-3311, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34245263

RESUMO

CONTEXT: Intrauterine growth restriction (IUGR) is an immediate outcome of an adverse womb environment, exposing newborns to developing cardiometabolic disorders later in life. OBJECTIVE: This study investigates the cardiac metabolic consequences and underlying mechanism of energy expenditure in developing fetuses under conditions of IUGR. METHODS: Using an animal model of IUGR characterized by uteroplacental vascular insufficiency, mitochondrial function, gene profiling, lipidomic analysis, and transcriptional assay were determined in fetal cardiac tissue and cardiomyocytes. RESULTS: IUGR fetuses exhibited an upregulation of key genes associated with fatty acid breakdown and ß-oxidation (Acadvl, Acadl, Acaa2), and mitochondrial carnitine shuttle (Cpt1a, Cpt2), instigating a metabolic gene reprogramming in the heart. Induction of Ech1, Acox1, Acox3, Acsl1, and Pex11a indicated a coordinated interplay with peroxisomal ß-oxidation and biogenesis mainly observed in females, suggesting sexual dimorphism in peroxisomal activation. Concurring with the sex-related changes, mitochondrial respiration rates were stronger in IUGR female fetal cardiomyocytes, accounting for enhanced adenosine 5'-triphosphate production. Mitochondrial biogenesis was induced in fetal hearts with elevated expression of Ppargc1a transcript specifically in IUGR females. Lipidomic analysis identified the accumulation of arachidonic, eicosapentaenoic, and docosapentaenoic polyunsaturated long-chain fatty acids (LCFAs) in IUGR fetal hearts, which leads to nuclear receptor peroxisome proliferator-activated receptor α (PPARα) transcriptional activation in cardiomyocytes. Also, the enrichment of H3K27ac chromatin marks to PPARα-responsive metabolic genes in IUGR fetal hearts outlines an epigenetic control in the early metabolic energy switch. CONCLUSION: This study describes a premature and sex-related remodeling of cardiac metabolism in response to an unfavorable intrauterine environment, with specific LCFAs that may serve as predictive effectors leading to IUGR.


Assuntos
Metabolismo Energético , Ácidos Graxos/metabolismo , Retardo do Crescimento Fetal/patologia , Coração Fetal/patologia , Mitocôndrias/patologia , Miócitos Cardíacos/patologia , Animais , Animais Recém-Nascidos , Feminino , Retardo do Crescimento Fetal/metabolismo , Coração Fetal/metabolismo , Masculino , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Oxirredução , Ratos Sprague-Dawley , Fatores Sexuais
11.
J Assist Reprod Genet ; 38(8): 2139-2149, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34009631

RESUMO

PURPOSE: Is the presence of cytoplasmic strings (CS) in human blastocysts associated with the probability of clinical pregnancy with fetal heart (CPFH) after transfer. METHODS: This case-control study involved 300 single blastocyst transfers. 150 of these resulted in a CPFH (cases) while 150 did not (controls). All embryos were cultured in Embryoscope+ and AI software (IVY) was used to select the blastocyst with the highest score from the cohort for transfer. An embryologist, blind to the transfer outcome, recorded the CS number, location, and duration of their activity. RESULTS: There was a significant difference in the number of blastocysts that contained CS, with 97.3% of women's blastocysts resulting in +CPFH containing the CS compared to 88.7% of blastocysts in women who did not have a pregnancy (p = 0.007, OR; 4.67, CI 95% 1.5-14.2). CS appeared 2.4 h earlier in embryo development in the +CPFH group compared to their negative counterparts (p = 0.007). There was a significant difference in the average number of CS/blastocyst with a higher number being present in those that achieved a clinical pregnancy (mean: 6.2, SD 2.9) compared to those that did not (mean: 4.6, SD 3.0) (p ≤ 0.0001). There was a significant increase in the number of vesicles seen traveling along the CS with more seen in the blastocysts resulting in a +CPFH (mean: 4.3 SD 2.1) compared to those in the -CPFH group (mean: 3.1, SD 2.1). CONCLUSION: This study has shown that the presence of cytoplasmic strings in human blastocysts is associated with the probability of clinical pregnancy with fetal heart.


Assuntos
Blastocisto/metabolismo , Estruturas Citoplasmáticas/genética , Transferência Embrionária , Coração Fetal/ultraestrutura , Adulto , Blastocisto/patologia , Blastocisto/ultraestrutura , Estudos de Casos e Controles , Criopreservação , Citoplasma/genética , Citoplasma/ultraestrutura , Estruturas Citoplasmáticas/metabolismo , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Feminino , Coração Fetal/metabolismo , Coração Fetal/patologia , Humanos , Gravidez , Taxa de Gravidez
12.
Int J Mol Sci ; 22(5)2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33807511

RESUMO

3'-5' cyclic nucleotide phosphodiesterases (PDEs) are a large family of enzymes playing a fundamental role in the control of intracellular levels of cAMP and cGMP. Emerging evidence suggested an important role of phosphodiesterases in heart formation, but little is known about the expression of phosphodiesterases during cardiac development. In the present study, the pattern of expression and enzymatic activity of phosphodiesterases was investigated at different stages of heart formation. C57BL/6 mice were mated and embryos were collected from 14.5 to 18.5 days of development. Data obtained by qRT-PCR and Western blot analysis showed that seven different isoforms are expressed during heart development, and PDE1C, PDE2A, PDE4D, PDE5A and PDE8A are modulated from E14.5 to E18.5. In heart homogenates, the total cAMP and cGMP hydrolytic activity is constant at the evaluated times, and PDE4 accounts for the majority of the cAMP hydrolyzing ability and PDE2A accounts for cGMP hydrolysis. This study showed that a subset of PDEs is expressed in developing mice heart and some of them are modulated to maintain constant nucleotide phosphodiesterase activity in embryonic and fetal heart.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Coração Fetal/metabolismo , Diester Fosfórico Hidrolases/metabolismo , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Animais , AMP Cíclico , GMP Cíclico/metabolismo , Feminino , Coração Fetal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidores de Fosfodiesterase/farmacologia
13.
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
14.
Sci Rep ; 11(1): 6608, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758249

RESUMO

Cardiac development is a dynamic process, temporally and spatially. When disturbed, it leads to congenital cardiac anomalies that affect approximately 1% of live births. Genetic variants in several loci lead to anomalies, with the transcription factor NKX2-5 being one of the largest. However, there are also non-genetic factors that influence cardiac malformations. We examined the hypothesis that hyperoxia may be beneficial and can rescue genetic cardiac anomalies induced by an Nkx2-5 mutation. Intermittent mild hyperoxia (40% PO2) was applied for 10 h per day to normal wild-type female mice mated with heterozygous Nkx2-5 mutant males from gestational day 8.5 to birth. Hyperoxia therapy reduced excessive trabeculation in Nkx2-5 mutant mice compared to normoxic conditions (ratio of trabecular layer relative to compact layer area, normoxia 1.84 ± 0.07 vs. hyperoxia 1.51 ± 0.04) and frequency of muscular ventricular septal defects per heart (1.53 ± 0.32 vs. 0.68 ± 0.15); however, the incidence of membranous ventricular septal defects in Nkx2-5 mutant hearts was not changed. Nkx2-5 mutant embryonic hearts showed defective coronary vessel organization, which was improved by intermittent mild hyperoxia. The results of our study showed that mild gestational hyperoxia therapy rescued genetic cardiac malformation induced by Nkx2-5 mutation in part.


Assuntos
Coração Fetal/embriologia , Comunicação Interventricular/terapia , Oxigenoterapia Hiperbárica/métodos , Animais , Feminino , Coração Fetal/anormalidades , Coração Fetal/metabolismo , Comunicação Interventricular/genética , Proteína Homeobox Nkx-2.5/genética , Camundongos , Mutação
15.
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
16.
Pediatr Res ; 89(3): 456-463, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32674111

RESUMO

BACKGROUND: Mitochondrial permeability transition pore (mPTP) closure triggers cardiomyocyte differentiation during development while pathological opening causes cell death during myocardial ischemia-reperfusion and heart failure. Ubiquinone modulates the mPTP; however, little is known about its mechanistic role in health and disease. We previously found excessive proton leak in newborn Fmr1 KO mouse forebrain caused by ubiquinone deficiency and increased open mPTP probability. Because of the physiological differences between the heart and brain during maturation, we hypothesized that developing Fmr1 KO cardiomyocyte mitochondria would demonstrate dissimilar features. METHODS: Newborn male Fmr1 KO mice and controls were assessed. Respiratory chain enzyme activity, ubiquinone content, proton leak, and oxygen consumption were measured in cardiomyocyte mitochondria. Cardiac function was evaluated via echocardiography. RESULTS: In contrast to controls, Fmr1 KO cardiomyocyte mitochondria demonstrated increased ubiquinone content and decreased proton leak. Leak was cyclosporine (CsA)-sensitive in controls and CsA-insensitive in Fmr1 KOs. There was no difference in absolute mitochondrial respiration or cardiac function between strains. CONCLUSION: These findings establish the newborn Fmr1 KO mouse as a novel model of excess ubiquinone and closed mPTP in the developing heart. Such a model may help provide insight into the biology of cardiac development and pathophysiology of neonatal heart failure. IMPACT: Ubiquinone is in excess and the mPTP is closed in the developing FXS heart. Strengthens evidence of open mPTP probability in the normally developing postnatal murine heart and provides new evidence for premature closure of the mPTP in Fmr1 mutants. Establishes a novel model of excess CoQ and a closed pore in the developing heart. Such a model will be a valuable tool used to better understand the role of ubiquinone and the mPTP in the neonatal heart in health and disease.


Assuntos
Modelos Animais de Doenças , Coração Fetal/metabolismo , Proteína do X Frágil de Retardo Mental/genética , Síndrome do Cromossomo X Frágil/metabolismo , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Ubiquinona/metabolismo , Animais , Atractilosídeo/análogos & derivados , Atractilosídeo/farmacologia , Ciclosporina/farmacologia , Transporte de Elétrons , Síndrome do Cromossomo X Frágil/genética , Guanosina Difosfato/farmacologia , Masculino , Camundongos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Consumo de Oxigênio , Força Próton-Motriz , Método Simples-Cego , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia
17.
Curr Mol Med ; 21(2): 165-175, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32520686

RESUMO

BACKGROUND: Severe fetal anemias can cause high output cardiac failure. Mitochondria are key regulators of cardiac function. However, the effects of an early phase of fetal anemia on the fetal heart and cardiac mitochondrial function are not known. OBJECTIVE: The aim of this study is to compare mitochondrial function and cardiac biochemical alterations in the fetal cardiac tissue between anemic and non-anemic fetuses. MATERIALS AND METHODS: A cross-sectional study was conducted in Fetuses affected by Hb Bart's disease (n=18) and non-anemic fetuses (n=10) at 17-20 weeks. Echocardiograms had been carried out in all cases to assess prenatal cardiac function. Cardiac tissues were collected after pregnancy termination for the determination of cardiac iron accumulation, mitochondrial function, including mitochondrial ROS production, mitochondrial depolarization and mitochondrial swelling, mitochondrial dynamics, inflammation, and apoptosis. RESULTS: Prenatal cardiac function evaluated by ultrasound was comparable between the Hb Bart's and non-anemic groups. In Bart's group, the levels of cardiac mitochondrial depolarization and swelling, and the TNF-α level were significantly higher, compared to the non-anemic group. On the contrary, anti-inflammatory (IL-10) levels were significantly lower in the Hb Bart's group. Additionally, active caspase-3 and Bcl-2 expression were also significantly higher (P= 0.001, P=0.035) in Bart's group. The mitochondrial fission protein expression, including p-DRP1/total DRP1, was significantly higher in Bart's group. However, there was no difference in cardiac iron accumulation levels between these two groups. CONCLUSION: Despite equivalent prenatal cardiac function and comparable cardiac iron accumulation in the Bart's and non-anemic groups, fetal anemia is significantly associated with cardiac mitochondrial dysfunction, increased mitochondrial fission, and increased inflammation and apoptosis. These findings indicate that an early phase of fetal anemia without cardiac iron overload can lead to cardiac mitochondrial dysfunction in fetuses with Hb Bart's.


Assuntos
Anemia/complicações , Doenças Fetais/patologia , Coração Fetal/patologia , Insuficiência Cardíaca/patologia , Hemoglobinas Anormais/metabolismo , Mitocôndrias Cardíacas/patologia , Anemia/diagnóstico por imagem , Anemia/patologia , Apoptose , Estudos de Casos e Controles , Estudos Transversais , Feminino , Doenças Fetais/diagnóstico por imagem , Doenças Fetais/etiologia , Coração Fetal/diagnóstico por imagem , Coração Fetal/metabolismo , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/etiologia , Hemoglobinas Anormais/análise , Humanos , Mediadores da Inflamação/metabolismo , Ferro/metabolismo , Mitocôndrias Cardíacas/metabolismo , Estresse Oxidativo , Gravidez , Ultrassonografia Pré-Natal
18.
J Cardiovasc Pharmacol ; 77(1): 94-99, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33136763

RESUMO

ABSTRACT: Embryonic epicardial cells make an important contribution to cardiac development. However, their proliferation mechanism is still unclear. Epicardial cells from E12.5 fetal hearts were used in our study. Agrin was used to treat these cells. The expression of Aurora B, Ki67, and pH3 was measured by quantitative reverse transcription-polymerase chain reaction and immunofluorescence. The proportion of cells in G1/S/G2 phase was determined by flow cytometry. The results showed that agrin significantly increased the expression of ki67, pH3, and Aurora B in epicardial cells. Flow cytometry results showed that agrin significantly increased the proportion of epicardial cells in S phase. However, blocking yes-associated protein significantly downregulated the levels of ki67, pH3, and Aurora B and the proportion of epicardial cells in S phase. Thus, our results suggest that agrin may promote the proliferation of epicardial cells by regulating the yes-associated protein activity. This may be useful in exploring heart development mechanisms and preventing congenital heart disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Agrina/farmacologia , Proliferação de Células/efeitos dos fármacos , Coração Fetal/efeitos dos fármacos , Pericárdio/efeitos dos fármacos , Animais , Aurora Quinase B/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Coração Fetal/metabolismo , Histonas/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Pericárdio/metabolismo , Fosforilação , Proteínas de Sinalização YAP
19.
Hypertens Pregnancy ; 39(4): 429-443, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32791955

RESUMO

OBJECTIVE: This study aimed to investigate the effects of melatonin on cardiac oxidative stress and apoptosis in the fetal heart in RUPP rats. METHODS: The fetal heart samples were obtained from melatonin administrated RUPP rats. RESULTS: Our results indicate that preeclampsia exacerbated by melatonin deficiency triggers hypoxic conditions, both mis/un-folded protein response, oxidative stress-induced DNA damage and apoptosis. Melatonin treatment provided significant therapeutic effects on fetal hearts via regulating all these stress response at cellular and molecular levels. CONCLUSION: Melatonin may be considered as a potential molecule for development of preventive strategies to reduce the PE induced risk of cardiovascular diseases in offspring.


Assuntos
Apoptose/efeitos dos fármacos , Coração Fetal/efeitos dos fármacos , Melatonina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Pressão Sanguínea/fisiologia , Feminino , Coração Fetal/metabolismo , Pinealectomia , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Ratos , Ratos Wistar , Útero/irrigação sanguínea
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...