Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Eur J Pharmacol ; 960: 176112, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37879426

RESUMEN

Kaempferol is a natural flavonoid compound that exhibits various pharmacological actions. However, there are few reports regarding the role of kaempferol in cardiovascular abnormalities. This study aimed to assess whether kaempferol could prevent cardiovascular malfunction and hypertrophy provoked by chronic inhibition of nitric oxide (NO) formation in rats. Rats (180-200 g) were treated daily with Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME) (40 mg/kg, in drinking water) for five weeks concomitant with kaempferol (oral administration) at a dose of 20 mg/kg or 40 mg/kg or lisinopril (5 mg/kg). Kaempferol partially prevented the progression of hypertension provoked by NO inhibition (p < 0.05). Left ventricular malfunction and hypertrophy present in hypertensive rats were alleviated by concurrent administration of kaempferol (p < 0.05). Furthermore, L-NAME rats had increased sympathetic nerve-mediated vasoconstriction and decreased acetylcholine-induced vasorelaxation and aortic wall thickening, which were resolved by kaempferol treatment (p < 0.05). Kaempferol restored tissue superoxide formation, malondialdehyde, catalase activity, plasma nitric oxide metabolites, tumor necrosis factor-alpha (TNF-α) and interleukin-6 in L-NAME rats (p < 0.05). Overexpression of tumor necrosis factor receptor 2 (TNFR2), phosphatidylinositol 3-kinases (PI3K), AKT serine/threonine kinase 1 (Akt1) and smad2/3 in heart tissue and upregulation of tumor necrosis factor receptor 1 (TNFR1), phosphorylated nuclear factor-kappaB (p-NF-κB) and transforming growth factor beta 1 (TGF-ß1) in vascular tissue were suppressed by kaempferol (p < 0.05). In conclusion, kaempferol exerts antihypertensive, cardioprotective, antioxidant, and anti-inflammatory effects in NO-dependent hypertensive rats. The underlying mechanisms of kaempferol in preventing cardiovascular changes induced by L-NAME were due to the suppression of the TNF-α pathway.


Asunto(s)
Anomalías Cardiovasculares , Hipertensión , Ratas , Animales , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Quempferoles/farmacología , Quempferoles/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Antioxidantes/farmacología , Aorta/metabolismo , Hipertrofia/metabolismo , Anomalías Cardiovasculares/complicaciones , Anomalías Cardiovasculares/metabolismo , Presión Sanguínea
2.
Int J Mol Sci ; 22(21)2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34768894

RESUMEN

The neurons secreting oxytocin (OXY) and vasopressin (AVP) are located mainly in the supraoptic, paraventricular, and suprachiasmatic nucleus of the brain. Oxytocinergic and vasopressinergic projections reach several regions of the brain and the spinal cord. Both peptides are released from axons, soma, and dendrites and modulate the excitability of other neuroregulatory pathways. The synthesis and action of OXY and AVP in the peripheral organs (eye, heart, gastrointestinal system) is being investigated. The secretion of OXY and AVP is influenced by changes in body fluid osmolality, blood volume, blood pressure, hypoxia, and stress. Vasopressin interacts with three subtypes of receptors: V1aR, V1bR, and V2R whereas oxytocin activates its own OXTR and V1aR receptors. AVP and OXY receptors are present in several regions of the brain (cortex, hypothalamus, pons, medulla, and cerebellum) and in the peripheral organs (heart, lungs, carotid bodies, kidneys, adrenal glands, pancreas, gastrointestinal tract, ovaries, uterus, thymus). Hypertension, myocardial infarction, and coexisting factors, such as pain and stress, have a significant impact on the secretion of oxytocin and vasopressin and on the expression of their receptors. The inappropriate regulation of oxytocin and vasopressin secretion during ischemia, hypoxia/hypercapnia, inflammation, pain, and stress may play a significant role in the pathogenesis of cardiovascular diseases.


Asunto(s)
Anomalías Cardiovasculares , Oxitocina/metabolismo , Vasopresinas/metabolismo , Axones/metabolismo , Encéfalo/metabolismo , Anomalías Cardiovasculares/etiología , Anomalías Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Humanos , Hipertensión/etiología , Hipertensión/metabolismo , Pulmón/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Neuronas/metabolismo , Neurofisinas/metabolismo , Precursores de Proteínas/metabolismo , Receptores de Oxitocina/metabolismo
3.
Int J Mol Sci ; 22(9)2021 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068698

RESUMEN

Immune, neuroendocrine, and autonomic nervous system dysregulation in anorexia nervosa lead to cardiovascular complications that can potentially result in increased morbidity and mortality. It is suggested that a complex non-invasive assessment of cardiovascular autonomic regulation-cardiac vagal control, sympathetic vascular activity, and cardiovascular reflex control-could represent a promising tool for early diagnosis, personalized therapy, and monitoring of therapeutic interventions in anorexia nervosa particularly at a vulnerable adolescent age. In this view, we recommend to consider in the diagnostic route, at least in the subset of patients with peripheral microvascular symptoms, a nailfold video-capillaroscopy as an easy not invasive tool for the early assessing of possible cardiovascular involvement.


Asunto(s)
Anorexia Nerviosa/patología , Anomalías Cardiovasculares/patología , Enfermedades Vasculares Periféricas/patología , Anorexia Nerviosa/complicaciones , Anorexia Nerviosa/inmunología , Anorexia Nerviosa/metabolismo , Anomalías Cardiovasculares/complicaciones , Anomalías Cardiovasculares/inmunología , Anomalías Cardiovasculares/metabolismo , Frecuencia Cardíaca/fisiología , Humanos , Sistema Inmunológico/patología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Enfermedades Vasculares Periféricas/complicaciones , Enfermedades Vasculares Periféricas/inmunología , Enfermedades Vasculares Periféricas/metabolismo , Nervio Vago/metabolismo , Nervio Vago/patología
4.
Biomed Res Int ; 2020: 5073762, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32851077

RESUMEN

Vascular endothelial insulin signaling is critical for the maintenance of vascular and metabolic homeostasis. We have previously shown that in hypertensive Dahl rats, impaired vascular insulin action is linked to angiotensin II activation of the NFκB inflammatory pathway. Macrophage polarization (M1) has implicated in hypertensive and metabolic diseases. Here, we investigated the effect of macrophage depletion using liposome-encapsulated clodronate (LEC) on endothelial insulin resistance and cardiovascular remodeling in Dahl salt-sensitive (DS) rats. High salt intake (HS) for 5 weeks increased systolic blood pressure (SBP: 192 ± 5 vs. 144 ± 4 mmHg in NS, p < 0.05), aortic and cardiac hypertrophy, cardiac fibrosis, and impaired acetylcholine- and insulin-induced vasorelaxation, accompanied by impaired insulin activation of endothelial nitric oxide synthases (eNOS)/NO signaling. HS rats had a significant increase in CD68 (a monocyte/macrophage marker) expression in the aorta and the heart. LEC reduced SBP (168 ± 5 mmHg, p < 0.05) and cardiovascular injury and improved acetylcholine- and insulin-mediated vasorelaxation and insulin signaling molecules with a reduction in the macrophage infiltration in the aorta and the heart. HS rats also manifested an increase in the aortic expressions of inflammatory cytokines, including the ratio of phosphorylated inhibitory kappa B (Iκb)/Iκb, tumor necrosis factor α, and phosphorylated c-Jun N-terminal kinase (JNK) and oxidative stress, which were reduced in HS/LEC rats. Our results suggest that in salt-sensitive hypertension, macrophage may importantly contribute to endothelial insulin resistance, vascular inflammation, and injury. These findings support the idea that macrophages may be a new target for immunotherapy of vasculopathy in hypertensive and metabolic disorders.


Asunto(s)
Anomalías Cardiovasculares/genética , Hipertensión/metabolismo , Resistencia a la Insulina/genética , Cloruro de Sodio/metabolismo , Angiotensina II/genética , Animales , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/patología , Anomalías Cardiovasculares/prevención & control , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Hipertensión/genética , Hipertensión/patología , Hipertensión/prevención & control , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratas , Cloruro de Sodio/efectos adversos , Cloruro de Sodio Dietético/farmacología
5.
Pediatr Pulmonol ; 55(10): 2674-2682, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32662935

RESUMEN

BACKGROUND: Cilia are cell membrane-bound organelles responsible for airway mucus clearance, establishment of left-right organ asymmetry, cardiogenesis, and many other functions in utero. Phenotypic features suggestive of respiratory ciliary dyskinesia among patients with heterotaxy syndrome, defined as complex cardiovascular malformations (CVM) and situs ambiguus (SA), has not been adequately explored. OBJECTIVES: We hypothesized that there is a greater incidence of phenotypic features consistent with ciliary dyskinesia among patients with heterotaxy syndrome compared to patients with other CVM and laterality defects without heterotaxy syndrome. METHODS: Thirty six subjects were identified by medical record search and divided into four groups based on situs status and type of CVM as follows: SA and complex CVM (group 1); SA and simple CVM (group 2); situs solitus and complex CVM (group 3); and situs solitus and simple CVM (group 4). Phenotype was assessed with a clinical questionnaire, nasal nitric oxide (NO) level, and pulmonary function testing. Those with complex CVM underwent additional testing for variants in genes involved in ciliary structure and function. RESULTS: The mean nasal NO level was significantly lower among all subjects with complex CVM regardless of situs anomalies (groups 1 and 3). There was no significant difference in respiratory symptoms or lung function among the four groups. No bi-allelic genetic mutations were detected among patients with complex CVM. CONCLUSIONS: This study identified a relatively lower mean nasal NO level, suggestive of relative ciliary dyskinesia, among subjects with complex CVM. Pulmonary function and clinical symptoms did not reflect significant pulmonary disease among those with complex CVM.


Asunto(s)
Anomalías Cardiovasculares/metabolismo , Trastornos de la Motilidad Ciliar/metabolismo , Óxido Nítrico/metabolismo , Situs Inversus/metabolismo , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Cavidad Nasal , Fenotipo , Adulto Joven
6.
Am J Med Genet A ; 179(6): 1047-1052, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30941910

RESUMEN

Kosaki overgrowth syndrome is a recently described syndrome characterized by distinctive facial features, brain white matter lesions, and developmental delay. Germline activating heterozygous PDGFRB mutations have been reported in this condition. Systemic connective tissue-type findings have been described in some individuals. We describe a 19-year-old Caucasian female with a history of hydrocephalus, Dandy-Walker malformation, cervical spine arachnoid cyst, progressive scoliosis, and overgrowth. Her physical exam included distinctive craniofacial dysmorphism, as well as soft and hyperextensible skin. Cardiovascular imaging during adolescence revealed saccular aneurysms in both coronary artery systems and subtle tortuosity of the cervical vertebral arteries. Exome sequencing trio analysis identified a de novo previously reported pathogenic variant in PDGFRB, c.1696T>C (p.[Trp566Arg]). Further functional studies included platelet-derived growth factor cellular metabolic pathway activity that confirmed the variant causes a constitutive activation of the PI3K-AKT pathway. This is the first report to characterize the activating nature of this PDGFRB variant. We also highlight the connective tissue findings seen in Kosaki overgrowth syndrome and recommend baseline echocardiographic evaluation in all individuals with this condition with particular emphasis on coronary arteries.


Asunto(s)
Anomalías Cardiovasculares/etiología , Anomalías Cardiovasculares/metabolismo , Trastornos del Crecimiento/complicaciones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Anomalías Cardiovasculares/diagnóstico , Metabolismo Energético , Facies , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/diagnóstico , Trastornos del Crecimiento/genética , Humanos , Imagen por Resonancia Magnética , Fenotipo , Fosforilación , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Secuenciación del Exoma , Adulto Joven
7.
PLoS One ; 13(11): e0207251, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30408103

RESUMEN

The CXCL12-CXCR4 pathway has crucial roles in stem cell homing and maintenance, neuronal guidance, cancer progression, inflammation, remote-conditioning, cell migration and development. Recently, work in chick suggested that signalling via CXCR4 in neural crest cells (NCCs) has a role in the 22q11.2 deletion syndrome (22q11.2DS), a disorder where haploinsufficiency of the transcription factor TBX1 is responsible for the major structural defects. We tested this idea in mouse models. Our analysis of genes with altered expression in Tbx1 mutant mouse models showed down-regulation of Cxcl12 in pharyngeal surface ectoderm and rostral mesoderm, both tissues with the potential to signal to migrating NCCs. Conditional mutagenesis of Tbx1 in the pharyngeal surface ectoderm is associated with hypo/aplasia of the 4th pharyngeal arch artery (PAA) and interruption of the aortic arch type B (IAA-B), the cardiovascular defect most typical of 22q11.2DS. We therefore analysed constitutive mouse mutants of the ligand (CXCL12) and receptor (CXCR4) components of the pathway, in addition to ectodermal conditionals of Cxcl12 and NCC conditionals of Cxcr4. However, none of these typical 22q11.2DS features were detected in constitutively or conditionally mutant embryos. Instead, duplicated carotid arteries were observed, a phenotype recapitulated in Tie-2Cre (endothelial) conditional knock outs of Cxcr4. Previous studies have demonstrated genetic interaction between signalling pathways and Tbx1 haploinsufficiency e.g. FGF, WNT, SMAD-dependent. We therefore tested for possible epistasis between Tbx1 and the CXCL12 signalling axis by examining Tbx1 and Cxcl12 double heterozygotes as well as Tbx1/Cxcl12/Cxcr4 triple heterozygotes, but failed to identify any exacerbation of the Tbx1 haploinsufficient arch artery phenotype. We conclude that CXCL12 signalling via NCC/CXCR4 has no major role in the genesis of the Tbx1 loss of function phenotype. Instead, the pathway has a distinct effect on remodelling of head vessels and interventricular septation mediated via CXCL12 signalling from the pharyngeal surface ectoderm and second heart field to endothelial cells.


Asunto(s)
Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Quimiocina CXCL12/deficiencia , Receptores CXCR4/deficiencia , Proteínas de Dominio T Box/deficiencia , Animales , Aorta Torácica/anomalías , Aorta Torácica/embriología , Aorta Torácica/metabolismo , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Sistema Cardiovascular/embriología , Quimiocina CXCL12/genética , Síndrome de DiGeorge/enzimología , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/metabolismo , Modelos Animales de Enfermedad , Epistasis Genética , Femenino , Haploinsuficiencia , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Cresta Neural/metabolismo , Embarazo , Receptores CXCR4/genética , Transducción de Señal/genética , Proteínas de Dominio T Box/genética
8.
Genes Cells ; 23(11): 938-951, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30151851

RESUMEN

Integrins are transmembrane molecules that facilitate cell-to-cell and cell-to-extracellular matrix (ECM) interactions. Integrin molecules are heterodimers that consist of α- and ß-subunits. The integrin ß1 gene is widely expressed in vivo and is the major ß molecule in many tissues; however, tissue-specific roles of integrin ß1 are still elusive. In this study, we investigated integrin ß1 function in endothelial cells of zebrafish. An integrin ß1b mutant zebrafish exhibited morphological abnormalities in blood vessel formation, cephalic hemorrhage and a decreased responsiveness to tactile stimulation during development. To determine the role of integrin ß1b in vascular formation, we developed a Gal4/UAS-mediated conditional inactivation of integrin ß1 by expressing the cytoplasmic region of integrin ß1 that acts as a dominant-negative (DN) isoform. Expression of integrin ß1 DN in endothelial cells induced blood vessel abnormalities as in integrin ß1b mutants. These results show that endothelial cells require integrin activity for the formation and/or maintenance of blood vessels in zebrafish. Furthermore, our time-lapse recording visualized the breakpoint of cephalic vessels and the hemorrhage onset. Taken together, our tissue-specific inactivation of integrin ß1 in zebrafish is powerful tools for functional analysis of integrin ß1 in developing tissues.


Asunto(s)
Anomalías Cardiovasculares/patología , Embrión no Mamífero/patología , Endotelio Vascular/patología , Hemorragia/patología , Integrina beta1/metabolismo , Mutación , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Embrión no Mamífero/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Integrina beta1/genética , Pez Cebra/genética , Pez Cebra/metabolismo
9.
Sci Rep ; 8(1): 7745, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773828

RESUMEN

Genetically modified mice models suggest an important role for G-protein-coupled receptor kinase 5 (GRK5) in the pathophysiology of obesity and related disorders. We investigated whether single nucleotide polymorphisms (SNPs) in the gene encoding GRK5 affect cardiometabolic traits in humans. We genotyped 3 common SNPs in intron 1 (rs1980030, rs10466210, rs9325562) and one SNP in intron 3 (rs10886471) of GRK5 in 2332 subjects at risk for type 2 diabetes. Total- and visceral fat mass were measured by magnetic resonance (MR) tomography and liver fat content by 1H-MR spectroscopy. Insulin secretion and sensitivity were estimated during an OGTT and measured during the euglycemic, hyperinsulinemic clamp (n = 498). Carriers of the minor allele of rs10466210 and rs1980030 had higher total- and LDL-cholesterol levels (p = 0.0018 and p = 0.0031, respectively, for rs10466210; p = 0.0035 and p = 0.0081, respectively, for rs1980030), independently of gender, age, BMI and lipid-lowering drugs. The effects of rs10466210 withstood Bonferroni correction. Similar associations were observed with apolipoprotein B levels (p = 0.0034 and p = 0.0122, respectively). Carriers of the minor allele of rs10466210 additionally displayed a trend for higher intima-media thickness of the carotid artery (p = 0.075). GRK5 may represent a novel target for strategies aiming at lowering LDL-cholesterol levels and at modifying cardiovascular risk.


Asunto(s)
Anomalías Cardiovasculares/etiología , Grosor Intima-Media Carotídeo , LDL-Colesterol/sangre , Diabetes Mellitus Tipo 2/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Resistencia a la Insulina , Polimorfismo de Nucleótido Simple/genética , Adulto , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/patología , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Insulina/metabolismo , Lípidos/sangre , Masculino , Persona de Mediana Edad
10.
Birth Defects Res ; 109(10): 791-804, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28509418

RESUMEN

The most impressive phenotypic appearance of sirenomelia is the presence of a 180°-rotated, axially positioned, single lower limb. Associated gastrointestinal and genitourinary anomalies are almost always present. This rare anomaly is still the subject of ongoing controversies concerning its nosology, pathogenesis, and possible genetic etiology. Sirenomelia can be part of a syndromic continuum, overlapping with other complex conditions including caudal dysgenesis and VATER/VACTERL/VACTERL-H associations, which could all be part of a heterogeneous spectrum, and originate from an early defect in blastogenesis. It is imaginable that different "primary field defects," whether or not genetically based, induce a spectrum of caudal malformations. In the current study, we review the contemporary hypotheses and conceptual approaches regarding the etiology and pathogenesis of sirenomelia, especially in the context of concomitant conditions. To expand on the latter, we included the external and internal dysmorphology of one third trimester sirenomelic fetus from our anatomical museum collection, in which multiple concomitant but discordant anomalies were observed compared with classic sirenomelia, and was diagnosed as VACTERL-H association with sirenomelia. Birth Defects Research 109:791-804, 2017. © 2017 The Authors. Birth Defects Research Published by Wiley Periodicals, Inc.


Asunto(s)
Ectromelia/metabolismo , Ectromelia/fisiopatología , Anomalías Múltiples/patología , Canal Anal/anomalías , Canal Anal/metabolismo , Canal Anal/fisiopatología , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/fisiopatología , Anomalías del Sistema Digestivo/metabolismo , Anomalías del Sistema Digestivo/fisiopatología , Ectromelia/complicaciones , Ectromelia/diagnóstico , Esófago/anomalías , Esófago/metabolismo , Esófago/fisiopatología , Feto/anomalías , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/fisiopatología , Humanos , Hidrocefalia/metabolismo , Hidrocefalia/fisiopatología , Riñón/anomalías , Riñón/metabolismo , Riñón/fisiopatología , Deformidades Congénitas de las Extremidades/metabolismo , Deformidades Congénitas de las Extremidades/fisiopatología , Anomalías Musculoesqueléticas/metabolismo , Anomalías Musculoesqueléticas/fisiopatología , Columna Vertebral/anomalías , Columna Vertebral/metabolismo , Columna Vertebral/fisiopatología , Tráquea/anomalías , Tráquea/metabolismo , Tráquea/fisiopatología , Anomalías Urogenitales/etiología , Anomalías Urogenitales/fisiopatología
11.
PLoS Genet ; 13(3): e1006687, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28346476

RESUMEN

The 22q11.2 deletion syndrome (22q11.2DS; velo-cardio-facial syndrome; DiGeorge syndrome) is a congenital anomaly disorder in which haploinsufficiency of TBX1, encoding a T-box transcription factor, is the major candidate for cardiac outflow tract (OFT) malformations. Inactivation of Tbx1 in the anterior heart field (AHF) mesoderm in the mouse results in premature expression of pro-differentiation genes and a persistent truncus arteriosus (PTA) in which septation does not form between the aorta and pulmonary trunk. Canonical Wnt/ß-catenin has major roles in cardiac OFT development that may act upstream of Tbx1. Consistent with an antagonistic relationship, we found the opposite gene expression changes occurred in the AHF in ß-catenin loss of function embryos compared to Tbx1 loss of function embryos, providing an opportunity to test for genetic rescue. When both alleles of Tbx1 and one allele of ß-catenin were inactivated in the Mef2c-AHF-Cre domain, 61% of them (n = 34) showed partial or complete rescue of the PTA defect. Upregulated genes that were oppositely changed in expression in individual mutant embryos were normalized in significantly rescued embryos. Further, ß-catenin was increased in expression when Tbx1 was inactivated, suggesting that there may be a negative feedback loop between canonical Wnt and Tbx1 in the AHF to allow the formation of the OFT. We suggest that alteration of this balance may contribute to variable expressivity in 22q11.2DS.


Asunto(s)
Anomalías Cardiovasculares/genética , Síndrome de DiGeorge/genética , Modelos Animales de Enfermedad , Proteínas de Dominio T Box/genética , beta Catenina/genética , Animales , Apoptosis/genética , Anomalías Cardiovasculares/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/genética , Síndrome de DiGeorge/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/metabolismo , Tronco Arterial/citología , Tronco Arterial/embriología , Tronco Arterial/metabolismo , beta Catenina/metabolismo
12.
Environ Toxicol Pharmacol ; 46: 292-300, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27525560

RESUMEN

Glyphosate is a broad spectrum herbicide used aggressively in agricultural practices as well as home garden care. Although labeled "safe" by the chemical industry, doses tested by industry do not mimic chronic exposures to sublethal doses that organisms in the environment are exposed to over long periods of time. Given the widespread uses of and exposure to glyphosate, studies on developmental toxicity are needed. Here we utilize the zebrafish vertebrate model system to study early effects of glyphosate on the developing heart. Treatment by embryo soaking with 50µg/ml glyphosate starting at gastrulation results in structural abnormalities in the atrium and ventricle, irregular heart looping, situs inversus as well as decreased heartbeats by 48h as determined by live imaging and immunohistochemistry. Vasculature in the body was also affected as determined using fli-1 transgenic embryos. To determine if the effects noted at 48h post fertilization are due to early stage alterations in myocardial precursors, we also investigate cardiomyocyte development with a Mef2 antibody and by mef2ca in situ hybridization and find alterations in the Mef2/mef2ca staining patterns during early cardiac patterning stages. We conclude that glyphosate is developmentally toxic to the zebrafish heart.


Asunto(s)
Anomalías Cardiovasculares/inducido químicamente , Embrión no Mamífero/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Glicina/análogos & derivados , Corazón/efectos de los fármacos , Pez Cebra , Animales , Animales Modificados Genéticamente , Cardiotoxicidad , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/patología , Anomalías Cardiovasculares/fisiopatología , Embrión no Mamífero/anomalías , Glicina/toxicidad , Proteínas Fluorescentes Verdes/genética , Corazón/embriología , Corazón/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Factores Reguladores Miogénicos/genética , Proteína Proto-Oncogénica c-fli-1/genética , Pez Cebra/anomalías , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Glifosato
13.
Mol Cell Biol ; 35(23): 3990-4005, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26391956

RESUMEN

p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements, cell proliferation, attachment, and migration in a variety of cellular contexts, including endothelial cells. However, the role of endothelial Pak in embryo development has not been reported, and currently, there is no consensus on the endothelial function of individual Pak isoforms, in particular p21-activated kinase 2 (Pak2), the main Pak isoform expressed in endothelial cells. In this work, we employ genetic and molecular studies that show that Pak2, but not Pak1, is a critical mediator of development and maintenance of endothelial cell function. Endothelial depletion of Pak2 leads to early embryo lethality due to flawed blood vessel formation in the embryo body and yolk sac. In adult endothelial cells, Pak2 depletion leads to severe apoptosis and acute angiogenesis defects, and in adult mice, endothelial Pak2 deletion leads to increased vascular permeability. Furthermore, ubiquitous Pak2 deletion is lethal in adult mice. We show that many of these defects are mediated through a newly unveiled Pak2/Bmk1 pathway. Our results demonstrate that endothelial Pak2 is essential during embryogenesis and also for adult blood vessel maintenance, and they also pinpoint the Bmk1/Erk5 pathway as a critical mediator of endothelial Pak2 signaling.


Asunto(s)
Endotelio/embriología , Endotelio/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Animales , Permeabilidad Capilar , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Pérdida del Embrión , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Endotelio/citología , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Masculino , Ratones Endogámicos C57BL , Interferencia de ARN , Quinasas p21 Activadas/genética
14.
PLoS One ; 10(4): e0123965, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25860837

RESUMEN

The Dlg1 gene encodes a member of the MAGUK protein family involved in the polarization of epithelial cells. Null mutant mice for the Dlg1 gene (Dlg1-/- mice) exhibit respiratory failure and cyanosis, and die soon after birth. However, the cause of this neonatal lethality has not been determined. In the present study, we further examined Dlg1-/- mice and found severe defects in the cardiovascular system, including ventricular septal defect, persistent truncus arteriosus, and double outlet right ventricle, which would cause the neonatal lethality. These cardiovascular phenotypes resemble those of mutant mice lacking planar cell polarity (PCP) genes and support a recent notion that DLG1 is involved in the PCP pathway. We assessed the degree of involvement of DLG1 in the development of other organs, as the cochlea, intestine, and skeleton, in which PCP signaling has been suggested to play a role. In the organ of Corti, tissue elongation was inhibited accompanied by disorganized arrangement of the hair cell rows, while the orientation of the stereocilia bundle was normal. In the sternum, cleft sternum, abnormal calcification pattern of cartilage, and disorganization of chondrocytes were observed. Furthermore, shortening of the intestine, sternum, and long bones of the limbs was observed. These phenotypes of Dlg1-/- mice involving cellular disorganization and insufficient tissue elongation strongly suggest a defect in the convergent extension movements in these mice. Thus, our present results provide a possibility that DLG1 is particularly required for convergent extension among PCP signaling-dependent processes.


Asunto(s)
Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Morfogénesis/genética , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Animales , Desarrollo Óseo/genética , Desarrollo Óseo/fisiología , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Polaridad Celular/genética , Polaridad Celular/fisiología , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Homólogo 1 de la Proteína Discs Large , Femenino , Corazón Fetal/crecimiento & desarrollo , Corazón Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/metabolismo , Intestinos/embriología , Intestinos/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Embarazo , Proteínas Asociadas a SAP90-PSD95 , Transducción de Señal
15.
Artículo en Inglés | MEDLINE | ID: mdl-23716477

RESUMEN

BACKGROUND: Heart defects are the most common abnormalities in infants of diabetic mothers. Cardiac malformation is associated with altered expression of the genes in the transforming growth factor ß system, including inhibin ßA, which forms activin-A as a homodimer and functions through its effectors, Smad2 and Smad3. This study aimed to investigate the role of activin-A in diabetes-induced cardiac malformations. METHODS: Diabetes mellitus in female mice (C57BL/6J) was induced via intravenous injection of streptozotocin. The expression of inhibin ßA protein and phosphorylation of Smad2 and Smad3 in the embryonic hearts were examined using immunohistochemical, in situ proximity ligation, and immunoblot assays. Embryos and endocardial cushions of nondiabetic mice were cultured in a high concentration of glucose and treated with activin-A. Mitosis was examined using BrdU incorporation assay and immunohistochemistry of phosphorylated histone H3. Migration of the endocardial cells was assessed using a collagen-based cell migration assay. RESULTS: The levels of inhibin ßA expression and Smad2 and Smad3 activation were significantly reduced by maternal diabetes. Treatment with activin-A significantly increased cell proliferation in the myocardium and migration of endocardial cells, compared with those in vehicle-treated high glucose group, to the level in the euglycemic control group. CONCLUSIONS: Maternal diabetes suppresses the expression of inhibin ßA protein, as well as the activation of Smad2 and Smad3. Activin-A rescues cell proliferation in the myocardium and migration of the endocardial cells suppressed by hyperglycemia. The activin-Smad2/3 signaling system appears to play a role in cardiac malformation in diabetic embryopathy.


Asunto(s)
Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/etiología , Subunidades beta de Inhibinas/metabolismo , Embarazo en Diabéticas , Animales , Bromodesoxiuridina/metabolismo , Anomalías Cardiovasculares/tratamiento farmacológico , Anomalías Cardiovasculares/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Endocardio/metabolismo , Endocardio/patología , Femenino , Inmunohistoquímica , Subunidades beta de Inhibinas/farmacología , Subunidades beta de Inhibinas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Fosforilación , Embarazo , Embarazo en Diabéticas/metabolismo , Proteína smad3/metabolismo
16.
Prenat Diagn ; 32(11): 1041-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22855024

RESUMEN

OBJECTIVE: Periconceptional folate supplementation prevents a number of congenital anomalies (CA). The aim of our study was to investigate the association of 11 polymorphisms in the folate-metabolizing genes with the risk of having an offspring with CA in the Russian ethnic group. METHOD: We genotyped 280 mothers having a CA-affected pregnancy and 390 control mothers. The most common malformations among the cases were CA of the nervous, urinary, and cardiovascular systems, and these groups were analyzed separately. RESULTS: In the whole group of CA, we revealed the associations of MTHFR C677T and MTR A2756G loci with increased risk of CA-affected pregnancy. In the group of CA of the cardiovascular system, we observed an association of MTHFR A1298C with decreased risk and an association of MTR A2756G with increased risk of CA. After the Bonferroni correction, only the association between the genotype MTR 2756GG and the risk of having a fetus with CA of the cardiovascular system remained statistically significant (OR = 4.99, P = 0.03). CONCLUSION: These findings indicate that locus A2756G in the MTR gene may play a role in susceptibility to CA of the cardiovascular system in West Siberia, but further research is necessary to confirm the association.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Anomalías Congénitas/genética , Predisposición Genética a la Enfermedad , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo de Nucleótido Simple , Adulto , Anomalías Cardiovasculares/epidemiología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Estudios de Casos y Controles , Anomalías Congénitas/metabolismo , Femenino , Genotipo , Humanos , Malformaciones del Sistema Nervioso/epidemiología , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Embarazo , Siberia/epidemiología , Anomalías Urogenitales/epidemiología , Anomalías Urogenitales/genética , Anomalías Urogenitales/metabolismo
17.
J Clin Invest ; 122(6): 2006-17, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22622036

RESUMEN

Human vascular malformations cause disease as a result of changes in blood flow and vascular hemodynamic forces. Although the genetic mutations that underlie the formation of many human vascular malformations are known, the extent to which abnormal blood flow can subsequently influence the vascular genetic program and natural history is not. Loss of the SH2 domain-containing leukocyte protein of 76 kDa (SLP76) resulted in a vascular malformation that directed blood flow through mesenteric lymphatic vessels after birth in mice. Mesenteric vessels in the position of the congenital lymphatic in mature Slp76-null mice lacked lymphatic identity and expressed a marker of blood vessel identity. Genetic lineage tracing demonstrated that this change in vessel identity was the result of lymphatic endothelial cell reprogramming rather than replacement by blood endothelial cells. Exposure of lymphatic vessels to blood in the absence of significant flow did not alter vessel identity in vivo, but lymphatic endothelial cells exposed to similar levels of shear stress ex vivo rapidly lost expression of PROX1, a lymphatic fate-specifying transcription factor. These findings reveal that blood flow can convert lymphatic vessels to blood vessels, demonstrating that hemodynamic forces may reprogram endothelial and vessel identity in cardiovascular diseases associated with abnormal flow.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anomalías Cardiovasculares/metabolismo , Células Endoteliales/metabolismo , Proteínas de Homeodominio/biosíntesis , Vasos Linfáticos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Velocidad del Flujo Sanguíneo , Anomalías Cardiovasculares/patología , Línea Celular , Células Endoteliales/patología , Proteínas de Homeodominio/genética , Humanos , Vasos Linfáticos/anomalías , Vasos Linfáticos/patología , Ratones , Ratones Mutantes , Fosfoproteínas/genética , Proteínas Supresoras de Tumor/genética
18.
Hum Mol Genet ; 21(6): 1248-59, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22116938

RESUMEN

Growth factor signaling results in dramatic phenotypic changes in cells, which require commensurate alterations in cellular metabolism. Mutations in SLC2A10/GLUT10, a member of the facilitative glucose transporter family, are associated with altered transforming growth factor-ß (TGFß) signaling in patients with arterial tortuosity syndrome (ATS). The objective of this work was to test whether SLC2A10/GLUT10 can serve as a link between TGFß-related transcriptional regulation and metabolism during development. In zebrafish embryos, knockdown of slc2a10 using antisense morpholino oligonucleotide injection caused a wavy notochord and cardiovascular abnormalities with a reduced heart rate and blood flow, which was coupled with an incomplete and irregular vascular patterning. This was phenocopied by treatment with a small-molecule inhibitor of TGFß receptor (tgfbr1/alk5). Array hybridization showed that the changes at the transcriptome level caused by the two treatments were highly correlated, revealing that a reduced tgfbr1 signaling is a key feature of ATS in early zebrafish development. Interestingly, a large proportion of the genes, which were specifically dysregulated after glut10 depletion gene and not by tgfbr1 inhibition, play a major role in mitochondrial function. Consistent with these results, slc2a10 morphants showed decreased respiration and reduced TGFß reporter gene activity. Finally, co-injection of antisense morpholinos targeting slc2a10 and smad7 (a TGFß inhibitor) resulted in a partial rescue of smad7 morphant phenotypes, suggesting scl2a10/glut10 functions downstream of smads. Taken together, glut10 is essential for cardiovascular development by facilitating both mitochondrial respiration and TGFß signaling.


Asunto(s)
Anomalías Cardiovasculares/etiología , Proteínas Facilitadoras del Transporte de la Glucosa/fisiología , Mitocondrias/metabolismo , Notocorda/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Anomalías Cardiovasculares/metabolismo , Anomalías Cardiovasculares/patología , Luciferasas/metabolismo , Mitocondrias/patología , Datos de Secuencia Molecular , Morfolinos/farmacología , Mutación/genética , Notocorda/patología , Fenotipo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Transcriptoma , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Pez Cebra/crecimiento & desarrollo
19.
Development ; 138(10): 1925-34, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490058

RESUMEN

Protein O-glucosylation is a conserved post-translational modification that occurs on epidermal growth factor-like (EGF) repeats harboring the C(1)-X-S-X-P-C(2) consensus sequence. The Drosophila protein O-glucosyltransferase (Poglut) Rumi regulates Notch signaling, but the contribution of protein O-glucosylation to mammalian Notch signaling and embryonic development is not known. Here, we show that mouse Rumi encodes a Poglut, and that Rumi(-/-) mouse embryos die before embryonic day 9.5 with posterior axis truncation and severe defects in neural tube development, somitogenesis, cardiogenesis and vascular remodeling. Rumi knockdown in mouse cell lines results in cellular and molecular phenotypes of loss of Notch signaling without affecting Notch ligand binding. Biochemical, cell culture and cross-species transgenic experiments indicate that a decrease in Rumi levels results in reduced O-glucosylation of Notch EGF repeats, and that the enzymatic activity of Rumi is key to its regulatory role in the Notch pathway. Genetic interaction studies show that removing one copy of Rumi in a Jag1(+/-) (jagged 1) background results in severe bile duct morphogenesis defects. Altogether, our data indicate that addition of O-glucose to EGF repeats is essential for mouse embryonic development and Notch signaling, and that Jag1-induced signaling is sensitive to the gene dosage of the protein O-glucosyltransferase Rumi. Given that Rumi(-/-) embryos show more severe phenotypes compared to those displayed by other global regulators of canonical Notch signaling, Rumi is likely to have additional important targets during mammalian development.


Asunto(s)
Desarrollo Embrionario/fisiología , Glucosiltransferasas/metabolismo , Receptores Notch/metabolismo , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Conductos Biliares Intrahepáticos/anomalías , Conductos Biliares Intrahepáticos/metabolismo , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Línea Celular , Proteínas de Drosophila , Desarrollo Embrionario/genética , Factor de Crecimiento Epidérmico/genética , Femenino , Dosificación de Gen , Glucosiltransferasas/deficiencia , Glucosiltransferasas/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Hígado/anomalías , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Fenotipo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal
20.
Toxicol Appl Pharmacol ; 254(2): 100-26, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21296098

RESUMEN

Reactions between chemicals used to disinfect drinking water and compounds present in source waters produce chemical mixtures containing hundreds of disinfection byproducts (DBPs). Although the results have been somewhat inconsistent, some epidemiological studies suggest associations may exist between DBP exposures and adverse developmental outcomes. The potencies of individual DBPs in rodent and rabbit developmental bioassays suggest that no individual DBP can account for the relative risk estimates reported in the positive epidemiologic studies, leading to the hypothesis that these outcomes could result from the toxicity of DBP mixtures. As a first step in a mixtures risk assessment for DBP developmental effects, this paper identifies developmentally toxic DBPs and examines data relevant to the mode of action (MOA) for DBP developmental toxicity. We identified 24 developmentally toxic DBPs and four adverse developmental outcomes associated with human DBP exposures: spontaneous abortion, cardiovascular defects, neural tube defects, and low birth weight infancy. A plausible MOA, involving hormonal disruption of pregnancy, is delineated for spontaneous abortion, which some epidemiologic studies associate with total trihalomethane and bromodichloromethane exposures. The DBP data for the other three outcomes were inadequate to define key MOA steps.


Asunto(s)
Aborto Espontáneo/epidemiología , Anomalías Cardiovasculares/epidemiología , Desinfectantes/toxicidad , Recién Nacido de Bajo Peso , Defectos del Tubo Neural/epidemiología , Abastecimiento de Agua , Aborto Espontáneo/inducido químicamente , Aborto Espontáneo/metabolismo , Animales , Anomalías Cardiovasculares/inducido químicamente , Anomalías Cardiovasculares/metabolismo , Desinfectantes/metabolismo , Femenino , Humanos , Recién Nacido de Bajo Peso/crecimiento & desarrollo , Recién Nacido de Bajo Peso/metabolismo , Recién Nacido , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/metabolismo , Embarazo , Medición de Riesgo , Purificación del Agua/métodos , Abastecimiento de Agua/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA