Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Birth Defects Res ; 114(17): 1101-1111, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36114760

RESUMO

BACKGROUND: Environmental factors may influence the development of tetralogy of Fallot (TOF), and DNA methylation patterns may reveal specific chemical signatures of perturbations during cardiac development. We investigated whether blood and buccal cells could be viable surrogates for myocardium. METHODS: We measured epigenome-wide DNA methylation at 866,895 5'-cytosine-phosphate-guanine-3' (CpG) sites in blood (n=3), buccal cells (n=3), and right ventricular myocardium (n=4) collected from infants with TOF and compared the percent of differentially methylated CpG sites across tissue types. Gene-specific DNA methylation profiles were also analyzed for ten representative genes associated with heart development. Welch's ANOVAs compared general methylation between tissue types. RESULTS: Comparison of DNA methylation profiles across blood, buccal, and myocardium suggested myocardium and buccal samples were most similar, differing in DNA methylation at only 1.3% (11,386) of CpG sites whereas myocardium and blood were most dissimilar, having 146,857 statistically dissimilar methylated CpG sites (~17% dissimilarity; adjusted p < 0.01 for each site). Buccal swabs were significantly more variable (p < .001) than either blood or myocardial samples. In gene-specific analyses, SCO2, GATA4, NOTCH4, WNT7A, and DKK2 showed conserved DNA methylation profiles across tissue types, while HAND1, JAG1, NKX2-5, TBX5 and TBX20 showed more distinctive tissue-specific patterns of DNA methylation. CONCLUSIONS: Compared with blood, buccal tissue more closely mirrors the myocardial methylome, with >10-fold similarity. Nevertheless, both buccal and blood tissue capture highly conserved DNA methylation patterns at specific genetic loci related to cardiac development. Buccal cheek swabs may be a useful surrogate tissue type for future investigations of TOF-specific epigenetic profiles.


Assuntos
Metilação de DNA , Tetralogia de Fallot , Citosina , Metilação de DNA/genética , Guanina , Humanos , Lactente , Mucosa Bucal , Fosfatos , Tetralogia de Fallot/genética
2.
Alcohol Clin Exp Res ; 46(8): 1433-1448, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35692084

RESUMO

BACKGROUND: Excessive alcohol consumption during pregnancy is associated with high risk of congenital heart defects, but it is unclear how alcohol specifically affects heart development during the acute aftermath of a maternal binge drinking episode. We hypothesize that administration of a single maternal binge dose of alcohol to pregnant mice at embryonic day 9.5 (E9.5) causes perturbations in the expression patterns of specific genes in the developing heart in the acute period (1-3 days) following the binge episode. To test this hypothesis and identify strong candidate ethanol-sensitive target genes of interest, we adapted a mouse binge alcohol model that is associated with a high incidence of congenital heart defects as described below. METHODS/RESULTS: Pregnant mice were administered a single dose of alcohol (2.5 g/kg in saline) or control (saline alone) via oral gavage. To evaluate the impact of maternal binge alcohol on cardiac gene expression profiles, we isolated embryonic hearts from both groups (n = 5/group) at 24, 48, and 72 h post-gavage for transcriptomic analyses. RNA was extracted and evaluated using quantitative RNA-sequencing (RNA-Seq) methods. To identify a cohort of binge-altered cardiac genes, we set the threshold for change at >2.0-fold difference with adjusted p < 0.05 versus control.  RNA-Seq analysis of cardiac gene expression revealed that of the 17 genes that were altered within the first 48 h post-binge, with the largest category consisting of transcription factors (Alx1, Alx4, HoxB7, HoxD8, and Runx2), followed by signaling molecules (Adamts18, Dkk2, Rtl1, and Wnt7a). Furthermore, multiple comparative and pathway analyses suggested that several of the candidate genes identified through differential RNA-Seq analysis may interact through certain common pathways. To investigate this further, we performed gene-specific qPCR analyses for three representative candidate targets: Runx2, Wnt7a, and Mlxipl. Notably, only Wnt7a showed significantly (p < 0.05) decreased expression in response to maternal binge alcohol in the qPCR assays. CONCLUSIONS: These findings identify Wnt7a and a short list of potential other candidate genes and pathways for further study, which could provide mechanistic insights into how maternal binge alcohol consumption produces congenital cardiac malformations.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas , Cardiopatias Congênitas , Consumo de Bebidas Alcoólicas/genética , Animais , Consumo Excessivo de Bebidas Alcoólicas/genética , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Feminino , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/genética , Camundongos , Gravidez , RNA , Transcriptoma , Proteínas Wnt/genética
3.
Front Cardiovasc Med ; 8: 634774, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33898535

RESUMO

Antiretroviral therapy (ART) has significantly reduced the rate of mortality in HIV infected population, but people living with HIV (PLWH) show higher rates of cardiovascular disease (CVD). However, the effect of antiretroviral (ARV) drug treatment on cardiac cells is not clear. In this study, we explored the effect of ARV drugs in cardiomyocyte epigenetic remodeling. Primary cardiomyocytes were treated with a combination of four ARV drugs (ritonavir, abacavir, atazanavir, and lamivudine), and epigenetic changes were examined. Our data suggest that ARV drugs treatment significantly reduces acetylation at H3K9 and H3K27 and promotes methylation at H3K9 and H3K27, which are histone marks for gene expression activation and gene repression, respectively. Besides, ARV drugs treatment causes pathological changes in the cell through increased production of reactive oxygen species (ROS) and cellular hypertrophy. Further, the expression of chromatin remodeling enzymes was monitored in cardiomyocytes treated with ARV drugs using PCR array. The PCR array data indicated that the expression of epigenetic enzymes was differentially regulated in the ARV drugs treated cardiomyocytes. Consistent with the PCR array result, SIRT1, SUV39H1, and EZH2 protein expression was significantly upregulated in ARV drugs treated cardiomyocytes. Furthermore, gene expression analysis of the heart tissue from HIV+ patients showed that the expression of SIRT1, SUV39H1, and EZH2 was up-regulated in patients with a history of ART. Additionally, we found that expression of SIRT1 can protect cardiomyocytes in presence of ARV drugs through reduction of cellular ROS and cellular hypertrophy. Our results reveal that ARV drugs modulate the epigenetic histone markers involved in gene expression, and play a critical role in histone deacetylation at H3K9 and H3K27 during cellular stress. This study may lead to development of novel therapeutic strategies for the treatment of CVD in PLWH.

4.
Diabetologia ; 64(1): 181-194, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33052459

RESUMO

AIMS/HYPOTHESIS: Melanocortin 4 receptor (MC4R) mutation is the most common cause of known monogenic obesity in humans. Unexpectedly, humans and rodents with MC4R deficiency do not develop hyperglycaemia despite chronic obesity and insulin resistance. To explain the underlying mechanisms for this phenotype, we determined the role of MC4R in glucose homeostasis in the presence and absence of obesity in mice. METHODS: We used global and hypothalamus-specific MC4R-deficient mice to investigate the brain regions that contribute to glucose homeostasis via MC4R. We performed oral, intraperitoneal and intravenous glucose tolerance tests in MC4R-deficient mice that were either obese or weight-matched to their littermate controls to define the role of MC4R in glucose regulation independently of changes in body weight. To identify the integrative pathways through which MC4R regulates glucose homeostasis, we measured renal and adrenal sympathetic nerve activity. We also evaluated glucose homeostasis in adrenaline (epinephrine)-deficient mice to investigate the role of adrenaline in mediating the effects of MC4R in glucose homeostasis. We employed a graded [13C6]glucose infusion procedure to quantify renal glucose reabsorption in MC4R-deficient mice. Finally, we measured the levels of renal glucose transporters in hypothalamus-specific MC4R-deficient mice and adrenaline-deficient mice using western blotting to ascertain the molecular mechanisms underlying MC4R control of glucose homeostasis. RESULTS: We found that obese and weight-matched MC4R-deficient mice exhibited improved glucose tolerance due to elevated glucosuria, not enhanced beta cell function. Moreover, MC4R deficiency selectively in the paraventricular nucleus of the hypothalamus (PVH) is responsible for reducing the renal threshold for glucose as measured by graded [13C6]glucose infusion technique. The MC4R deficiency suppressed renal sympathetic nerve activity by 50% in addition to decreasing circulating adrenaline and renal GLUT2 levels in mice, which contributed to the elevated glucosuria. We further report that adrenaline-deficient mice recapitulated the increased excretion of glucose in urine observed in the MC4R-deficient mice. Restoration of circulating adrenaline in both the MC4R- and adrenaline-deficient mice reversed their phenotype of improved glucose tolerance and elevated glucosuria, demonstrating the role of adrenaline in mediating the effects of MC4R on glucose reabsorption. CONCLUSIONS/INTERPRETATION: These findings define a previously unrecognised function of hypothalamic MC4R in glucose reabsorption mediated by adrenaline and renal GLUT2. Taken together, our findings indicate that elevated glucosuria due to low sympathetic tone explains why MC4R deficiency does not cause hyperglycaemia despite inducing obesity and insulin resistance. Graphical abstract.


Assuntos
Hexoses/metabolismo , Homeostase/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Bases de Schiff/metabolismo , Animais , Glicemia/metabolismo , Cruzamentos Genéticos , Epinefrina/deficiência , Epinefrina/fisiologia , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 2/fisiologia , Glicosúria/fisiopatologia , Hipotálamo/química , Insulina/sangue , Resistência à Insulina/fisiologia , Rim/inervação , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/fisiopatologia , Receptor Tipo 4 de Melanocortina/deficiência , Sistema Nervoso Simpático/fisiopatologia
5.
J Neurochem ; 152(3): 299-314, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31677273

RESUMO

Adrenal chromaffin cells release epinephrine (EPI) and norepinephrine (NE) into the bloodstream as part of the homeostatic response to situations like stress. Here we utilized EPI-deficient mice generated by knocking out (KO) the phenylethanolamine N-methyltransferase (Pnmt) gene. These Pnmt-KO mice were bred to homozygosis but displayed no major phenotype. The lack of EPI was partially compensated by an increase in NE, suggesting that EPI storage was optimized in adrenergic cells. Electron microscopy showed that despite the lack of EPI, chromaffin granules retain their shape and general appearance. This indicate that granules from adrenergic or noradrenergic cells preserve their characteristics even though they contain only NE. Acute insulin injection largely reduced the EPI content in wild-type animals, with a minimal reduction in NE, whereas there was only a partial reduction in NE content in Pnmt-KO mice. The analysis of exocytosis by amperometry revealed a reduction in the quantum size (-30%) and Imax (-21%) of granules in KO cells relative to the wild-type granules, indicating a lower affinity of NE for the granule matrix of adrenergic cells. As amperometry cannot distinguish between adrenergic or noradrenergic cells, it would suggest even a larger reduction in the affinity for the matrix. Therefore, our results demonstrate that adrenergic cells retain their structural characteristics despite the almost complete absence of EPI. Furthermore, the chromaffin granule matrix from adrenergic cells is optimized to accumulate EPI, with NE being a poor substitute. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Assuntos
Células Cromafins/metabolismo , Grânulos Cromafim/metabolismo , Epinefrina/metabolismo , Norepinefrina/metabolismo , Animais , Exocitose/fisiologia , Masculino , Camundongos , Camundongos Knockout , Feniletanolamina N-Metiltransferase/deficiência , Feniletanolamina N-Metiltransferase/genética
6.
Front Physiol ; 10: 713, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31354504

RESUMO

In this mini-review, we provide an overview of phenylethanolamine-N-methyl transferase (Pnmt)-derived cardiomyocytes (PdCMs), a recently discovered cardiomyocyte subpopulation. We discuss their anatomical localization, physiological characteristics, possible function, and future perspectives. Their unique distribution in the heart, electrical activity, Ca2+ transient properties, and potential role in localized adrenergic signaling are discussed.

7.
J Biol Chem ; 293(18): 6925-6941, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29540484

RESUMO

Cardiac energy demands during early embryonic periods are sufficiently met through glycolysis, but as development proceeds, the oxidative phosphorylation in mitochondria becomes increasingly vital. Adrenergic hormones are known to stimulate metabolism in adult mammals and are essential for embryonic development, but relatively little is known about their effects on metabolism in the embryonic heart. Here, we show that embryos lacking adrenergic stimulation have ∼10-fold less cardiac ATP compared with littermate controls. Despite this deficit in steady-state ATP, neither the rates of ATP formation nor degradation was affected in adrenergic hormone-deficient hearts, suggesting that ATP synthesis and hydrolysis mechanisms were fully operational. We thus hypothesized that adrenergic hormones stimulate metabolism of glucose to provide chemical substrates for oxidation in mitochondria. To test this hypothesis, we employed a metabolomics-based approach using LC/MS. Our results showed glucose 1-phosphate and glucose 6-phosphate concentrations were not significantly altered, but several downstream metabolites in both glycolytic and pentose-phosphate pathways were significantly lower compared with controls. Furthermore, we identified glyceraldehyde-3-phosphate dehydrogenase and glucose-6-phosphate dehydrogenase as key enzymes in those respective metabolic pathways whose activity was significantly (p < 0.05) and substantially (80 and 40%, respectively) lower in adrenergic hormone-deficient hearts. Addition of pyruvate and to a lesser extent ribose led to significant recovery of steady-state ATP concentrations. These results demonstrate that without adrenergic stimulation, glucose metabolism in the embryonic heart is severely impaired in multiple pathways, ultimately leading to insufficient metabolic substrate availability for successful transition to aerobic respiration needed for survival.


Assuntos
Coração/embriologia , Metabolômica , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Via de Pentose Fosfato , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Epinefrina/metabolismo , Feminino , Glucose/metabolismo , Glucose-6-Fosfato/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glucofosfatos/metabolismo , Gliceraldeído 3-Fosfato Desidrogenase (NADP+)/metabolismo , Glicólise , Hidrólise , Cetona Oxirredutases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Fosforilação , Gravidez
8.
Hum Mol Genet ; 27(9): 1533-1544, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29452352

RESUMO

Cardiac calsequestrin (Casq2) associates with the ryanodine receptor 2 channel in the junctional sarcoplasmic reticulum to regulate Ca2+ release into the cytoplasm. Patients carrying mutations in CASQ2 display low resting heart rates under basal conditions and stress-induced polymorphic ventricular tachycardia (CPVT). In this study, we generate and characterize novel conditional deletion and conditional rescue mouse models to test the influence of developmental programs on the heart rate and CPVT phenotypes. We also compare the requirements for Casq2 function in the cardiac conduction system (CCS) and in working cardiomyocytes. Our study shows that the CPVT phenotype is dependent upon concurrent loss of Casq2 function in both the CCS and in working cardiomyocytes. Accordingly, restoration of Casq2 in only the CCS prevents CPVT. In addition, occurrence of CPVT is independent of the developmental history of Casq2-deficiency. In contrast, resting heart rate depends upon Casq2 gene activity only in the CCS and upon developmental history. Finally, our data support a model where low basal heart rate is a significant risk factor for CPVT.


Assuntos
Calsequestrina/metabolismo , Taquicardia Ventricular/metabolismo , Tamoxifeno/farmacologia , Animais , Cálcio/metabolismo , Calsequestrina/genética , Feminino , Frequência Cardíaca/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Mutantes , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Taquicardia Ventricular/genética
9.
Biochem Biophys Res Commun ; 495(4): 2547-2552, 2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29288665

RESUMO

Mouse embryos that lack the ability to produce the adrenergic hormones, norepinephrine (NE) and epinephrine (EPI), due to disruption of the dopamine beta-hydroxylase (Dbh-/-) gene inevitably perish from heart failure during mid-gestation. Since adrenergic stimulation is well-known to enhance calcium signaling in developing as well as adult myocardium, and impairments in calcium signaling are typically associated with heart failure, we hypothesized that adrenergic-deficient embryonic hearts would display deficiencies in cardiac calcium signaling relative to adrenergic-competent controls at a developmental stage immediately preceding the onset of heart failure, which first appears beginning or shortly after mouse embryonic day 10.5 (E10.5). To test this hypothesis, we used ratiometric fluorescent calcium imaging techniques to measure cytosolic calcium transients, [Ca2+]i in isolated E10.5 mouse hearts. Our results show that spontaneous [Ca2+]i oscillations were intact and robustly responded to a variety of stimuli including extracellular calcium (5 mM), caffeine (5 mM), and NE (100 nM) in a manner that was indistinguishable from controls. Further, we show similar patterns of distribution (via immunofluorescent histochemical staining) and activity (via patch-clamp recording techniques) for the major voltage-gated plasma membrane calcium channel responsible for the L-type calcium current, ICa,L, in adrenergic-deficient and control embryonic cardiac cells. These results demonstrate that despite the absence of vital adrenergic hormones that consistently leads to embryonic lethality in vivo, intracellular and extracellular calcium signaling remain essentially intact and functional in embryonic mouse hearts through E10.5. These findings suggest that adrenergic stimulation is not required for the development of intracellular calcium oscillations or extracellular calcium signaling through ICa,L and that aberrant calcium signaling does not likely contribute to the onset of heart failure in this model.


Assuntos
Adrenérgicos/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Epinefrina/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Norepinefrina/metabolismo , Animais , Camundongos , Camundongos Knockout
10.
Sci Rep ; 7: 40687, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28084430

RESUMO

In the present study, channelrhodopsin 2 (ChR2) was specifically introduced into murine cells expressing the Phenylethanolamine n-methyltransferase (Pnmt) gene, which encodes for the enzyme responsible for conversion of noradrenaline to adrenaline. The new murine model enabled the identification of a distinctive class of Pnmt-expressing neuroendocrine cells and their descendants (i.e. Pnmt+ cell derived cells) within the heart. Here, we show that Pnmt+ cells predominantly localized to the left side of the adult heart. Remarkably, many of the Pnmt+ cells in the left atrium and ventricle appeared to be working cardiomyocytes based on their morphological appearance and functional properties. These Pnmt+ cell derived cardiomyocytes (PdCMs) are similar to conventional myocytes in morphological, electrical and contractile properties. By stimulating PdCMs selectively with blue light, we were able to control cardiac rhythm in the whole heart, isolated tissue preparations and single cardiomyocytes. Our new murine model effectively demonstrates functional dissection of cardiomyocyte subpopulations using optogenetics, and opens new frontiers of exploration into their physiological roles in normal heart function as well as their potential application for selective cardiac repair and regeneration strategies.


Assuntos
Frequência Cardíaca , Miocárdio/citologia , Miócitos Cardíacos/fisiologia , Optogenética , Feniletanolamina N-Metiltransferase/genética , Animais , Sinalização do Cálcio , Fenômenos Eletrofisiológicos , Imunofluorescência , Expressão Gênica , Genes Reporter , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Optogenética/métodos , Feniletanolamina N-Metiltransferase/metabolismo
11.
Nat Commun ; 7: 12604, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27576662

RESUMO

Vesicle fusion is executed via formation of an Ω-shaped structure (Ω-profile), followed by closure (kiss-and-run) or merging of the Ω-profile into the plasma membrane (full fusion). Although Ω-profile closure limits release but recycles vesicles economically, Ω-profile merging facilitates release but couples to classical endocytosis for recycling. Despite its crucial role in determining exocytosis/endocytosis modes, how Ω-profile merging is mediated is poorly understood in endocrine cells and neurons containing small ∼30-300 nm vesicles. Here, using confocal and super-resolution STED imaging, force measurements, pharmacology and gene knockout, we show that dynamic assembly of filamentous actin, involving ATP hydrolysis, N-WASP and formin, mediates Ω-profile merging by providing sufficient plasma membrane tension to shrink the Ω-profile in neuroendocrine chromaffin cells containing ∼300 nm vesicles. Actin-directed compounds also induce Ω-profile accumulation at lamprey synaptic active zones, suggesting that actin may mediate Ω-profile merging at synapses. These results uncover molecular and biophysical mechanisms underlying Ω-profile merging.


Assuntos
Actinas/metabolismo , Membrana Celular/metabolismo , Fusão de Membrana , Modelos Biológicos , Animais , Bovinos , Células Cromafins , Endocitose , Exocitose , Feminino , Técnicas de Inativação de Genes , Processamento de Imagem Assistida por Computador , Lampreias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia/métodos , Imagem Molecular/métodos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Cultura Primária de Células , Vesículas Secretórias/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo
12.
Am J Physiol Endocrinol Metab ; 308(5): E402-13, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516547

RESUMO

As development proceeds from the embryonic to fetal stages, cardiac energy demands increase substantially, and oxidative phosphorylation of ADP to ATP in mitochondria becomes vital. Relatively little, however, is known about the signaling mechanisms regulating the transition from anaerobic to aerobic metabolism that occurs during the embryonic period. The main objective of this study was to test the hypothesis that adrenergic hormones provide critical stimulation of energy metabolism during embryonic/fetal development. We examined ATP and ADP concentrations in mouse embryos lacking adrenergic hormones due to targeted disruption of the essential dopamine ß-hydroxylase (Dbh) gene. Embryonic ATP concentrations decreased dramatically, whereas ADP concentrations rose such that the ATP/ADP ratio in the adrenergic-deficient group was nearly 50-fold less than that found in littermate controls by embryonic day 11.5. We also found that cardiac extracellular acidification and oxygen consumption rates were significantly decreased, and mitochondria were significantly larger and more branched in adrenergic-deficient hearts. Notably, however, the mitochondria were intact with well-formed cristae, and there was no significant difference observed in mitochondrial membrane potential. Maternal administration of the adrenergic receptor agonists isoproterenol or l-phenylephrine significantly ameliorated the decreases in ATP observed in Dbh-/- embryos, suggesting that α- and ß-adrenergic receptors were effective modulators of ATP concentrations in mouse embryos in vivo. These data demonstrate that adrenergic hormones stimulate cardiac energy metabolism during a critical period of embryonic development.


Assuntos
Adrenérgicos/farmacologia , Doenças do Sistema Nervoso Autônomo/embriologia , Doenças do Sistema Nervoso Autônomo/fisiopatologia , Dopamina beta-Hidroxilase/deficiência , Dopamina beta-Hidroxilase/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Cardiopatias , Norepinefrina/deficiência , Adrenérgicos/metabolismo , Animais , Doenças do Sistema Nervoso Autônomo/genética , Doenças do Sistema Nervoso Autônomo/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Embrião de Mamíferos , Epinefrina/metabolismo , Epinefrina/farmacologia , Feminino , Coração/efeitos dos fármacos , Coração/embriologia , Coração/inervação , Cardiopatias/embriologia , Cardiopatias/genética , Cardiopatias/metabolismo , Isoproterenol/farmacologia , Troca Materno-Fetal/efeitos dos fármacos , Camundongos , Camundongos Knockout , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Gravidez , Regulação para Cima/efeitos dos fármacos
13.
J Pharmacol Exp Ther ; 351(2): 243-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25161169

RESUMO

It has been suggested that there is a link between epinephrine synthesis and the development of ß2-adrenoceptor-mediated effects, but it remains to be determined whether this development is triggered by epinephrine. The aim of this study was to characterize ß-adrenoceptor-mediated relaxation and facilitation of norepinephrine release in the aorta of phenylethanolamine-N-methyltransferase-knockout (Pnmt-KO) mice. Catecholamines were quantified by reverse-phase high-performance liquid chromatography-electrochemical detection. Aortic rings were mounted in a myograph to determine concentration-response curves to selective ß1- or ß2-adrenoceptor agonists in the absence or presence of selective ß1- or ß2-adrenoceptor antagonists. Aortic rings were also preincubated with [(3)H]norepinephrine to measure tritium overflow elicited by electrical stimulation in the presence of increasing concentrations of nonselective ß- or selective ß2-adrenoceptor agonists. ß2-Adrenoceptor protein density was evaluated by Western blotting and ß2-adrenoceptor localization by immunohistochemistry. Epinephrine is absent in Pnmt-KO mice. The potency and the maximal effect of the ß2-adrenoceptor agonist terbutaline were lower in Pnmt-KO than in wild-type (WT) mice. The selective ß2-adrenoceptor antagonist ICI 118,551 [(±)-erythro-(S*,S*)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride] antagonized the relaxation caused by terbutaline in WT but not in Pnmt-KO mice. Isoproterenol and terbutaline induced concentration-dependent increases in tritium overflow in WT mice only. ß2-Adrenoceptor protein density was decreased in membrane aorta homogenates of Pnmt-KO mice, and this finding was supported by immunofluorescence confocal microscopy. In conclusion, epinephrine is crucial for ß2-adrenoceptor-mediated vasodilation and facilitation of norepinephrine release. In the absence of epinephrine, ß2-adrenoceptor protein density was decreased in aorta cell membranes, thus potentially hindering its functional activity.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Aorta/efeitos dos fármacos , Epinefrina/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Vasodilatação/efeitos dos fármacos , Animais , Aorta/metabolismo , Catecolaminas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Estimulação Elétrica/métodos , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Norepinefrina/metabolismo , Feniletanolamina N-Metiltransferase/metabolismo , Terbutalina/farmacologia
14.
Am J Stem Cells ; 2(3): 137-54, 2013 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-24396707

RESUMO

Phenylethanolamine n-methyltransferase (Pnmt) catalyzes the conversion of norepinephrine into epinephrine, and thus serves as a marker of adrenergic cells. In adults, adrenergic cells are present in the adrenal medullae and the central and peripheral (sympathetic) nervous systems where they play key roles in stress responses and a variety of other functions. During early embryonic development, however, Pnmt first appears in the heart where it is associated with specialized myocytes in the pacemaking and conduction system. There is a transient surge in cardiac Pnmt expression beginning when the first myocardial contractions occur, before any nerve-like or neural crest cells appear in the heart. This early expression of Pnmt denotes a mesodermal origin of these "Instrinsic Cardiac Adrenergic" (ICA) cells. Interestingly, Pnmt+ cells are found in all four chambers of the developing heart, but by adult stages, are found primarily concentrated on the left side of the heart. This regionalized expression occurs in the left atrium and in specific regions of the left ventricle roughly corresponding to basal, mid, and apical sections. A second distinct population of Pnmt-expressing (Pnmt+) cells enters the embryonic heart from invading neural crest, and these "Neural Crest-Derived" (NCD) Pnmt+ cells appear to give rise to a subpopulation(s) of cardiac neurons. Pnmt expression thus serves as a marker not only for adrenergic cells, but also for precursor or "primer" cells destined to become specialized myocytes and neurons in the heart. This review discusses the distribution of Pnmt in the heart during development, including the types of cells where it is expressed, and their potential use for regenerative medicine therapies for cardiovascular disease.

15.
Mol Biotechnol ; 54(2): 350-60, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22706789

RESUMO

Adrenaline and noradrenaline are important neurotransmitter hormones that mediate physiological stress responses in adult mammals, and are essential for cardiovascular function during a critical period of embryonic/fetal development. In this study, we describe a novel mouse model system for identifying and characterizing adrenergic cells. Specifically, we generated a reporter mouse strain in which a nuclear-localized enhanced green fluorescent protein gene (nEGFP) was inserted into exon 1 of the gene encoding Phenylethanolamine n-methyltransferase (Pnmt), the enzyme responsible for production of adrenaline from noradrenaline. Our analysis demonstrates that this knock-in mutation effectively marks adrenergic cells in embryonic and adult mice. We see expression of nEGFP in Pnmt-expressing cells of the adrenal medulla in adult animals. We also note that nEGFP expression recapitulates the restricted expression of Pnmt in the embryonic heart. Finally, we show that nEGFP and Pnmt expressions are each induced in parallel during the in vitro differentiation of pluripotent mouse embryonic stem cells into beating cardiomyocytes. Thus, this new mouse genetic model should be useful for the identification and functional characterization of adrenergic cells in vitro and in vivo.


Assuntos
Medula Suprarrenal/metabolismo , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Medula Suprarrenal/citologia , Animais , Células-Tronco Embrionárias/metabolismo , Epinefrina/genética , Epinefrina/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Mutação , Miócitos Cardíacos/metabolismo , Norepinefrina/genética , Norepinefrina/metabolismo , Feniletanolamina N-Metiltransferase/genética , Feniletanolamina N-Metiltransferase/metabolismo , Células-Tronco Pluripotentes/metabolismo
16.
Physiol Genomics ; 44(19): 934-47, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22911456

RESUMO

Adrenergic hormones are essential for early heart development. To gain insight into understanding how these hormones influence heart development, we evaluated genomic expression changes in embryonic hearts from adrenergic-deficient and wild-type control mice. To perform this study, we used a mouse model with targeted disruption of the Dopamine ß-hydroxylase (Dbh) gene, whose product is responsible for enzymatic conversion of dopamine into norepinephrine. Embryos homozygous for the null allele (Dbh(-/-)) die from heart failure beginning as early as embryonic day 10.5 (E10.5). To assess underlying causes of heart failure, we isolated hearts from Dbh(-/-) and Dbh(+/+) embryos prior to manifestation of the phenotype and examined gene expression changes using genomic Affymetrix 430A 2.0 arrays, which enabled simultaneous evaluation of >22,000 genes. We found that only 22 expressed genes showed a significant twofold or greater change, representing ~0.1% of the total genes analyzed. More than half of these genes are associated with either metabolism (31%) or signal transduction (22%). Remarkably, several of the altered genes encode for proteins that are directly involved in retinoic acid (RA) biosynthesis and transport. Subsequent evaluation showed that RA concentrations were significantly elevated by an average of ~3-fold in adrenergic-deficient (Dbh(-/-)) embryos compared with controls, thereby suggesting that RA may be an important downstream mediator of adrenergic action during embryonic heart development.


Assuntos
Vias Biossintéticas/genética , Dopamina beta-Hidroxilase/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Insuficiência Cardíaca/genética , Coração/embriologia , Tretinoína/metabolismo , Animais , Dopamina beta-Hidroxilase/deficiência , Ecocardiografia , Regulação da Expressão Gênica no Desenvolvimento/genética , Insuficiência Cardíaca/enzimologia , Camundongos , Camundongos Knockout , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real
17.
Biochem Biophys Res Commun ; 423(3): 536-41, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22683331

RESUMO

To determine if adrenergic hormones play a critical role in the functional development of the cardiac pacemaking and conduction system, we employed a mouse model where adrenergic hormone production was blocked due to targeted disruption of the dopamine ß-hydroxylase (Dbh) gene. Immunofluorescent histochemical evaluation of the major gap junction protein, connexin 43, revealed that its expression was substantially decreased in adrenergic-deficient (Dbh-/-) relative to adrenergic-competent (Dbh+/+ and Dbh+/-) mouse hearts at embryonic day 10.5 (E10.5), whereas pacemaker and structural protein staining appeared similar. To evaluate cardiac electrical conduction in these hearts, we cultured them on microelectrode arrays (8×8, 200 µm apart). Our results show a significant slowing of atrioventricular conduction in adrenergic-deficient hearts compared to controls (31.4±6.4 vs. 15.4±1.7 ms, respectively, p<0.05). To determine if the absence of adrenergic hormones affected heart rate and rhythm, mouse hearts from adrenergic-competent and deficient embryos were cultured ex vivo at E10.5, and heart rates were measured before and after challenge with the ß-adrenergic receptor agonist, isoproterenol (0.5 µM). On average, all hearts showed increased heart rate responses following isoproterenol challenge, but a significant (p<0.05) 225% increase in the arrhythmic index (AI) was observed only in adrenergic-deficient hearts. These results show that adrenergic hormones may influence heart development by stimulating connexin 43 expression, facilitating atrioventricular conduction, and helping to maintain cardiac rhythm during a critical phase of embryonic development.


Assuntos
Arritmias Cardíacas/embriologia , Arritmias Cardíacas/genética , Dopamina beta-Hidroxilase/genética , Condutividade Elétrica , Coração/embriologia , Coração/fisiopatologia , Animais , Conexina 43/biossíntese , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/fisiopatologia , Frequência Cardíaca Fetal/genética , Frequência Cardíaca Fetal/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Camundongos , Camundongos Knockout
19.
PLoS One ; 6(7): e22811, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21818395

RESUMO

Adrenaline and noradrenaline are produced within the heart from neuronal and non-neuronal sources. These adrenergic hormones have profound effects on cardiovascular development and function, yet relatively little information is available about the specific tissue distribution of adrenergic cells within the adult heart. The purpose of the present study was to define the anatomical localization of cells derived from an adrenergic lineage within the adult heart. To accomplish this, we performed genetic fate-mapping experiments where mice with the cre-recombinase (Cre) gene inserted into the phenylethanolamine-n-methyltransferase (Pnmt) locus were cross-mated with homozygous Rosa26 reporter (R26R) mice. Because Pnmt serves as a marker gene for adrenergic cells, offspring from these matings express the ß-galactosidase (ßGAL) reporter gene in cells of an adrenergic lineage. ßGAL expression was found throughout the adult mouse heart, but was predominantly (89%) located in the left atrium (LA) and ventricle (LV) (p<0.001 compared to RA and RV), where many of these cells appeared to have cardiomyocyte-like morphological and structural characteristics. The staining pattern in the LA was diffuse, but the LV free wall displayed intermittent non-random staining that extended from the apex to the base of the heart, including heavy staining of the anterior papillary muscle along its perimeter. Three-dimensional computer-aided reconstruction of XGAL+ staining revealed distribution throughout the LA and LV, with specific finger-like projections apparent near the mid and apical regions of the LV free wall. These data indicate that adrenergic-derived cells display distinctive left-sided distribution patterns in the adult mouse heart.


Assuntos
Envelhecimento/metabolismo , Epinefrina/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Actinina/metabolismo , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/metabolismo , Animais , Ativação Enzimática , Imunofluorescência , Processamento de Imagem Assistida por Computador , Camundongos , Microscopia Confocal , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Feniletanolamina N-Metiltransferase/metabolismo , Coloração e Rotulagem , beta-Galactosidase/metabolismo
20.
BMC Biotechnol ; 11: 62, 2011 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-21635760

RESUMO

BACKGROUND: Gene therapy continues to hold great potential for treating many different types of disease and dysfunction. Safe and efficient techniques for gene transfer and expression in vivo are needed to enable gene therapeutic strategies to be effective in patients. Currently, the most commonly used methods employ replication-defective viral vectors for gene transfer, while physical gene transfer methods such as biolistic-mediated ("gene-gun") delivery to target tissues have not been as extensively explored. In the present study, we evaluated the efficacy of biolistic gene transfer techniques in vivo using non-invasive bioluminescent imaging (BLI) methods. RESULTS: Plasmid DNA carrying the firefly luciferase (LUC) reporter gene under the control of the human Cytomegalovirus (CMV) promoter/enhancer was transfected into mouse skin and liver using biolistic methods. The plasmids were coupled to gold microspheres (1 µm diameter) using different DNA Loading Ratios (DLRs), and "shot" into target tissues using a helium-driven gene gun. The optimal DLR was found to be in the range of 4-10. Bioluminescence was measured using an In Vivo Imaging System (IVIS-50) at various time-points following transfer. Biolistic gene transfer to mouse skin produced peak reporter gene expression one day after transfer. Expression remained detectable through four days, but declined to undetectable levels by six days following gene transfer. Maximum depth of tissue penetration following biolistic transfer to abdominal skin was 200-300 µm. Similarly, biolistic gene transfer to mouse liver in vivo also produced peak early expression followed by a decline over time. In contrast to skin, however, liver expression of the reporter gene was relatively stable 4-8 days post-biolistic gene transfer, and remained detectable for nearly two weeks. CONCLUSIONS: The use of bioluminescence imaging techniques enabled efficient evaluation of reporter gene expression in vivo. Our results demonstrate that different tissues show different expression kinetics following gene transfer of the same reporter plasmid to different mouse tissues in vivo. We evaluated superficial (skin) and abdominal organ (liver) targets, and found that reporter gene expression peaked within the first two days post-transfer in each case, but declined most rapidly in the skin (3-4 days) compared to liver (10-14 days). This information is essential for designing effective gene therapy strategies in different target tissues.


Assuntos
Biolística/métodos , Genes Reporter , Medições Luminescentes/métodos , Animais , Citomegalovirus/genética , Ouro/química , Humanos , Fígado/metabolismo , Luciferases de Vaga-Lume/genética , Camundongos , Plasmídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...