Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 444 Suppl 1: S308-S324, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29807017

RESUMEN

Carotid body glomus cells mediate essential reflex responses to arterial blood hypoxia. They are dopaminergic and secrete growth factors that support dopaminergic neurons, making the carotid body a potential source of patient-specific cells for Parkinson's disease therapy. Like adrenal chromaffin cells, which are also hypoxia-sensitive, glomus cells are neural crest-derived and require the transcription factors Ascl1 and Phox2b; otherwise, their development is little understood at the molecular level. Here, analysis in chicken and mouse reveals further striking molecular parallels, though also some differences, between glomus and adrenal chromaffin cell development. Moreover, histology has long suggested that glomus cell precursors are 'émigrés' from neighbouring ganglia/nerves, while multipotent nerve-associated glial cells are now known to make a significant contribution to the adrenal chromaffin cell population in the mouse. We present conditional genetic lineage-tracing data from mice supporting the hypothesis that progenitors expressing the glial marker proteolipid protein 1, presumably located in adjacent ganglia/nerves, also contribute to glomus cells. Finally, we resolve a paradox for the 'émigré' hypothesis in the chicken - where the nearest ganglion to the carotid body is the nodose, in which the satellite glia are neural crest-derived, but the neurons are almost entirely placode-derived - by fate-mapping putative nodose neuronal 'émigrés' to the neural crest.


Asunto(s)
Cuerpo Carotídeo/embriología , Células Cromafines/metabolismo , Pericitos/metabolismo , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Hipoxia de la Célula/fisiología , Embrión de Pollo , Pollos/metabolismo , Ratones , Ratones Noqueados , Proteína Proteolipídica de la Mielina/fisiología , Cresta Neural/metabolismo , Neuronas/metabolismo , Pericitos/fisiología , Factores de Transcripción/metabolismo
2.
Pediatr Res ; 84(3): 458-465, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29976969

RESUMEN

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.


Asunto(s)
Betametasona/uso terapéutico , Conducto Arterioso Permeable/tratamiento farmacológico , Conducto Arterial/efectos de los fármacos , Animales , Ecocardiografía , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Recien Nacido Prematuro , Exposición Materna , Ratones , Oxígeno/metabolismo , Papio , Reacción en Cadena de la Polimerasa , Prostaglandinas/metabolismo
3.
Am J Physiol Heart Circ Physiol ; 311(3): H572-81, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27371685

RESUMEN

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.


Asunto(s)
Conducto Arterial/efectos de los fármacos , Fluoxetina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sertralina/farmacología , Vasoconstricción/efectos de los fármacos , Animales , Aorta/metabolismo , Conducto Arterial/metabolismo , Femenino , Inmunohistoquímica , Ratones , Miografía , Síndrome de Circulación Fetal Persistente , Embarazo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Physiol Genomics ; 46(13): 457-66, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24790087

RESUMEN

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.


Asunto(s)
Conducto Arterial/metabolismo , Transcriptoma , Grado de Desobstrucción Vascular/genética , Animales , Conducto Arterioso Permeable/genética , Conducto Arterioso Permeable/metabolismo , Embrión de Mamíferos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Vasodilatación/genética
5.
Am J Physiol Heart Circ Physiol ; 307(5): H732-40, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24993047

RESUMEN

Sepsis is strongly associated with patency of the ductus arteriosus (PDA) in critically ill newborns. Inflammation and the aminoglycoside antibiotics used to treat neonatal sepsis cause smooth muscle relaxation, but their contribution to PDA is unknown. We examined whether: 1) lipopolysaccharide (LPS) or inflammatory cytokines cause relaxation of the ex vivo mouse DA; 2) the aminoglycosides gentamicin, tobramycin, or amikacin causes DA relaxation; and 3) newborn infants treated with aminoglycosides have an increased risk of symptomatic PDA (sPDA). Changes in fetal mouse DA tone were measured by pressure myography in response to LPS, TNF-α, IFN-γ, macrophage-inflammatory protein 2, IL-15, IL-13, CXC chemokine ligand 12, or three aminoglycosides. A clinical database of inborn patients of all gestations was analyzed for association between sPDA and aminoglycoside treatment. Contrary to expectation, neither LPS nor any of the inflammatory mediators caused DA relaxation. However, each of the aminoglycosides caused concentration-dependent vasodilation in term and preterm mouse DAs. Pretreatment with indomethacin and N-(G)-nitro-L-arginine methyl ester did not prevent gentamicin-induced DA relaxation. Gentamicin-exposed DAs developed less oxygen-induced constriction than unexposed DAs. Among 488,349 infants who met the study criteria, 40,472 (8.3%) had sPDA. Confounder-adjusted odds of sPDA were higher in gentamicin-exposed infants, <25 wk and >32 wk. Together, these findings suggest that factors other than inflammation contribute to PDA. Aminoglycoside-induced vasorelaxation and inhibition of oxygen-induced DA constriction support the paradox that antibiotic treatment of sepsis may contribute to DA relaxation. This association was also found in newborn infants, suggesting that antibiotic selection may be an important consideration in efforts to reduce sepsis-associated PDA.


Asunto(s)
Conducto Arterioso Permeable/fisiopatología , Conducto Arterial/efectos de los fármacos , Gentamicinas/farmacología , Sepsis/complicaciones , Vasodilatación , Animales , Quimiocina CXCL12/farmacología , Conducto Arterial/fisiopatología , Conducto Arterioso Permeable/etiología , Humanos , Técnicas In Vitro , Indometacina/farmacología , Recién Nacido , Interferón gamma/farmacología , Interleucinas/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Factor de Necrosis Tumoral alfa/farmacología
6.
Reproduction ; 148(6): 607-21, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25252651

RESUMEN

The mouse model has greatly contributed to understanding molecular mechanisms involved in the regulation of progesterone (P4) plus estrogen (E)-dependent blastocyst implantation process. However, little is known about contributory molecular mechanisms of the P4-only-dependent blastocyst implantation process that occurs in species such as hamsters, guineapigs, rabbits, pigs, rhesus monkeys, and perhaps humans. We used the hamster as a model of P4-only-dependent blastocyst implantation and carried out cross-species microarray (CSM) analyses to reveal differentially expressed genes at the blastocyst implantation site (BIS), in order to advance the understanding of molecular mechanisms of implantation. Upregulation of 112 genes and downregulation of 77 genes at the BIS were identified using a mouse microarray platform, while use of the human microarray revealed 62 up- and 38 down-regulated genes at the BIS. Excitingly, a sizable number of genes (30 up- and 11 down-regulated genes) were identified as a shared pool by both CSMs. Real-time RT-PCR and in situ hybridization validated the expression patterns of several up- and down-regulated genes identified by both CSMs at the hamster and mouse BIS to demonstrate the merit of CSM findings across species, in addition to revealing genes specific to hamsters. Functional annotation analysis found that genes involved in the spliceosome, proteasome, and ubiquination pathways are enriched at the hamster BIS, while genes associated with tight junction, SAPK/JNK signaling, and PPARα/RXRα signalings are repressed at the BIS. Overall, this study provides a pool of genes and evidence of their participation in up- and down-regulated cellular functions/pathways at the hamster BIS.


Asunto(s)
Implantación del Embrión/genética , Genes/genética , Mesocricetus/genética , Transcriptoma/genética , Animales , Cricetinae , Regulación hacia Abajo/genética , Femenino , Humanos , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de la Especie , Regulación hacia Arriba/genética
7.
Pediatr Res ; 76(3): 238-44, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24941212

RESUMEN

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.


Asunto(s)
Acetaminofén/administración & dosificación , Conducto Arterioso Permeable/tratamiento farmacológico , Conducto Arterial/efectos de los fármacos , Antagonistas de Prostaglandina/administración & dosificación , Vasoconstrictores/administración & dosificación , Administración Intravenosa , Administración Oral , Animales , Presión Sanguínea/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Conducto Arterial/metabolismo , Conducto Arterial/fisiopatología , Conducto Arterial/cirugía , Conducto Arterioso Permeable/diagnóstico , Conducto Arterioso Permeable/metabolismo , Conducto Arterioso Permeable/fisiopatología , Conducto Arterioso Permeable/cirugía , Humanos , Indometacina/farmacología , Recién Nacido , Ligadura , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
8.
J Mol Cell Cardiol ; 59: 86-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454087

RESUMEN

Persistent patency of the ductus arteriosus (PDA) is a common problem in preterm infants. The antacid cimetidine is a potent antagonist of the H2 histamine receptor but it also inhibits certain cytochrome P450 enzymes (CYPs), which may affect DA patency. We examined whether cimetidine contributes to PDA and is mediated by CYP inhibition rather than H2 blockade. Analysis of a clinical trial to prevent lung injury in premature infants revealed a significant association between cimetidine treatment and PDA. Cimetidine and ranitidine, both CYP inhibitors as well as H2 blockers, caused relaxation of the term and preterm mouse DA. CYP enzymes that are inhibited by cimetidine were expressed in DA subendothelial smooth muscle. The selective CYP3A inhibitor ketoconazole induced greater DA relaxation than cimetidine, whereas famotidine and other H2 antagonists with less CYP inhibitory effects caused less dilation. Histamine receptors were developmentally regulated and localized in DA smooth muscle. However, cimetidine caused DA relaxation in histamine-deficient mice, consistent with CYP inhibition, not H2 antagonism, as the mechanism for PDA. Oxygen-induced DA constriction was inhibited by both cimetidine and famotidine. These studies show that antacids and other compounds with CYP inhibitory properties pose a significant and previously unrecognized risk for PDA in critically ill newborn infants.


Asunto(s)
Cimetidina/efectos adversos , Sistema Enzimático del Citocromo P-450/metabolismo , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterioso Permeable/metabolismo , Antagonistas de los Receptores H2 de la Histamina/efectos adversos , Humanos , Inmunohistoquímica , Recién Nacido , Cetoconazol/efectos adversos , Reacción en Cadena de la Polimerasa , Ensayos Clínicos Controlados Aleatorios como Asunto , Ranitidina/efectos adversos , Receptores Histamínicos/metabolismo , Estudios Retrospectivos
9.
Reproduction ; 146(5): 419-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23929901

RESUMEN

Alkaline phosphatase (AP) activity has been demonstrated in the uterus of several species, but its importance in the uterus, in general and during pregnancy, is yet to be revealed. In this study, we focused on identifying AP isozyme types and their hormonal regulation, cell type, and event-specific expression and possible functions in the hamster uterus during the cycle and early pregnancy. Our RT-PCR and in situ hybridization studies demonstrated that among the known Akp2, Akp3, Akp5, and Akp6 murine AP isozyme genes, hamster uteri express only Akp2 and Akp6; both genes are co-expressed in luminal epithelial cells. Studies in cyclic and ovariectomized hamsters established that while progesterone (P4) is the major uterine Akp2 inducer, both P4 and estrogen are strong Akp6 regulators. Studies in preimplantation uteri showed induction of both genes and the activity of their encoded isozymes in luminal epithelial cells during uterine receptivity. However, at the beginning of implantation, Akp2 showed reduced expression in luminal epithelial cells surrounding the implanted embryo. By contrast, expression of Akp6 and its isozyme was maintained in luminal epithelial cells adjacent to, but not away from, the implanted embryo. Following implantation, stromal transformation to decidua was associated with induced expressions of only Akp2 and its isozyme. We next demonstrated that uterine APs dephosphorylate and detoxify endotoxin lipopolysaccharide at their sites of production and activity. Taken together, our findings suggest that uterine APs contribute to uterine receptivity, implantation, and decidualization in addition to their role in protection of the uterus and pregnancy against bacterial infection.


Asunto(s)
Fosfatasa Alcalina/biosíntesis , Decidua/enzimología , Implantación del Embrión , Inducción Enzimática , Lipopolisacáridos/metabolismo , Placentación , Útero/enzimología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Cricetinae , Decidua/citología , Decidua/inmunología , Decidua/fisiología , Endometrio/citología , Endometrio/enzimología , Endometrio/inmunología , Endometrio/fisiología , Infecciones por Escherichia coli/inmunología , Ciclo Estral , Femenino , Inmunidad Innata , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Lipopolisacáridos/toxicidad , Mesocricetus , Ovariectomía , Fosforilación , Embarazo , ARN Mensajero/metabolismo , Útero/citología , Útero/inmunología , Útero/fisiología
10.
Pediatr Res ; 72(2): 122-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22565502

RESUMEN

BACKGROUND: Increased oxygen tension at birth regulates physiologic events that are essential to postnatal survival, but the accompanying oxidative stress may also generate isoprostanes. We hypothesized that isoprostanes regulate ductus arteriosus (DA) function during postnatal vascular transition. METHODS: Isoprostanes were measured by gas chromatography-mass spectrometry. DA tone was assessed by pressure myography. Gene expression was measured by quantitative PCR. RESULTS: Oxygen exposure was associated with increased 8-iso-prostaglandin (PG)F2α in newborn mouse lungs. Both 8-iso-PGE2 and 8-iso-PGF2α induced concentration-dependent constriction of the isolated term DA, which was reversed by the thromboxane A2 (TxA2) receptor antagonist SQ29548. SQ29548 pretreatment unmasked an isoprostane-induced DA dilation mediated by the EP4 PG receptor. Exposure of the preterm DA to 8-iso-PGE2 caused unexpected DA relaxation that was reversed by EP4 antagonism. In contrast, exposure to 8-iso-PGF2α caused preterm DA constriction via TxA2 receptor activation. Further investigation revealed the predominance of the TxA2 receptor at term, whereas the EP4 receptor was expressed and functionally active from mid-gestation onward. CONCLUSION: This study identifies a novel physiological role for isoprostanes during postnatal vascular transition and provide evidence that oxidative stress may act on membrane lipids to produce vasoactive mediators that stimulate physiological DA closure at birth or induce pathological patency of the preterm DA.


Asunto(s)
Conducto Arterioso Permeable/metabolismo , Conducto Arterial/crecimiento & desarrollo , Isoprostanos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Vasodilatación/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Compuestos Bicíclicos Heterocíclicos con Puentes , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Conducto Arterial/efectos de los fármacos , Conducto Arterioso Permeable/fisiopatología , Ácidos Grasos Insaturados , Femenino , Cromatografía de Gases y Espectrometría de Masas , Perfilación de la Expresión Génica , Hidrazinas/farmacología , Isoprostanos/farmacología , Ratones , Miografía , Estrés Oxidativo/fisiología , Oxígeno/análisis , Embarazo , Nacimiento Prematuro/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores
11.
Dev Biol ; 318(1): 112-25, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18423437

RESUMEN

It is unknown whether or not tight junction formation plays any role in morula to blastocyst transformation that is associated with development of polarized trophoblast cells and fluid accumulation. Tight junctions are a hallmark of polarized epithelial cells and zonula occludens-1 (ZO-1) is a known key regulator of tight junction formation. Here we show that ZO-1 protein is first expressed during compaction of 8-cell embryos. This stage-specific appearance of ZO-1 suggests its participation in morula to blastocyst transition. Consistent with this idea, we demonstrate that ZO-1 siRNA delivery inside the blastomeres of zona-weakened embryos using electroporation not only knocks down ZO-1 gene and protein expressions, but also inhibits morula to blastocyst transformation in a concentration-dependent manner. In addition, ZO-1 inactivation reduced the expression of Cdx2 and Oct-4, but not ZO-2 and F-actin. These results provide the first evidence that ZO-1 is involved in blastocyst formation from the morula by regulating accumulation of fluid and differentiation of nonpolar blastomeres to polar trophoblast cells.


Asunto(s)
Blastocisto/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/metabolismo , Mórula/metabolismo , Fosfoproteínas/metabolismo , Actinas/metabolismo , Animales , Blastocisto/citología , Factor de Transcripción CDX2 , Cadherinas/metabolismo , Electroporación , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Mórula/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfoproteínas/genética , Embarazo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Uniones Estrechas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína de la Zonula Occludens-1 , Proteína de la Zonula Occludens-2
12.
Biol Reprod ; 81(6): 1131-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19684335

RESUMEN

Cytosolic phospholipase A2 (cPLA2, PLA2G4A) catalyzes the release of arachidonic acid for prostaglandin synthesis by cyclooxygenase 1 (PTGS1) and cyclooxygenase 2 (PTGS2). Mice with Pla2g4a deficiency have parturition delay and other reproductive deficits, including deferred onset of implantation, crowding of implantation sites, and small litters. In this study, we examined the contribution of PLA2G4A to parturition in mice. Pla2g4a mRNA and protein expression were discretely localized in the term and preterm uterine luminal epithelium and colocalized with Ptgs1, but not Ptgs2, expression. The levels of PGE2, PGF2alpha, 6-keto-PGF1alpha, and TxB2 were significantly decreased in Pla2g4a-null uterine tissues, similar to Ptgs1-null uteri, consistent with predominance of PLA2G4A-PTGS1-mediated prostaglandin synthesis in preparation for murine parturition. Litter size was strongly associated with the timing of parturition in Pla2g4a-null mice but could not fully account for the parturition delay. Pla2g4a-null females that received PGE2 + carbaprostacyclin at the time of implantation delivered earlier (20.5 +/- 0.2 days vs. 21.6 +/- 0.2 days, P < 0.01), although litter size was not improved (4.6 vs. 4.4 pups per litter, P = 0.6). After correction for small litter size, multivariate analysis indicated that Pla2g4a-null mice given prostaglandin treatment to improve implantation timing had gestational length that was similar to wild-type and Pla2g4a heterozygous mice. These results indicate that, despite specific Pla2g4a expression and function in term gestation uteri, the delayed parturition phenotype in Pla2g4a-null mice is primarily due to deferral of implantation. The role of PLA2G4A in timely parturition appears to be critically related to its actions in early pregnancy.


Asunto(s)
Implantación del Embrión/fisiología , Fosfolipasas A2 Grupo IV/fisiología , Parto/fisiología , Útero/metabolismo , Análisis de Varianza , Animales , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/genética , Femenino , Fosfolipasas A2 Grupo IV/genética , Inmunohistoquímica , Hibridación in Situ , Tamaño de la Camada/efectos de los fármacos , Tamaño de la Camada/genética , Tamaño de la Camada/fisiología , Espectrometría de Masas , Ratones , Ratones Noqueados , Parto/genética , Embarazo , Prostaglandinas/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Regresión , Estadísticas no Paramétricas , Factores de Tiempo
13.
Pediatr Res ; 66(2): 155-61, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19390487

RESUMEN

Although prostaglandin E2 (PGE2) vasodilates the ductus arteriosus, tocolysis with cyclooxygenase (COX) inhibitors delays postnatal ductus arteriosus closure. We used fetal mice and sheep to determine whether PGE2 has a role in the development of ductus contractility that is distinct from its function as a vasodilator. Prolonged exposure of fetal ductus to PGE2 in vitro increased the expression of CaL- and K+-channel genes (CaLalpha1c, CaLbeta2, Kir6.1, and Kv1.5, which regulate oxygen-induced constriction) without affecting the genes that regulate Rho-kinase-mediated calcium sensitization. Conversely, chronic exposure to COX inhibitors in utero decreased expression of CaL- and K+-channel genes, without affecting Rho-kinase-associated genes. Chronic COX inhibition in utero decreased the ductus' in vitro contractile response to stimuli that use CaL- and K+-channels (like O2 and K+), whereas the response to stimuli that act through Rho-kinase-mediated pathways (like U46619) was not significantly affected. Phosphodiesterase expression, which decreases the ductus' sensitivity to cAMP- or cGMP-dependent vasodilators, was increased by PGE2 exposure and decreased by COX inhibition, respectively. These studies identify potential downstream effectors of a PGE2-mediated, developmental program, regulating oxygen-induced ductus closure. Alterations in these effectors may explain the increased risk of patent ductus arteriosus (PDA) after in utero COX inhibition.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/metabolismo , Dinoprostona/metabolismo , Conducto Arterial/metabolismo , Vasoconstricción/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Ratones , Oxígeno/metabolismo , Embarazo , Ovinos
14.
Prostaglandins Other Lipid Mediat ; 88(3-4): 89-96, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19049898

RESUMEN

Nitric oxide (NO) and cyclooxygenase (COX)-derived prostaglandins are critical regulators of the fetal ductus arteriosus. To examine the interaction of these pathways within the ductus wall, the ductus arteriosus of term and preterm fetal mice was evaluated by pressurized myography. The isolated preterm ductus was more sensitive to NOS inhibition than at term. Sequential NOS and COX inhibition caused 36% constriction of the preterm ductus regardless of drug order. In contrast, constriction of the term ductus was dependent on the sequence of inhibition; NOS inhibition prior to COX inhibition produced greater constriction than when inhibitors were given in reverse order (36+/-6% versus 23+/-5%). Selective COX-1 or COX-2 inhibition prior to N(G)-nitro-l-arginine methyl ester (l-NAME) induced the expected degree of constriction. However, NOS inhibition followed by selective COX-2 inhibition caused unexpected ductal dilation. These findings are consistent with NO-induced activation of COX in the ductus arteriosus wall and the production of a COX-2-derived constrictor prostanoid that contributes to the balance of vasoactive forces that maintain fetal ductus arteriosus tone.


Asunto(s)
Conducto Arterial/efectos de los fármacos , Feto/irrigación sanguínea , Óxido Nítrico/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Interacciones Farmacológicas , Femenino , Técnicas In Vitro , Indometacina/farmacología , Ratones , Miografía , NG-Nitroarginina Metil Éster/farmacología , Donantes de Óxido Nítrico/farmacología , Embarazo , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos
15.
Endocrinology ; 159(1): 490-505, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029054

RESUMEN

Cyclooxygenase (COX)-derived prostaglandins stimulate uterine contractions and prepare the cervix for parturition. Prior reports suggest Cox-1 knockout (KO) mice exhibit delayed parturition due to impaired luteolysis, yet the mechanism for late-onset delivery remains unclear. Here, we examined key factors for normal onset of parturition to determine whether any could account for the delayed parturition phenotype. Pregnant Cox-1KO mice did not display altered timing of embryo implantation or postimplantation growth. Although messenger RNAs of contraction-associated proteins (CAPs) were differentially expressed between Cox-1KO and wild-type (WT) myometrium, there were no differences in CAP agonist-induced intracellular calcium release, spontaneous or oxytocin (OT)-induced ex vivo uterine contractility, or in vivo uterine contractile pressure. Delayed parturition in Cox-1KO mice persisted despite exogenous OT treatment. Progesterone (P4) withdrawal, by ovariectomy or administration of the P4-antagonist RU486, diminished the delayed parturition phenotype of Cox-1KO mice. Because antepartum P4 levels do not decline in Cox-1KO females, P4-treated WT mice were examined for the effect of this hormone on in vivo uterine contractility and ex vivo cervical dilation. P4-treated WT mice had delayed parturition but normal uterine contractility. Cervical distensibility was decreased in Cox-1KO mice on the day of expected delivery and reduced in WT mice with long-term P4 treatment. Collectively, these findings show that delayed parturition in Cox-1KO mice is the result of impaired luteolysis and cervical dilation, despite the presence of strong uterine contractions.


Asunto(s)
Maduración Cervical , Cuello del Útero/metabolismo , Ciclooxigenasa 1/metabolismo , Luteólisis , Proteínas de la Membrana/metabolismo , Miometrio/metabolismo , Embarazo Prolongado/metabolismo , Contracción Uterina , Abortivos Esteroideos/farmacología , Abortivos Esteroideos/uso terapéutico , Animales , Células Cultivadas , Maduración Cervical/efectos de los fármacos , Cuello del Útero/efectos de los fármacos , Cuello del Útero/patología , Ciclooxigenasa 1/genética , Femenino , Técnicas In Vitro , Luteólisis/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones Endogámicos , Ratones Noqueados , Mifepristona/farmacología , Mifepristona/uso terapéutico , Miometrio/efectos de los fármacos , Miometrio/patología , Ovariectomía/efectos adversos , Oxitócicos/farmacología , Oxitócicos/uso terapéutico , Oxitocina/farmacología , Oxitocina/uso terapéutico , Embarazo , Embarazo Prolongado/tratamiento farmacológico , Embarazo Prolongado/patología , Embarazo Prolongado/prevención & control , Progesterona/metabolismo , Contracción Uterina/efectos de los fármacos
16.
PLoS One ; 13(5): e0197439, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29771989

RESUMEN

Bacterial DNA has been reported in the placenta and amniotic fluid by several independent groups of investigators. However, it's taxonomic overlap with fetal and maternal bacterial DNA in different sites has been poorly characterized. Here, we determined the presence of bacterial DNA in the intestines and placentas of fetal mice at gestational day 17 (n = 13). These were compared to newborn intestines (n = 15), maternal sites (mouth, n = 6; vagina, n = 6; colon, n = 7; feces, n = 8), and negative controls to rule out contamination. The V4 region of the bacterial 16S rRNA gene indicated a pattern of bacterial DNA in fetal intestine similar to placenta but with higher phylogenetic diversity than placenta or newborn intestine. Firmicutes were the most frequently assignable phylum. SourceTracker analysis suggested the placenta as the most commonly identifiable origin for fetal bacterial DNA, but also over 75% of fetal gut genera overlapped with maternal oral and vaginal taxa but not with maternal or newborn feces. These data provide evidence for the presence of bacterial DNA in the mouse fetus.


Asunto(s)
Líquido Amniótico/metabolismo , ADN Bacteriano/análisis , Mucosa Intestinal/metabolismo , Intestinos/embriología , Placenta/metabolismo , Placenta/microbiología , Animales , Femenino , Ratones , Embarazo , ARN Ribosómico 16S/genética , Vagina/metabolismo , Vagina/microbiología
17.
Sci Rep ; 7(1): 6835, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754971

RESUMEN

Monitoring cervical structure and composition during pregnancy has high potential for prediction of preterm birth (PTB), a problem affecting 15 million newborns annually. We use in vivo Raman spectroscopy, a label-free, light-based method that provides a molecular fingerprint to non-invasively investigate normal and impaired cervical remodeling. Prostaglandins stimulate uterine contractions and are clinically used for cervical ripening during pregnancy. Deletion of cyclooxygenase-1 (Cox-1), an enzyme involved in production of these prostaglandins, results in delayed parturition in mice. Contrary to expectation, Cox-1 null mice displayed normal uterine contractility; therefore, this study sought to determine whether cervical changes could explain the parturition differences in Cox-1 null mice and gestation-matched wild type (WT) controls. Raman spectral changes related to extracellular matrix proteins, lipids, and nucleic acids were tracked over pregnancy and found to be significantly delayed in Cox-1 null mice at term. A cervical basis for the parturition delay was confirmed by other ex vivo tests including decreased tissue distensibility, hydration, and elevated progesterone levels in the Cox-1 null mice at term. In conclusion, in vivo Raman spectroscopy non-invasively detected abnormal remodeling in the Cox-1 null mouse, and clearly demonstrated that the cervix plays a key role in their delayed parturition.


Asunto(s)
Cuello del Útero/metabolismo , Nacimiento a Término/metabolismo , Animales , Cuello del Útero/patología , Cuello del Útero/fisiología , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Metabolismo de los Lípidos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ácidos Nucleicos/metabolismo , Espectrometría Raman , Nacimiento a Término/genética , Contracción Uterina
18.
Endocrinology ; 147(1): 130-40, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16179408

RESUMEN

Biochemical changes of cervical connective tissue, including progressive disorganization of the collagen network and increased water content, occur during gestation to allow for cervical dilatation during labor, but the mechanisms that regulate cervical fluid balance are not fully understood. We examined whether aquaporins (AQPs), a family of membrane channel proteins that facilitate water transport, help mediate fluid balance in the mouse cervix during parturition. Of the 13 known murine AQPs, AQP0-2, 6, 7, 9, 11, and 12 were absent or at the limits of detection. By Northern blot and real-time PCR, AQP3 expression was low in nongravid and mid-pregnancy cervices with peak expression on d 19 and postpartum d 1 (PP1). AQP4 expression was generally low throughout pregnancy but showed a small upward trend at the time of parturition. AQP5 and AQP8 expression were significantly increased on d 12-15 but fell to nongravid/baseline by d 19 and PP1. By in situ hybridization and immunohistochemistry, AQP3 was preferentially expressed in basal cell layers of the cervical epithelium, whereas AQP4, 5, and 8 were primarily expressed in apical cell layers. Females with LPS-induced preterm labor had similar trends in AQP4, 5, and 8 expression to mice with natural labor at term gestation. Mice with delayed cervical remodeling due to deletion of the steroid 5alpha-reductase type 1 gene showed significant reduction in the levels of AQP3, 4, and 8 on d 19 or PP1. Together, these studies suggest that AQPs 3, 4, 5, and 8 regulate distinct aspects of cervical water balance during pregnancy and parturition.


Asunto(s)
Acuaporinas/genética , Cuello del Útero/fisiología , Parto/fisiología , Preñez/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Lipopolisacáridos/farmacología , Ratones , Trabajo de Parto Prematuro/inducido químicamente , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
19.
PLoS One ; 10(11): e0143243, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26600013

RESUMEN

The uterine myometrium (UT-myo) is a therapeutic target for preterm labor, labor induction, and postpartum hemorrhage. Stimulation of intracellular Ca2+-release in UT-myo cells by oxytocin is a final pathway controlling myometrial contractions. The goal of this study was to develop a dual-addition assay for high-throughput screening of small molecular compounds, which could regulate Ca2+-mobilization in UT-myo cells, and hence, myometrial contractions. Primary murine UT-myo cells in 384-well plates were loaded with a Ca2+-sensitive fluorescent probe, and then screened for inducers of Ca2+-mobilization and inhibitors of oxytocin-induced Ca2+-mobilization. The assay exhibited robust screening statistics (Z´ = 0.73), DMSO-tolerance, and was validated for high-throughput screening against 2,727 small molecules from the Spectrum, NIH Clinical I and II collections of well-annotated compounds. The screen revealed a hit-rate of 1.80% for agonist and 1.39% for antagonist compounds. Concentration-dependent responses of hit-compounds demonstrated an EC50 less than 10µM for 21 hit-antagonist compounds, compared to only 7 hit-agonist compounds. Subsequent studies focused on hit-antagonist compounds. Based on the percent inhibition and functional annotation analyses, we selected 4 confirmed hit-antagonist compounds (benzbromarone, dipyridamole, fenoterol hydrobromide and nisoldipine) for further analysis. Using an ex vivo isometric contractility assay, each compound significantly inhibited uterine contractility, at different potencies (IC50). Overall, these results demonstrate for the first time that high-throughput small-molecules screening of myometrial Ca2+-mobilization is an ideal primary approach for discovering modulators of uterine contractility.


Asunto(s)
Calcio/metabolismo , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Miometrio/metabolismo , Contracción Uterina , Útero/metabolismo , Útero/fisiología , Animales , Calcio/agonistas , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Miometrio/citología , Miometrio/efectos de los fármacos , Oxitocina/farmacología , Embarazo , Cultivo Primario de Células , Reproducibilidad de los Resultados , Útero/efectos de los fármacos
20.
Endocrinology ; 144(4): 1533-41, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639938

RESUMEN

The periimplantation period is marked by edematous changes in the uterus. In the mouse, increased uterine vascular permeability occurs in response to estrogen and certain vasoactive mediators, but the mechanisms that regulate fluid transport during implantation are not fully understood. Aquaporins (AQPs) are a family of membrane channel proteins that facilitate bulk water transport. To assess their role in implantation, we examined the expression of AQPs 0-9 in the mouse uterus on d 1-8 of pregnancy. Our results show distinct uterine expression patterns for AQP1, AQP4, and AQP5. AQP1 is localized to the inner circular myometrium throughout the periimplantation period. AQP4 is highly expressed in the luminal epithelium on d 1 of pregnancy but barely detectable at the time of implantation. AQP5 is expressed at low levels in the glandular epithelium during early pregnancy but is markedly increased on d 5. By immunohistochemistry, AQP5 is localized in the basolateral region of the uterine glands. Treatment of adult ovariectomized mice with replacement steroids demonstrates an estrogen-induced shift in AQP1 signals from the myometrium to the uterine stromal vasculature, suggesting a role in uterine fluid imbibition. In contrast, AQP5 is induced only in estrogen-treated, progesterone-primed uteri. We also observed expression of AQP8 in the inner-cell mass and AQP9 in the mural trophectoderm of the implanting blastocyst. Collectively, these results suggest that members of the AQP family are involved in embryo and uterine fluid homeostasis during implantation.


Asunto(s)
Acuaporinas/genética , Acuaporinas/metabolismo , Desarrollo Embrionario/fisiología , Estrógenos/farmacología , Canales Iónicos , Proteínas de la Membrana , Progesterona/farmacología , Animales , Acuaporina 1 , Acuaporina 4 , Acuaporina 5 , Desarrollo Embrionario/efectos de los fármacos , Epitelio/metabolismo , Femenino , Expresión Génica/fisiología , Homeostasis/fisiología , Masculino , Ratones , Embarazo , Útero/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA