Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 325(4): H687-H701, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37566109

RESUMO

The ductus arteriosus (DA) is a vascular shunt that allows oxygenated blood to bypass the developing lungs in utero. Fetal DA patency requires vasodilatory signaling via the prostaglandin E2 (PGE2) receptor EP4. However, in humans and mice, disrupted PGE2-EP4 signaling in utero causes unexpected patency of the DA (PDA) after birth, suggesting another role for EP4 during development. We used EP4-knockout (KO) mice and acute versus chronic pharmacological approaches to investigate EP4 signaling in DA development and function. Expression analyses identified EP4 as the primary EP receptor in the DA from midgestation to term; inhibitor studies verified EP4 as the primary dilator during this period. Chronic antagonism recapitulated the EP4 KO phenotype and revealed a narrow developmental window when EP4 stimulation is required for postnatal DA closure. Myography studies indicate that despite reduced contractile properties, the EP4 KO DA maintains an intact oxygen response. In newborns, hyperoxia constricted the EP4 KO DA but survival was not improved, and permanent remodeling was disrupted. Vasomotion and increased nitric oxide (NO) sensitivity in the EP4 KO DA suggest incomplete DA development. Analysis of DA maturity markers confirmed a partially immature EP4 KO DA phenotype. Together, our data suggest that EP4 signaling in late gestation plays a key developmental role in establishing a functional term DA. When disrupted in EP4 KO mice, the postnatal DA exhibits signaling and contractile properties characteristic of an immature DA, including impairments in the first, muscular phase of DA closure, in addition to known abnormalities in the second permanent remodeling phase.NEW & NOTEWORTHY EP4 is the primary EP receptor in the ductus arteriosus (DA) and is critical during late gestation for its development and eventual closure. The "paradoxical" patent DA (PDA) phenotype of EP4-knockout mice arises from a combination of impaired contractile potential, altered signaling properties, and a failure to remodel associated with an underdeveloped immature vessel. These findings provide new mechanistic insights into women who receive NSAIDs to treat preterm labor, whose infants have unexplained PDA.


Assuntos
Permeabilidade do Canal Arterial , Canal Arterial , Camundongos , Animais , Recém-Nascido , Feminino , Gravidez , Humanos , Canal Arterial/metabolismo , Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Permeabilidade do Canal Arterial/genética , Camundongos Knockout
2.
Pediatr Res ; 84(3): 458-465, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29976969

RESUMO

BACKGROUND: Although studies involving preterm infants ≤34 weeks gestation report a decreased incidence of patent ductus arteriosus after antenatal betamethasone, studies involving younger gestation infants report conflicting results. METHODS: We used preterm baboons, mice, and humans (≤276/7 weeks gestation) to examine betamethasone's effects on ductus gene expression and constriction both in vitro and in vivo. RESULTS: In mice, betamethasone increased the sensitivity of the premature ductus to the contractile effects of oxygen without altering the effects of other contractile or vasodilatory stimuli. Betamethasone's effects on oxygen sensitivity could be eliminated by inhibiting endogenous prostaglandin/nitric oxide signaling. In mice and baboons, betamethasone increased the expression of several developmentally regulated genes that mediate oxygen-induced constriction (K+ channels) and inhibit vasodilator signaling (phosphodiesterases). In human infants, betamethasone increased the rate of ductus constriction at all gestational ages. However, in infants born ≤256/7 weeks gestation, betamethasone's contractile effects were only apparent when prostaglandin signaling was inhibited, whereas at 26-27 weeks gestation, betamethasone's contractile effects were apparent even in the absence of prostaglandin inhibitors. CONCLUSIONS: We speculate that betamethasone's contractile effects may be mediated through genes that are developmentally regulated. This could explain why betamethasone's effects vary according to the infant's developmental age at birth.


Assuntos
Betametasona/uso terapêutico , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Animais , Ecocardiografia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Recém-Nascido Prematuro , Exposição Materna , Camundongos , Oxigênio/metabolismo , Papio , Reação em Cadeia da Polimerase , Prostaglandinas/metabolismo
3.
Am J Physiol Heart Circ Physiol ; 311(3): H572-81, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27371685

RESUMO

Use of selective serotonin reuptake inhibitors (SSRIs) is common during pregnancy. Fetal exposure to SSRIs is associated with persistent pulmonary hypertension of the newborn (PPHN); however, a direct link between the two has yet to be established. Conversely, it is well known that PPHN can be caused by premature constriction of the ductus arteriosus (DA), a fetal vessel connecting the pulmonary and systemic circulations. We hypothesized that SSRIs could induce in utero DA constriction. Using isolated vessels and whole-animal models, we sought to determine the effects of two commonly prescribed SSRIs, fluoxetine and sertraline, on the fetal mouse DA. Cannulated vessel myography studies demonstrated that SSRIs caused concentration-dependent DA constriction and made vessels less sensitive to prostaglandin-induced dilation. Moreover, in vivo studies showed that SSRI-exposed mice had inappropriate DA constriction in utero. Taken together, these findings establish that SSRIs promote fetal DA constriction and provide a potential mechanism by which SSRIs could contribute to PPHN.


Assuntos
Canal Arterial/efeitos dos fármacos , Fluoxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sertralina/farmacologia , Vasoconstrição/efeitos dos fármacos , Animais , Aorta/metabolismo , Canal Arterial/metabolismo , Feminino , Imuno-Histoquímica , Camundongos , Miografia , Síndrome da Persistência do Padrão de Circulação Fetal , Gravidez , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Mol Cell Cardiol ; 69: 88-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24508561

RESUMO

Vascular smooth muscle cells (VSMCs) are derived from distinct embryonic origins. Vessels originating from differing smooth muscle cell populations have distinct vascular and pathological properties involving calcification, atherosclerosis, and structural defects such as aneurysm and coarctation. We hypothesized that domains within a single vessel, such as the aorta, vary in phenotype based on embryonic origin. Gene profiling and myographic analyses demonstrated that embryonic ascending and descending aortic domains exhibited distinct phenotypes. In vitro analyses demonstrated that VSMCs from each region were dissimilar in terms of cytoskeletal and migratory properties, and retention of different gene expression patterns. Using the same analysis, we found that these same two domains are indistinguishable in the adult vessel. Our data demonstrate that VSMCs from different embryonic origins are functionally distinct in the embryonic mouse, but converge to assume a common phenotype in the aorta of healthy adults. These findings have fundamental implications for aortic development, function and disease progression.


Assuntos
Aorta/embriologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Variação Genética , Músculo Liso Vascular/embriologia , Animais , Aorta/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Músculo Liso Vascular/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Physiol Genomics ; 46(13): 457-66, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24790087

RESUMO

Failure of the ductus arteriosus (DA) to close at birth can lead to serious complications. Conversely, certain profound congenital cardiac malformations require the DA to be patent until corrective surgery can be performed. In each instance, clinicians have a very limited repertoire of therapeutic options at their disposal - indomethacin or ibuprofen to close a patent DA (PDA) and prostaglandin E1 to maintain patency of the DA. Neither treatment is specific to the DA and both may have deleterious off-target effects. Therefore, more therapeutic options specifically targeted to the DA should be considered. We hypothesized the DA possesses a unique genetic signature that would set it apart from other vessels. A microarray was used to compare the genetic profiles of the murine DA and ascending aorta (AO). Over 4,000 genes were differentially expressed between these vessels including a subset of ion channel-related genes. Specifically, the alpha and beta subunits of large-conductance calcium-activated potassium (BKCa) channels are enriched in the DA. Gain- and loss-of-function studies showed inhibition of BKCa channels caused the DA to constrict, while activation caused DA relaxation even in the presence of O2. This study identifies subsets of genes that are enriched in the DA that may be used to develop DA-specific drugs. Ion channels that regulate DA tone, including BKCa channels, are promising targets. Specifically, BKCa channel agonists like NS1619 maintain DA patency even in the presence of O2 and may be clinically useful.


Assuntos
Canal Arterial/metabolismo , Transcriptoma , Grau de Desobstrução Vascular/genética , Animais , Permeabilidade do Canal Arterial/genética , Permeabilidade do Canal Arterial/metabolismo , Embrião de Mamíferos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Vasodilatação/genética
6.
Pediatr Res ; 76(3): 238-44, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24941212

RESUMO

BACKGROUND: We evaluated the clinical effectiveness of variable courses of paracetamol on patent ductus arteriosus (PDA) closure and examined its effect on the in vitro term and preterm murine ductus arteriosus (DA). METHODS: Neonates received one of the following three paracetamol regimens: short course of oral paracetamol (SCOP), long course of oral paracetamol (LCOP), and intravenous paracetamol (IVP) for 2-6 d. Pressure myography was used to examine changes in vasomotor tone of the preterm and term mouse DA in response to paracetamol or indomethacin. Their effect on prostaglandin synthesis by DA explants was measured by mass spectroscopy. RESULTS: Twenty-one preterm infants were included. No changes in PDA hemodynamics were seen in SCOP infants (n = 5). The PDA became less significant and eventually closed in six LCOP infants (n = 7). PDA closure was achieved in eight IVP infants (n = 9). On pressure myograph, paracetamol induced a concentration-dependent constriction of the term mouse DA, up to 30% of baseline (P < 0.01), but required >1 µmol/l. Indomethacin induced greater DA constriction and suppression of prostaglandin synthesis (P < 0.05). CONCLUSION: The clinical efficacy of paracetamol on PDA closure may depend on the duration of treatment and the mode of administration. Paracetamol is less potent than indomethacin for constriction of the mouse DA in vitro.


Assuntos
Acetaminofen/administração & dosagem , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Antagonistas de Prostaglandina/administração & dosagem , Vasoconstritores/administração & dosagem , Administração Intravenosa , Administração Oral , Animais , Pressão Sanguínea/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Canal Arterial/metabolismo , Canal Arterial/fisiopatologia , Canal Arterial/cirurgia , Permeabilidade do Canal Arterial/diagnóstico , Permeabilidade do Canal Arterial/metabolismo , Permeabilidade do Canal Arterial/fisiopatologia , Permeabilidade do Canal Arterial/cirurgia , Humanos , Indometacina/farmacologia , Recém-Nascido , Ligadura , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA