Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 42(7): 886-902, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35477279

RESUMEN

BACKGROUND: The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. METHODS: We generated mice deficient in DOCK4' used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells' used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. RESULTS: Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. CONCLUSIONS: Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.


Asunto(s)
Células Endoteliales , Proteínas de Unión al GTP rho , Uniones Adherentes/metabolismo , Animales , Permeabilidad Capilar/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Pulmón/metabolismo , Ratones , Proteínas de Unión al GTP rho/metabolismo
2.
Circ Res ; 117(2): 166-77, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-26034041

RESUMEN

RATIONALE: Human and murine Vcam1 promoters contain 2 adjacent nuclear factor-κB (NF-κB)-binding elements. Both are essential for cytokine-induced transcription of transiently transfected promoter-reporter constructs. However, the relevance of these insights to regulation of the endogenous Vcam1 gene and to pathophysiological processes in vivo remained unknown. OBJECTIVE: Determine the role of the 5' NF-κB-binding element in expression of the endogenous Vcam1 gene. METHODS AND RESULTS: Homologous recombination in embryonic stem cells was used to inactivate the 5' NF-κB element in the Vcam1 promoter and alter 3 nucleotides in the 5' untranslated region to allow direct comparison of wild-type versus mutant allele RNA expression and chromatin configuration in heterozygous mice. Systemic treatment with inflammatory cytokines or endotoxin (lipopolysaccharide) induced lower expression of the mutant allele relative to wild-type by endothelial cells in the aorta, heart, and lungs. The mutant allele also showed lower endothelial expression in 2-week atherosclerotic lesions in Vcam1 heterozygous/low-density lipoprotein receptor-deficient mice fed a cholesterol-rich diet. In vivo chromatin immunoprecipitation assays of heart showed diminished lipopolysaccharide-induced association of RNA polymerase 2 and NF-κB p65 with the mutant promoter. In contrast, expression of mutant and wild-type alleles was comparable in intimal cells of wire-injured carotid artery and 4- to 12-week atherosclerotic lesions. CONCLUSIONS: This study highlights differences between in vivo and in vitro promoter analyses, and reveals a differential role for a NF-κB transcriptional response element in endothelial vascular cell adhesion molecule-1 expression induced by inflammatory cytokines or a cholesterol-rich diet versus intimal cell expression in atherosclerotic lesions and injured arteries.


Asunto(s)
Regiones no Traducidas 5'/genética , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Elementos de Respuesta/efectos de la radiación , Factor de Transcripción ReIA/metabolismo , Túnica Íntima/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Animales , Aterosclerosis/etiología , Aterosclerosis/patología , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Colesterol en la Dieta/efectos adversos , Selectina E/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Mapeo de Interacción de Proteínas , ARN Polimerasa II/metabolismo , Receptores de LDL/deficiencia , Elementos de Respuesta/genética , Transcripción Genética , Túnica Íntima/patología , Molécula 1 de Adhesión Celular Vascular/biosíntesis
3.
J Pathol Inform ; 13: 8, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35136675

RESUMEN

BACKGROUND: Digital pathology operations that precede viewing by a pathologist have a substantial impact on costs and fidelity of the digital image. Scan time and file size determine throughput and storage costs, whereas tissue omission during digital capture ("dropouts") compromises downstream interpretation. We compared how these variables differ across scanners. METHODS: A 212 slide set randomly selected from a gynecologic-gestational pathology practice was used to benchmark scan time, file size, and image completeness. Workflows included the Hamamatsu S210 scanner (operated under default and optimized profiles) and the Leica GT450. Digital tissue dropouts were detected by the aligned overlay of macroscopic glass slide camera images (reference) with images created by the slide scanners whole slide images. RESULTS: File size and scan time were highly correlated within each platform. Differences in GT450, default S210, and optimized S210 performance were seen in average file size (1.4 vs. 2.5 vs. 3.4 GB) and scan time (93 vs. 376 vs. 721 s). Dropouts were seen in 29.5% (186/631) of successful scans overall: from a low of 13.7% (29/212) for the optimized S210 profile, followed by 34.6% (73/211) for the GT450 and 40.4% (84/208) for the default profile S210 profile. Small dislodged fragments, "shards," were dropped in 22.2% (140/631) of slides, followed by tissue marginalized at the glass slide edges, 6.2% (39/631). "Unique dropouts," those for which no equivalent appeared elsewhere in the scan, occurred in only three slides. Of these, 67% (2/3) were "floaters" or contaminants from other cases. CONCLUSIONS: Scanning speed and resultant file size vary greatly by scanner type, scanner operation settings, and clinical specimen mix (tissue type, tissue area). Digital image fidelity as measured by tissue dropout frequency and dropout type also varies according to the tissue type and scanner. Dropped tissues very rarely (1/631) represent actual specimen tissues that are not represented elsewhere in the scan, so in most cases cannot alter the diagnosis. Digital pathology platforms vary in their output efficiency and image fidelity to the glass original and should be matched to the intended application.

4.
J Clin Invest ; 117(3): 812-22, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17304352

RESUMEN

We rescued the embryonic lethality of global PPARgamma knockout by breeding Mox2-Cre (MORE) mice with floxed PPARgamma mice to inactivate PPARgamma in the embryo but not in trophoblasts and created a generalized PPARgamma knockout mouse model, MORE-PPARgamma knockout (MORE-PGKO) mice. PPARgamma inactivation caused severe lipodystrophy and insulin resistance; surprisingly, it also caused hypotension. Paradoxically, PPARgamma agonists had the same effect. We showed that another mouse model of lipodystrophy was hypertensive, ruling out the lipodystrophy as a cause. Further, high salt loading did not correct the hypotension in MORE-PGKO mice. In vitro studies showed that the vasculature from MORE-PGKO mice was more sensitive to endothelial-dependent relaxation caused by muscarinic stimulation, but was not associated with changes in eNOS expression or phosphorylation. In addition, vascular smooth muscle had impaired contraction in response to alpha-adrenergic agents. The renin-angiotensin-aldosterone system was mildly activated, consistent with increased vascular capacitance or decreased volume. These effects are likely mechanisms contributing to the hypotension. Our results demonstrated that PPARgamma is required to maintain normal adiposity and insulin sensitivity in adult mice. Surprisingly, genetic loss of PPARgamma function, like activation by agonists, lowered blood pressure, likely through a mechanism involving increased vascular relaxation.


Asunto(s)
Muerte Fetal/genética , Hipotensión/genética , Resistencia a la Insulina/genética , Lipodistrofia/genética , PPAR gamma/fisiología , Animales , Femenino , Lipodistrofia/patología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR gamma/deficiencia , PPAR gamma/genética , Fosforilación
5.
J Pathol Inform ; 9: 37, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30533276

RESUMEN

BACKGROUND: Digital Imaging and Communications in Medicine (DICOM®) is the standard for the representation, storage, and communication of medical images and related information. A DICOM file format and communication protocol for pathology have been defined; however, adoption by vendors and in the field is pending. Here, we implemented the essential aspects of the standard and assessed its capabilities and limitations in a multisite, multivendor healthcare network. METHODS: We selected relevant DICOM attributes, developed a program that extracts pixel data and pixel-related metadata, integrated patient and specimen-related metadata, populated and encoded DICOM attributes, and stored DICOM files. We generated the files using image data from four vendor-specific image file formats and clinical metadata from two departments with different laboratory information systems. We validated the generated DICOM files using recognized DICOM validation tools and measured encoding, storage, and access efficiency for three image compression methods. Finally, we evaluated storing, querying, and retrieving data over the web using existing DICOM archive software. RESULTS: Whole slide image data can be encoded together with relevant patient and specimen-related metadata as DICOM objects. These objects can be accessed efficiently from files or through RESTful web services using existing software implementations. Performance measurements show that the choice of image compression method has a major impact on data access efficiency. For lossy compression, JPEG achieves the fastest compression/decompression rates. For lossless compression, JPEG-LS significantly outperforms JPEG 2000 with respect to data encoding and decoding speed. CONCLUSION: Implementation of DICOM allows efficient access to image data as well as associated metadata. By leveraging a wealth of existing infrastructure solutions, the use of DICOM facilitates enterprise integration and data exchange for digital pathology.

6.
Circ Res ; 97(4): 372-9, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-16051889

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)-gamma is required for adipogenesis but is also found in the cardiovascular system, where it has been proposed to oppose inflammatory pathways and act as a growth suppressor. PPAR-gamma agonists, thiazolidinediones (TZDs), inhibit cardiomyocyte growth in vitro and in pressure overload models. Paradoxically, TZDs also induce cardiac hypertrophy in animal models. To directly determine the role of cardiomyocyte PPAR-gamma, we have developed a cardiomyocyte-specific PPAR-gamma-knockout (CM-PGKO) mouse model. CM-PGKO mice developed cardiac hypertrophy with preserved systolic cardiac function. Treatment with a TZD, rosiglitazone, induced cardiac hypertrophy in both littermate control mice and CM-PGKO mice and activated distinctly different hypertrophic pathways from CM-PGKO. CM-PGKO mice were found to have increased expression of cardiac embryonic genes (atrial natriuretic peptide and beta-myosin heavy chain) and elevated nuclear factor kappaB activity in the heart, effects not found by rosiglitazone treatment. Rosiglitazone increased cardiac phosphorylation of p38 mitogen-activated protein kinase independent of PPAR-gamma, whereas rosiglitazone induced phosphorylation of extracellular signal-related kinase 1/2 in the heart dependent of PPAR-gamma. Phosphorylation of c-Jun N-terminal kinases was not affected by rosiglitazone or CM-PGKO. Surprisingly, despite hypertrophy, Akt phosphorylation was suppressed in CM-PGKO mouse heart. These data show that cardiomyocyte PPAR-gamma suppresses cardiac growth and embryonic gene expression and inhibits nuclear factor kappaB activity in vivo. Further, rosiglitazone causes cardiac hypertrophy at least partially independent of PPAR-gamma in cardiomyocytes and through different mechanisms from CM-PGKO.


Asunto(s)
Cardiomegalia/etiología , Miocitos Cardíacos/fisiología , PPAR gamma/fisiología , Tiazolidinedionas/farmacología , Animales , Factor Natriurético Atrial/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Cadenas Pesadas de Miosina/genética , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , PPAR gamma/agonistas , PPAR gamma/genética , Fosforilación , Rosiglitazona , Sístole
7.
Mol Cell Biol ; 24(18): 8134-44, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15340074

RESUMEN

Integrin-linked kinase (ILK) is a phosphoinositide 3-kinase-dependent serine/threonine kinase that interacts with beta integrins. Here we show that endothelial cell (EC)-specific deletion of ILK in mice confers placental insufficiency with decreased labyrinthine vascularization, yielding no viable offspring. Deletion of ILK in zebra fish using antisense morpholino oligonucleotides results in marked patterning abnormalities of the vasculature and is similarly lethal. To dissect potential mechanisms responsible for these phenotypes, we performed ex vivo deletion of ILK from purified EC of adult mice. We observed downregulation of the active-conformation of beta1 integrins with a striking increase in EC apoptosis associated with activation of caspase 9. There was also reduced phosphorylation of the ILK kinase substrate, Akt. However, phenotypic rescue of ILK-deficient EC by wild-type ILK, but not by a constitutively active mutant of Akt, suggests regulation of EC survival by ILK in an Akt-independent manner. Thus, endothelial ILK plays a critical role in vascular development through integrin-matrix interactions and EC survival. These data have important implications for both physiological and pathological angiogenesis.


Asunto(s)
Endotelio Vascular/citología , Endotelio Vascular/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Supervivencia Celular , Células Cultivadas , ADN Complementario/genética , Endotelio Vascular/embriología , Femenino , Eliminación de Gen , Ratones , Ratones Transgénicos , Embarazo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
8.
PLoS One ; 11(8): e0160636, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27505464

RESUMEN

Peroxisome proliferator activated receptor gamma (PPARγ) is a pleiotropic ligand activated transcription factor that acts in several tissues to regulate adipocyte differentiation, lipid metabolism, insulin sensitivity and glucose homeostasis. PPARγ also regulates cardiomyocyte homeostasis and by virtue of its obligate role in placental development is required for embryonic survival. To determine the postnatal functions of PPARγ in vivo we studied globally deficient neonatal mice produced by epiblast-restricted elimination of PPARγ. PPARγ-rescued placentas support development of PPARγ-deficient embryos that are viable and born in near normal numbers. However, PPARγ-deficient neonatal mice show severe lipodystrophy, lipemia, hepatic steatosis with focal hepatitis, relative insulin deficiency and diabetes beginning soon after birth and culminating in failure to thrive and neonatal lethality between 4 and 10 days of age. These abnormalities are not observed with selective PPARγ2 deficiency or with deficiency restricted to hepatocytes, skeletal muscle, adipocytes, cardiomyocytes, endothelium or pancreatic beta cells. These observations suggest important but previously unappreciated functions for PPARγ1 in the neonatal period either alone or in combination with PPARγ2 in lipid metabolism, glucose homeostasis and insulin sensitivity.


Asunto(s)
Diabetes Mellitus/metabolismo , Insulina/sangre , Lipodistrofia/metabolismo , PPAR gamma/deficiencia , Animales , Animales Recién Nacidos , Femenino , Estratos Germinativos/metabolismo , Hepatitis/complicaciones , Hepatomegalia/complicaciones , Homeostasis , Hiperlipidemias/complicaciones , Hiperlipoproteinemias/complicaciones , Resistencia a la Insulina , Cetosis/complicaciones , Lipodistrofia/complicaciones , Ratones , Necrosis , Placenta/metabolismo , Embarazo
9.
Stem Cells Dev ; 22(21): 2815-24, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23767827

RESUMEN

Tissue oxygen tension regulates differentiation of multiple types of stem cells. In the placenta, hypoxia has been associated with abnormal trophoblast differentiation and placental insufficiency syndromes of preeclampsia (PE) and intrauterine growth restriction (IUGR). Peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated transcription factor involved in many cellular processes, including differentiation. We have previously shown that PPARγ-null trophoblast stem (TS) cells show a defect in differentiation to labyrinthine trophoblast, instead differentiating preferentially to trophoblast giant cells (TGC). Since PPARγ is known to be regulated by hypoxia in adipose tissue, we hypothesized that there may be a link between oxygen tension, PPARγ expression, and trophoblast differentiation. We found that hypoxia reduced PPARγ expression by a mechanism independent of both hypoxia-inducible factor (HIF) and histone deacetylases (HDACs). In addition, PPARγ partially rescued hypoxia-induced inhibition of labyrinthine differentiation in wild-type TS cells but was not required for hypoxia-induced inhibition of TGC differentiation. Finally, we show that induction of labyrinthine trophoblast differentiation by HDAC inhibitor treatment is independent of both PPARγ and Gcm1. We propose a model with two pathways for labyrinthine trophoblast differentiation of TS cells, one of which is dependent on PPARγ and inhibited by hypoxia. Since hypoxia is associated with PE and IUGR, we propose that PPARγ may at least partially mediate hypoxia-induced placental insufficiency and as such may be a promising therapeutic target for these disorders.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , PPAR gamma/genética , Trofoblastos/citología , Trofoblastos/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Western Blotting , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Células Cultivadas , Cicloheximida/farmacología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , PPAR gamma/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Inhibidores de la Síntesis de la Proteína/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
10.
Am J Reprod Immunol ; 66(3): 209-22, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21385270

RESUMEN

PROBLEM: Intrauterine bacterial infection during pregnancy may lead to adverse outcome. The objective of this study was to assess whether peptidoglycan (PGN) derived from Gram-positive bacteria induces trophoblast stem (TS) cell death or alters TS cell cytokine production. METHOD OF STUDY: Toll-like receptor (TLR) transcript expression was assessed by RT-PCR. Protein expression was determined by confocal microscopy or flow cytometry. 7-Aminoactinomycin D (7-AAD) staining was used to assess TS cell death. Morphological features of cell death were evaluated by transmission electron microscopy. The presence of cleaved caspase-3 and high mobility group box 1 (HMGB1) protein was examined by Western blot. Cytokine levels in cell supernatants were determined using a mouse cytokine 23-plex panel. RESULTS: Toll-like receptor 2 and TLR4 protein was expressed from the 1-cell stage through the blastocyst stage of murine embryo development. Murine TS cells expressed TLR2 and TLR6 but not TLR1 or TLR4 RNA. Only TLR2 protein was detected at the plasma membrane of TS cells. PGN induced TS cell death by a caspase-3-independent mechanism. The cell death pathway induced by PGN was morphologically consistent with necrosis. Finally, PGN induced HMGB1 release and increased MIP-1ß secretion while inhibiting the constitutive release of RANTES. CONCLUSION: Peptidoglycan-induced TS cell necrosis and the subsequent release of HMGB1 and MIP-1ß may regulate an infection-induced inflammatory response at the maternal-fetal interface and thus may play a role in the pathogenesis of infection-associated pregnancy complications.


Asunto(s)
Citocinas/inmunología , Peptidoglicano/farmacología , Células Madre/efectos de los fármacos , Células Madre/inmunología , Trofoblastos/efectos de los fármacos , Trofoblastos/inmunología , Animales , Caspasa 3/inmunología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/inmunología , Citocinas/biosíntesis , Femenino , Masculino , Ratones , Necrosis/inducido químicamente , Necrosis/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/inmunología , Embarazo , Receptor Toll-Like 1/biosíntesis , Receptor Toll-Like 1/inmunología , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 6/biosíntesis , Receptor Toll-Like 6/inmunología
11.
PLoS One ; 4(11): e8055, 2009 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-19956639

RESUMEN

BACKGROUND: Abnormal trophoblast differentiation and function is the basis of many placenta-based pregnancy disorders, including pre-eclampsia and fetal growth restriction. PPARgamma, a ligand-activated nuclear receptor, plays essential roles in placental development; null murine embryos die at midgestation due to abnormalities in all placental layers, in particular, small labyrinth and expanded giant cell layer. Previous studies have focused mostly on the role of PPARgamma in trophoblast invasion. Based on the previously reported role of PPARgamma in preadipocyte differentiation, we hypothesized that PPARgamma also plays a pivotal role in trophoblast differentiation. To test this hypothesis, we report derivation of wild-type and PPARgamma-null trophoblast stem (TS) cells. METHODOLOGY/PRINCIPAL FINDINGS: PPARgamma-null TS cells showed defects in both proliferation and differentiation, specifically into labyrinthine trophoblast. Detailed marker analysis and functional studies revealed reduced differentiation of all three labyrinthine lineages, and enhanced giant cell differentiation, particularly the invasive subtypes. In addition, rosiglitazone, a specific PPARgamma agonist, reduced giant cell differentiation, while inducing Gcm1, a key regulator in labyrinth. Finally, reintroducing PPARgamma into null TS cells, using an adenovirus, normalized invasion and partially reversed defective labyrinthine differentiation, as assessed both by morphology and marker analysis. CONCLUSIONS/SIGNIFICANCE: In addition to regulating trophoblast invasion, PPARgamma plays a predominant role in differentiation of labyrinthine trophoblast lineages, which, along with fetal endothelium, form the vascular exchange interface with maternal blood. Elucidating cellular and molecular mechanisms mediating PPARgamma action will help determine if modulating PPARgamma activity, for which clinical pharmacologic agonists already exist, might modify the course of pregnancy disorders associated with placental dysfunction.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Gigantes/citología , PPAR gamma/metabolismo , Trofoblastos/citología , Adenoviridae/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Cartilla de ADN/genética , Marcadores Genéticos , Ratones , Ratones Endogámicos C57BL , Rosiglitazona , Tiazolidinedionas/farmacología , Factores de Tiempo
12.
Am J Physiol Heart Circ Physiol ; 294(2): H979-85, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18156202

RESUMEN

Hyperpolarization current (I(f)) is an important player in controlling heart rate and is stimulated by cAMP and inhibited by members of the pertussis toxin-sensitive G-protein G(i)/G(o) family. We have successfully derived cardiocytes from embryonic stem cells lacking G(o) or G(i2) and G(i3). We have established that both basal and isoproterenol-stimulated activities of I(f) in these cardiocytes have typical nodal-atrial characteristics and are unaffected by targeted gene inactivation of the G proteins G(o) or G(i2) and G(i3). Under basal conditions, both G(o) and G(i) are required for muscarinic inhibition of I(f) activity via a mechanism that involves the generation of nitric oxide, whereas, with prior stimulation by beta-agonists, only G(o) is required and G(i) and nitric oxide production are not. Our findings establish an essential role for G(o) in the antiadrenergic effect of muscarinic agent on I(f).


Asunto(s)
Células Madre Embrionarias/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Carbacol/farmacología , Línea Celular , Polaridad Celular/fisiología , Relación Dosis-Respuesta a Droga , Electrofisiología , Canales Iónicos/metabolismo , Isoproterenol/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Agonistas Muscarínicos/farmacología , Óxido Nítrico/fisiología , Técnicas de Placa-Clamp , Nodo Sinoatrial/citología
13.
Biochem Biophys Res Commun ; 357(1): 139-43, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17418106

RESUMEN

Muscarinic receptor-mediated cardiac parasympathetic activity is essential for regulating heart rate and heart rate variability (HRV). It has not been clear which G(i)/G(o) protein is responsible for these effects. We addressed this question using knockout mice that lack G protein alpha(i2), alpha(i3), or alpha(o) specifically. Unlike previously reported, our alpha(o)-null mice had significantly more survivors with normal life span. Isolated hearts from alpha(o)-null mice demonstrated much less sensitivity to the negative chronotropic effects of the muscarinic agonist carbachol to lower heart rate at baseline and a more profound effect under the stimulation of the beta-adrenergic agonist isoproterenol. In the presence of parasympathetic activation indirectly produced by methoxamine, an alpha(1)-adrenergic agonist, alpha(o)-null mice showed markedly decreased HRV compared with wild-type control mice. These differences in heart rate and HRV were not observed in alpha(i2)-null or alpha(i3)-null mice. Our findings establish an essential role for alpha(o) G protein in the anti-adrenergic effect of carbachol on heart rate regulation.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Frecuencia Cardíaca/fisiología , Corazón/fisiología , Receptores Muscarínicos/metabolismo , Animales , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ratones , Ratones Noqueados
14.
Proc Natl Acad Sci U S A ; 101(21): 8005-10, 2004 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-15148373

RESUMEN

Endostatin, a 20-kDa fragment of collagen XVIII, is a potent angiogenesis inhibitor. E-selectin, an inducible leukocyte adhesion molecule specifically expressed by endothelial cells, has also been implicated in angiogenesis. By using in vivo, ex vivo, and in vitro angiogenic assays, we investigated the functional relationship between endostatin and E-selectin. In corneal micropocket assays, recombinant endostatin administered i.p. by osmotic pump inhibited basic fibroblast growth factor-induced angiogenesis in WT, but not E-selectin-deficient, mice. Similarly, endostatin inhibited vascular endothelial growth factor-stimulated endothelial sprout formation from aortic rings dissected from WT but not from E-selectin-deficient mice. To further explore this apparent requirement for E-selectin in endostatin action, we manipulated E-selectin expression in cultured human endothelial cells. When E-selectin was induced by IL-1beta, or lipopolysaccharide, human umbilical vein endothelial cells and human dermal microvascular endothelial cells each became markedly more sensitive to inhibition by endostatin in a vascular endothelial growth factor-induced cell migration assay. To dissociate E-selectin expression from other consequences of endothelial activation, human umbilical vein endothelial cells were transduced with an adenoviral human E-selectin expression construct; these cells also showed increased sensitivity to endostatin, and this effect required the E-selectin cytoplasmic domain. Taken together, these results indicate that E-selectin is required for the antiangiogenic activity of endostatin in vivo and ex vivo and confers endostatin sensitivity to nonresponsive human endothelial cells in vitro. E-selectin may be a useful predictor and modulator of endostatin efficacy in antiangiogenic therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Selectina E/metabolismo , Endostatinas/farmacología , Animales , Aorta/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Movimiento Celular , Células Cultivadas , Córnea/irrigación sanguínea , Citocinas/farmacología , Selectina E/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Eliminación de Gen , Células HL-60 , Humanos , Técnicas In Vitro , Leucocitos/citología , Leucocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Sensibilidad y Especificidad , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA