Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 34(9): 2023-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24990230

RESUMEN

OBJECTIVE: Reactive oxygen species (ROS) act as signaling molecules during angiogenesis; however, the mechanisms used for such signaling events remain unclear. Stromal cell-derived factor-1α (SDF-1α) is one of the most potent angiogenic chemokines. Here, we examined the role of ROS in the regulation of SDF-1α-dependent angiogenesis. APPROACH AND RESULTS: Bovine aortic endothelial cells were treated with SDF-1α, and intracellular ROS generation was monitored. SDF-1α treatment induced bovine aortic endothelial cell migration and ROS generation, with the majority of ROS generated by bovine aortic endothelial cells at the leading edge of the migratory cells. Antioxidants and nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitors blocked SDF-1α-induced endothelial migration. Furthermore, knockdown of either NOX5 or p22phox (a requisite subunit for NOX1/2/4 activation) significantly impaired endothelial motility and tube formation, suggesting that multiple NOXs regulate SDF-1α-dependent angiogenesis. Our previous study demonstrated that c-Jun N-terminal kinase 3 activity is essential for SDF-1α-dependent angiogenesis. Here, we identified that NOX5 is the dominant NOX required for SDF-1α-induced c-Jun N-terminal kinase 3 activation and that NOX5 and MAP kinase phosphatase 7 (MKP7; the c-Jun N-terminal kinase 3 phosphatase) associate with one another but decrease this interaction on SDF-1α treatment. Furthermore, MKP7 activity was inhibited by SDF-1α, and this inhibition was relieved by NOX5 knockdown, indicating that NOX5 promotes c-Jun N-terminal kinase 3 activation by blocking MKP7 activity. CONCLUSIONS: We conclude that NOX is required for SDF-1α signaling and that intracellular redox balance is critical for SDF-1α-induced endothelial migration and angiogenesis.


Asunto(s)
Quimiocina CXCL12/fisiología , Proteínas de la Membrana/fisiología , NADPH Oxidasas/fisiología , Neovascularización Fisiológica/fisiología , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Aorta , Azoles/farmacología , Bovinos , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Fosfatasas de Especificidad Dual/fisiología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Técnicas de Silenciamiento del Gen , Hiperglucemia/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/fisiología , Isoindoles , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteína Quinasa 10 Activada por Mitógenos/fisiología , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/fisiología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Neovascularización Fisiológica/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Oxidación-Reducción , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
2.
Hum Mol Genet ; 23(4): 1013-24, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24113144

RESUMEN

Gordon Holmes syndrome (GHS) is a rare Mendelian neurodegenerative disorder characterized by ataxia and hypogonadism. Recently, it was suggested that disordered ubiquitination underlies GHS though the discovery of exome mutations in the E3 ligase RNF216 and deubiquitinase OTUD4. We performed exome sequencing in a family with two of three siblings afflicted with ataxia and hypogonadism and identified a homozygous mutation in STUB1 (NM_005861) c.737C→T, p.Thr246Met, a gene that encodes the protein CHIP (C-terminus of HSC70-interacting protein). CHIP plays a central role in regulating protein quality control, in part through its ability to function as an E3 ligase. Loss of CHIP function has long been associated with protein misfolding and aggregation in several genetic mouse models of neurodegenerative disorders; however, a role for CHIP in human neurological disease has yet to be identified. Introduction of the Thr246Met mutation into CHIP results in a loss of ubiquitin ligase activity measured directly using recombinant proteins as well as in cell culture models. Loss of CHIP function in mice resulted in behavioral and reproductive impairments that mimic human ataxia and hypogonadism. We conclude that GHS can be caused by a loss-of-function mutation in CHIP. Our findings further highlight the role of disordered ubiquitination and protein quality control in the pathogenesis of neurodegenerative disease and demonstrate the utility of combining whole-exome sequencing with molecular analyses and animal models to define causal disease polymorphisms.


Asunto(s)
Anomalías Múltiples/enzimología , Ataxia Cerebelosa/enzimología , Hormona Liberadora de Gonadotropina/deficiencia , Hipogonadismo/enzimología , Ubiquitina-Proteína Ligasas/genética , Anomalías Múltiples/genética , Adolescente , Secuencia de Aminoácidos , Animales , Células COS , Ataxia Cerebelosa/genética , Cerebelo/metabolismo , Cerebelo/patología , Chlorocebus aethiops , Femenino , Estudios de Asociación Genética , Hormona Liberadora de Gonadotropina/genética , Humanos , Hipogonadismo/genética , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Ubiquitina-Proteína Ligasas/deficiencia , Adulto Joven
3.
Circ Res ; 111(5): 564-74, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22777006

RESUMEN

RATIONALE: Among the extracellular modulators of Bmp (bone morphogenetic protein) signaling, Bmper (Bmp endothelial cell precursor-derived regulator) both enhances and inhibits Bmp signaling. Recently we found that Bmper modulates Bmp4 activity via a concentration-dependent, endocytic trap-and-sink mechanism. OBJECTIVE: To investigate the molecular mechanisms required for endocytosis of the Bmper/Bmp4 and signaling complex and determine the mechanism of Bmper's differential effects on Bmp4 signaling. METHODS AND RESULTS: Using an array of biochemical and cell biology techniques, we report that LRP1 (LDL receptor-related protein 1), a member of the LDL receptor family, acts as an endocytic receptor for Bmper and a coreceptor of Bmp4 to mediate the endocytosis of the Bmper/Bmp4 signaling complex. Furthermore, we demonstrate that LRP1-dependent Bmper/Bmp4 endocytosis is essential for Bmp4 signaling, as evidenced by the phenotype of lrp1-deficient zebrafish, which have abnormal cardiovascular development and decreased Smad1/5/8 activity in key vasculogenic structures. CONCLUSIONS: Together, these data reveal a novel role for LRP1 in the regulation of Bmp4 signaling by regulating receptor complex endocytosis. In addition, these data introduce LRP1 as a critical regulator of vascular development. These observations demonstrate Bmper's ability to fine-tune Bmp4 signaling at the single-cell level, unlike the spatial regulatory mechanisms applied by other Bmp modulators.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/metabolismo , Endocitosis/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Portadoras/genética , Línea Celular , Movimiento Celular/fisiología , Células Endoteliales/citología , Células HEK293 , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Fenotipo , ARN Interferente Pequeño/genética , Receptores de LDL/genética , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/genética , Pez Cebra , Proteínas de Pez Cebra/genética
4.
J Mol Cell Cardiol ; 52(3): 526-37, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22085703

RESUMEN

Awareness of the regulation of cell signaling by post-translational ubiquitination has emerged over the past 2 decades. Like phosphorylation, post-translational modification of proteins with ubiquitin can result in the regulation of numerous cellular functions, for example, the DNA damage response, apoptosis, cell growth, and the innate immune response. In this review, we discuss recently published mechanisms by which the ubiquitin proteasome system regulates key signal transduction pathways in the heart, including MAPK JNK, calcineurin, FOXO, p53, and estrogen receptors α and ß. We then explore how ubiquitin proteasome system-specific regulation of these signal transduction pathways plays a role in the pathophysiology of common cardiac diseases, such as cardiac hypertrophy, heart failure, ischemia reperfusion injury, and diabetes. This article is part of a Special Section entitled "Post-translational Modification."


Asunto(s)
Miocardio/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Animales , Calcineurina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Corazón/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Receptores de Estrógenos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
5.
Circ Res ; 109(4): 453-62, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21817165

RESUMEN

Cardiac hypertrophy develops most commonly in response to hypertension and is an independent risk factor for the development of heart failure. The mechanisms by which cardiac hypertrophy may be reversed to reduce this risk have not been fully determined to the point where mechanism-specific therapies have been developed. Recently, proteases in the calpain family have been implicated in the regulation of the development of cardiac hypertrophy in preclinical animal models. In this review, we summarize the molecular mechanisms by which calpain inhibition has been shown to modulate the development of cardiac (specifically ventricular) hypertrophy. The context within which calpain inhibition might be developed for therapeutic intervention of cardiac hypertrophy is then discussed.


Asunto(s)
Calpaína/metabolismo , Cardiomegalia/enzimología , Ventrículos Cardíacos/enzimología , Remodelación Ventricular , Animales , Calpaína/antagonistas & inhibidores , Cardiomegalia/patología , Cardiomegalia/prevención & control , Fármacos Cardiovasculares/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Modelos Animales de Enfermedad , Diseño de Fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Humanos , Transducción de Señal/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
6.
Cell Biochem Funct ; 29(4): 334-41, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21506136

RESUMEN

During vascular development, endothelial cells are exposed to a variety of rapidly changing factors, including fluctuating oxygen levels. We have previously shown that ankyrin repeat and suppressor of cytokine signalling box protein 4 (ASB4) is the most highly differentially expressed gene in the vascular lineage during early differentiation and is expressed in the embryonic vasculature at a time when oxygen tension is rising because of the onset of placental blood flow. To further our understanding of the regulation of ASB4 expression in endothelial cells, we tested the effect of various stressors for their ability to alter ASB4 expression in the immortalized murine endothelial cell lines MS1 and SVR. ASB4 expression is decreased during hypoxic insult and shear stress, whereas it is increased in response to tumour necrosis factor alpha (TNF-α). Further investigation indicated that nuclear factor kappa B (NF-κB) is the responsible transcription factor involved in the TNF-α-induced upregulation of ASB4, placing ASB4 downstream of NF-κB in the TNF-α signalling cascade and identifying it as a potential regulator for TNF-α's numerous functions associated with inflammation, angiogenesis and apoptosis.


Asunto(s)
Endotelio Vascular/metabolismo , Oxígeno/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular Transformada , Regulación del Desarrollo de la Expresión Génica , Hipoxia/metabolismo , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Estrés Mecánico , Transfección
7.
J Biol Chem ; 286(12): 9929-34, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21257759

RESUMEN

Proteolysis within the cardiac sarcomere is a constantly evolving area of research. Three major pathways of proteolysis have been identified as being active within the cardiac sarcomere, namely the ubiquitin-proteasome system, autophagy, and the calpain system. The role of ubiquitin-proteasome system-mediated proteolysis in cardiovascular health and disease has been known for some time; however, it is now apparent that other proteolytic systems also aid in the stabilization of cardiac sarcomere structure and function. This minireview focuses on the individual as well as cooperative involvement of each of these three major pathways of proteolysis within the cardiac sarcomere.


Asunto(s)
Proteínas Musculares/metabolismo , Miocardio/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sarcómeros/metabolismo , Ubiquitina/metabolismo , Animales , Humanos
8.
Sci Signal ; 3(122): pe17, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20484115

RESUMEN

A healthy vasculature is an essential component of development and is regulated by different signaling pathways. One of the most critical pathways involved is the vascular endothelial growth factor (VEGF) pathway. Components of this pathway serve as the first marker of primitive endothelial cells and are instrumental in inducing the initial differentiation of endothelial cells and later refining them into either arteries or veins. However, the regulation of VEGF signaling remains a mystery, with most studies focusing on the downstream components of this signaling cascade. New evidence shows that the protein cerebral cavernous malformation 3 (CCM3) is a key regulator of the VEGF pathway, bringing to light a previously unknown component of the VEGF signaling axis and opening the door to an exciting new era of vasculogenic research.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Ratones , Ratones Noqueados , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(14): 5675-80, 2009 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-19307591

RESUMEN

The chemokine stromal cell-derived factor-1alpha (SDF-1alpha) is a pivotal player in angiogenesis. It is capable of influencing such cellular processes as tubulogenesis and endothelial cell migration, yet very little is known about the actual signaling events that mediate SDF-1alpha-induced endothelial cell function. In this report, we describe the identification of an intricate SDF-1alpha-induced signaling cascade that involves endothelial nitric oxide synthase (eNOS), JNK3, and MAPK phosphatase 7 (MKP7). We demonstrate that the SDF-1alpha-induced activation of JNK3, critical for endothelial cell migration, depends on the prior activation of eNOS. Specifically, activation of eNOS leads to production of NO and subsequent nitrosylation of MKP7, rendering the phosphatase inactive and unable to inhibit the activation of JNK3. These observations reinforce the importance of nitric oxide and S-nitrosylation in angiogenesis and provide a mechanistic pathway for SDF-1alpha-induced endothelial cell migration. In addition, the discovery of this interactive network of pathways provides novel and unexpected therapeutic targets for angiogenesis-dependent diseases.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/fisiología , Fosfatasas de Especificidad Dual/metabolismo , Endotelio Vascular/citología , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Bovinos , Células Cultivadas , Células Endoteliales , Humanos , Transducción de Señal
10.
Cardiovasc Res ; 81(3): 439-48, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18974044

RESUMEN

The assembly and maintenance of the cardiac sarcomere, which contains the basic contractile components of actin and myosin, are essential for cardiac function. While often described as a static structure, the sarcomere is actually dynamic and undergoes constant turnover, allowing it to adapt to physiological changes while still maintaining function. A host of new factors have been identified that play a role in the regulation of protein quality control in the sarcomere, including chaperones that mediate the assembly of sarcomere components and ubiquitin ligases that control their specific degradation. There is clear evidence of sarcomere disorganization in animal models lacking muscle-specific chaperone proteins, illustrating the importance of these molecules in sarcomere structure and function. Although ubiquitin ligases have been found within the sarcomere structure itself, the role of the ubiquitin proteasome system in cardiac sarcomere regulation, and the factors that control its activity, are only just now being elucidated. The number of ubiquitin ligases identified with specificity for sarcomere proteins, each with distinct target substrates, is growing, allowing for tight regulation of this system. In this review, we highlight the dynamic interplay between sarcomere-specific chaperones and ubiquitin-dependent degradation of sarcomere proteins that is necessary in order to maintain structure and function of the cardiac sarcomere.


Asunto(s)
Cardiomiopatías/metabolismo , Contracción Muscular , Proteínas Musculares/sangre , Miocardio/metabolismo , Sarcómeros/metabolismo , Actinas/metabolismo , Adaptación Fisiológica , Animales , Autofagia , Proteínas de Caenorhabditis elegans/metabolismo , Calpaína/metabolismo , Cardiomiopatías/fisiopatología , Desmina/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Miocardio/enzimología , Miosinas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sarcómeros/enzimología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Cadena B de alfa-Cristalina/metabolismo
11.
Circ Res ; 103(9): 929-39, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18948631

RESUMEN

The molecular differences between arteries and veins are genetically predetermined and are evident even before the first embryonic heart beat. Although ephrinB2 and EphB4 are expressed in cells that will ultimately differentiate into arteries and veins, respectively, many other genes have been shown to play a significant role in cell fate determination. The expression patterns of ephrinB2 and EphB4 are restricted to arterial-venous boundaries, and Eph/ephrin signaling provides repulsive cues at arterial-venous boundaries that are thought to prevent intermixing of arterial- and venous-fated cells. However, the maintenance of arterial-venous fate is susceptible to some degree of plasticity. Thus, in response to signals from the ambient microenvironment and shear stress, there is flow-mediated intercalation of the arteries and veins that ultimately leads to the formation of a functional, closed-loop circulation. In addition, cells in the blood vessels of each organ undergo epigenetic, morphological, and functional adaptive changes that are specific to the proximate function of their cognate organ(s). These adaptive changes result in an interorgan and intraorgan vessel heterogeneity that manifest clinically in a disparate response of different organs to identical risk factors and injury in the same animal. In this review, we focus on the molecular and physiological factors influencing arterial-venous heterogeneity between and within different organ(s). We explore arterial-venous differences in selected organs, as well as their respective endothelial cell architectural organization that results in their inter- and intraorgan heterogeneity.


Asunto(s)
Arterias/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Venas/metabolismo , Animales , Arterias/embriología , Arterias/crecimiento & desarrollo , Diferenciación Celular , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Hemodinámica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neovascularización Fisiológica , Transducción de Señal/genética , Venas/embriología , Venas/crecimiento & desarrollo
12.
Mol Cell Biol ; 28(12): 4018-25, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18411298

RESUMEN

During the course of biological aging, there is a gradual accumulation of damaged proteins and a concomitant functional decline in the protein degradation system. Protein quality control is normally ensured by the coordinated actions of molecular chaperones and the protein degradation system that collectively help to maintain protein homeostasis. The carboxyl terminus of Hsp70-interacting protein (CHIP), a ubiquitin ligase/cochaperone, participates in protein quality control by targeting a broad range of chaperone substrates for proteasome degradation via the ubiquitin-proteasome system, demonstrating a broad involvement of CHIP in maintaining cytoplasmic protein quality control. In the present study, we have investigated the influence that protein quality control exerts on the aging process by using CHIP-/- mice. CHIP deficiency in mice leads to a markedly reduced life span, along with accelerated age-related pathophysiological phenotypes. These features were accompanied by indications of accelerated cellular senescence and increased indices of oxidative stress. In addition, CHIP-/- mice exhibit a deregulation of protein quality control, as indicated by elevated levels of toxic oligomer proteins and a decline in proteasome activity. Taken together, these data reveal that impaired protein quality control contributes to cellular senescence and implicates CHIP-dependent quality control mechanisms in the regulation of mammalian longevity in vivo.


Asunto(s)
Envejecimiento , Ubiquitina-Proteína Ligasas/fisiología , Animales , Encéfalo/metabolismo , Senescencia Celular , Citoplasma/metabolismo , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Oxígeno/química , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/genética
13.
Biol Neonate ; 89(3): 147-58, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16210849

RESUMEN

Beta-adrenergic receptor (betaAR) activation has been shown to maintain heart rate during hypoxia and to rescue the fetus from the fetal lethality that occurs in the absence of norepinephrine. This study examines whether the same subtype of betaAR is responsible for survival and heart rate regulation. It also investigates which betaARs are located on the early fetal heart and whether they can be directly activated during hypoxia. Cultured E12.5 mouse fetuses were treated with subtype-specific betaAR antagonists to pharmacologically block betaARs during a hypoxic insult. Hypoxia alone reduced heart rate by 35-40% compared to prehypoxic levels. During hypoxia, heart rate was further reduced by 31% in the presence of a beta(1)AR antagonist, CGP20712A, at 100 nM, but not with a beta2 (ICI118551)- or a beta3 (SR59230A)-specific antagonist at 100 nM. Survival in utero was also mediated by beta1ARs. A beta1 partial agonist, xamoterol, rescued 74% of catecholamine-deficient (tyrosine-hydroxylase-null) pups to birth, a survival rate equivalent to that with a nonspecific betaAR agonist, isoproterenol (87%). Receptor autoradiography showed that beta1ARs were only found on the mouse heart at E12.5, while beta2ARs were localized to the liver and vasculature. To determine if the response to hypoxia was intrinsic to the heart, isolated fetal hearts were incubated under hypoxic conditions in the presence of a betaAR agonist. Heart rate was reduced to 25-30% by hypoxia alone, but was restored to 63% of prehypoxic levels with 100 nM isoproterenol. Restoration was completely prevented if beta1ARs were blocked with CGP20712A at 300 nM, a concentration that blocks beta1ARs, but not beta2- or beta3ARs. Our results demonstrate that beta1ARs are located on the heart of early fetal mice and that beta1AR stimulation maintains fetal heart rate during hypoxia and mediates survival in vivo.


Asunto(s)
Hipoxia Fetal/mortalidad , Frecuencia Cardíaca Fetal , Receptores Adrenérgicos beta 1/fisiología , Antagonistas Adrenérgicos beta/farmacología , Animales , Corazón Fetal/fisiopatología , Frecuencia Cardíaca Fetal/efectos de los fármacos , Imidazoles/farmacología , Isoproterenol/farmacología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Técnicas de Cultivo de Órganos , Oxígeno/análisis , Tasa de Supervivencia , Tirosina 3-Monooxigenasa/deficiencia
14.
Am J Physiol Heart Circ Physiol ; 284(6): H2069-77, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12574001

RESUMEN

Mice lacking catecholamines die before birth, some with cardiovascular abnormalities. To investigate the role of catecholamines in development, embryonic day 12.5 (E12.5) fetuses were cultured and heart rate monitored. Under optimal oxygenation, wild-type and catecholamine-deficient fetuses had the same initial heart rate (200-220 beats/min), which decreased by 15% in wild-type fetuses during 50 min of culture. During the same culture period, catecholamine-deficient fetuses dropped their heart rate by 35%. Hypoxia reduced heart rate of wild-type fetuses by 35-40% in culture and by 20% in utero, assessed by echocardiography. However, catecholamine-deficient fetuses exhibited greater hypoxia-induced bradycardia, reducing their heart rate by 70-75% in culture. Isoproterenol, a beta-adrenergic receptor (beta-AR) agonist, reversed this extreme bradycardia, restoring the rate of catecholamine-deficient fetuses to that of nonmutant siblings. Moreover, isoproterenol rescued 100% of catecholamine-deficient pups to birth in a dose-dependent, stereo-specific manner when administered in the dam's drinking water. An alpha-AR agonist was without effect. When wild-type fetuses were cultured with adrenoreceptor antagonists to create pharmacological nulls, blockade of alpha-ARs with 10 microM phentolamine or beta-ARs with 10 microM bupranolol alone or in combination did not reduce heart rate under optimal oxygenation. However, when combined with hypoxia, beta-AR blockade reduced heart rate by 35%. In contrast, the muscarinic blocker atropine and the alpha-AR antagonist phentolamine had no effect. These data suggest that beta-ARs mediate survival in vivo and regulate heart rate in culture. We hypothesize that norepinephrine, acting through beta-ARs, maintains fetal heart rate during periods of transient hypoxia that occur throughout gestation, and that catecholamine-deficient fetuses die because they cannot withstand hypoxia-induced bradycardia.


Asunto(s)
Catecolaminas/fisiología , Feto/fisiología , Frecuencia Cardíaca Fetal/fisiología , Receptores Adrenérgicos beta/fisiología , Sobrevida/fisiología , Antagonistas Adrenérgicos beta/farmacología , Animales , Animales Recién Nacidos , Vasos Sanguíneos/patología , Dopamina beta-Hidroxilasa/genética , Dopamina beta-Hidroxilasa/fisiología , Ecocardiografía Doppler , Epinefrina/fisiología , Femenino , Hipoxia/fisiopatología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Norepinefrina/fisiología , Técnicas de Cultivo de Órganos , Embarazo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...