Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 545(7654): 305-310, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28489816

RESUMEN

Cerebral cavernous malformations (CCMs) are a cause of stroke and seizure for which no effective medical therapies yet exist. CCMs arise from the loss of an adaptor complex that negatively regulates MEKK3-KLF2/4 signalling in brain endothelial cells, but upstream activators of this disease pathway have yet to be identified. Here we identify endothelial Toll-like receptor 4 (TLR4) and the gut microbiome as critical stimulants of CCM formation. Activation of TLR4 by Gram-negative bacteria or lipopolysaccharide accelerates CCM formation, and genetic or pharmacologic blockade of TLR4 signalling prevents CCM formation in mice. Polymorphisms that increase expression of the TLR4 gene or the gene encoding its co-receptor CD14 are associated with higher CCM lesion burden in humans. Germ-free mice are protected from CCM formation, and a single course of antibiotics permanently alters CCM susceptibility in mice. These studies identify unexpected roles for the microbiome and innate immune signalling in the pathogenesis of a cerebrovascular disease, as well as strategies for its treatment.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Hemangioma Cavernoso del Sistema Nervioso Central/inmunología , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Inmunidad Innata , Receptor Toll-Like 4/inmunología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Susceptibilidad a Enfermedades , Células Endoteliales/metabolismo , Femenino , Vida Libre de Gérmenes , Bacterias Gramnegativas/inmunología , Hemangioma Cavernoso del Sistema Nervioso Central/microbiología , Humanos , Inyecciones Intravenosas , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Masculino , Ratones , Transducción de Señal , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
2.
Nature ; 532(7597): 122-6, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27027284

RESUMEN

Cerebral cavernous malformations (CCMs) are common inherited and sporadic vascular malformations that cause strokes and seizures in younger individuals. CCMs arise from endothelial cell loss of KRIT1, CCM2 or PDCD10, non-homologous proteins that form an adaptor complex. How disruption of the CCM complex results in disease remains controversial, with numerous signalling pathways (including Rho, SMAD and Wnt/ß-catenin) and processes such as endothelial-mesenchymal transition (EndMT) proposed to have causal roles. CCM2 binds to MEKK3 (refs 7, 8, 9, 10, 11), and we have recently shown that CCM complex regulation of MEKK3 is essential during vertebrate heart development. Here we investigate this mechanism in CCM disease pathogenesis. Using a neonatal mouse model of CCM disease, we show that expression of the MEKK3 target genes Klf2 and Klf4, as well as Rho and ADAMTS protease activity, are increased in the endothelial cells of early CCM lesions. By contrast, we find no evidence of EndMT or increased SMAD or Wnt signalling during early CCM formation. Endothelial-specific loss of Map3k3 (also known as Mekk3), Klf2 or Klf4 markedly prevents lesion formation, reverses the increase in Rho activity, and rescues lethality. Consistent with these findings in mice, we show that endothelial expression of KLF2 and KLF4 is increased in human familial and sporadic CCM lesions, and that a disease-causing human CCM2 mutation abrogates the MEKK3 interaction without affecting CCM complex formation. These studies identify gain of MEKK3 signalling and KLF2/4 function as causal mechanisms for CCM pathogenesis that may be targeted to develop new CCM therapeutics.


Asunto(s)
Células Endoteliales/metabolismo , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas ADAM/metabolismo , Animales , Animales Recién Nacidos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Femenino , Hemangioma Cavernoso del Sistema Nervioso Central/etiología , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/deficiencia , MAP Quinasa Quinasa Quinasa 3/deficiencia , Masculino , Ratones , Unión Proteica , Proteínas de Unión al GTP rho/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-32393494

RESUMEN

The rise in multidrug-resistant (MDR) organisms portends a serious global threat to the health care system with nearly untreatable infectious diseases, including pneumonia and its often fatal sequelae, acute respiratory distress syndrome (ARDS) and sepsis. Gram-negative bacteria (GNB), including Acinetobacter baumannii, Pseudomonas aeruginosa, and carbapenemase-producing Klebsiella pneumoniae (CPKP), are among the World Health Organization's and National Institutes of Health's high-priority MDR pathogens for targeted development of new therapies. Here, we show that stabilizing the host's vasculature by genetic deletion or pharmacological inhibition of the small GTPase ADP-ribosylation factor 6 (ARF6) increases survival rates of mice infected with A. baumannii, P. aeruginosa, and CPKP. We show that the pharmacological inhibition of ARF6-GTP phenocopies endothelium-specific Arf6 disruption in enhancing the survival of mice with A. baumannii pneumonia, suggesting that inhibition is on target. Finally, we show that the mechanism of protection elicited by these small-molecule inhibitors acts by the restoration of vascular integrity disrupted by GNB lipopolysaccharide (LPS) activation of the TLR4/MyD88/ARNO/ARF6 pathway. By targeting the host's vasculature with small-molecule inhibitors of ARF6 activation, we circumvent microbial drug resistance and provide a potential alternative/adjunctive treatment for emerging and reemerging pathogens.


Asunto(s)
Acinetobacter baumannii , Infecciones por Bacterias Gramnegativas , Factor 6 de Ribosilación del ADP , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana Múltiple/genética , Bacterias Gramnegativas , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Ratones , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa
4.
Development ; 144(13): 2392-2401, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576770

RESUMEN

The placental labyrinth is the interface for gas and nutrient exchange between the embryo and the mother; hence its proper development is essential for embryogenesis. However, the molecular mechanism underlying development of the placental labyrinth, particularly in terms of its endothelial organization, is not well understood. Here, we determined that fibronectin leucine-rich transmembrane protein 2 (FLRT2), a repulsive ligand of the UNC5 receptor family for neurons, is unexpectedly expressed in endothelial cells specifically in the placental labyrinth. Mice lacking FLRT2 in endothelial cells exhibited embryonic lethality at mid-gestation, with systemic congestion and hypoxia. Although they lacked apparent deformities in the embryonic vasculature and heart, the placental labyrinths of these embryos exhibited aberrant alignment of endothelial cells, which disturbed the feto-maternal circulation. Interestingly, this vascular deformity was related to endothelial repulsion through binding to the UNC5B receptor. Our results suggest that the proper organization of the placental labyrinth depends on coordinated inter-endothelial repulsion, which prevents uncontrolled layering of the endothelium.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Organogénesis , Placenta/embriología , Placenta/metabolismo , Transducción de Señal , Animales , Supervivencia Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Femenino , Eliminación de Gen , Hipoxia/patología , Glicoproteínas de Membrana/deficiencia , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Receptores de Netrina , Placenta/irrigación sanguínea , Placenta/citología , Embarazo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo
5.
Circ Res ; 122(12): 1716-1721, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29720384

RESUMEN

RATIONALE: The clinical course of cerebral cavernous malformations is highly unpredictable, with few cross-sectional studies correlating proinflammatory genotypes and plasma biomarkers with prior disease severity. OBJECTIVE: We hypothesize that a panel of 24 candidate plasma biomarkers, with a reported role in the physiopathology of cerebral cavernous malformations, may predict subsequent clinically relevant disease activity. METHODS AND RESULTS: Plasma biomarkers were assessed in nonfasting peripheral venous blood collected from consecutive cerebral cavernous malformation subjects followed for 1 year after initial sample collection. A first cohort (N=49) was used to define the best model of biomarker level combinations to predict a subsequent symptomatic lesional hemorrhagic expansion within a year after the blood sample. We generated the receiver operating characteristic curves and area under the curve for each biomarker individually and each weighted linear combination of relevant biomarkers. The best model to predict lesional activity was selected as that minimizing the Akaike information criterion. In this cohort, 11 subjects experienced symptomatic lesional hemorrhagic expansion (5 bleeds and 10 lesional growths) within a year after the blood draw. Subjects had lower soluble CD14 (cluster of differentiation 14; P=0.05), IL (interleukin)-6 (P=0.04), and VEGF (vascular endothelial growth factor; P=0.0003) levels along with higher plasma levels of IL-1ß (P=0.008) and soluble ROBO4 (roundabout guidance receptor 4; P=0.03). Among the 31 weighted linear combinations of these 5 biomarkers, the best model (with the lowest Akaike information criterion value, 25.3) was the weighted linear combination including soluble CD14, IL-1ß, VEGF, and soluble ROBO4, predicting a symptomatic hemorrhagic expansion with a sensitivity of 86% and specificity of 88% (area under the curve, 0.90; P<0.0001). We then validated our best model in the second sequential independent cohort (N=28). CONCLUSIONS: This is the first study reporting a predictive association between plasma biomarkers and subsequent cerebral cavernous malformation disease clinical activity. This may be applied in clinical prognostication and stratification of cases in clinical trials.


Asunto(s)
Biomarcadores/sangre , Hemangioma Cavernoso del Sistema Nervioso Central/sangre , Adolescente , Adulto , Anciano , Área Bajo la Curva , Hemorragia Cerebral/etiología , Niño , Preescolar , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Hemangioma Cavernoso del Sistema Nervioso Central/complicaciones , Humanos , Interleucina-1beta/sangre , Interleucina-6/sangre , Receptores de Lipopolisacáridos/sangre , Masculino , Persona de Mediana Edad , Curva ROC , Receptores de Superficie Celular/sangre , Sensibilidad y Especificidad , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto Joven
6.
Microcirculation ; 26(5): e12540, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30825241

RESUMEN

OBJECTIVE: Accumulating evidence suggests the vascular endothelium plays a fundamental role in the pathophysiology of obesity by regulating the functional status of white adipose and systemic metabolism. Robo4 is expressed specifically in endothelial cells and increases vascular stability and inhibits angiogenesis. We sought to determine the role of Robo4 in modulating cardiometabolic function in response to high-fat feeding. METHODS: We examined exercise capacity, glucose tolerance, and white adipose tissue artery gene expression, endothelium-dependent dilation (EDD), and angiogenesis in wild type and Robo4 knockout (KO) mice fed normal chow (NC) or a high-fat diet (HFD). RESULTS: We found Robo4 deletion enhances exercise capacity in NC-fed mice and HFD markedly increased the expression of the Robo4 ligand, Slit2, in white adipose tissue. Deletion of Robo4 increased angiogenesis in white adipose tissue and protected against HFD-induced impairments in white adipose artery vasodilation and glucose intolerance. CONCLUSIONS: We demonstrate a novel functional role for Robo4 in endothelial cell function and metabolic homeostasis in white adipose tissue, with Robo4 deletion protecting against endothelial and metabolic dysfunction associated with a HFD. Our findings suggest that Robo4-dependent signaling pathways may be a novel target in anti-obesity therapy.


Asunto(s)
Tejido Adiposo Blanco , Arterias , Grasas de la Dieta/efectos adversos , Endotelio Vascular , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Receptores de Superficie Celular , Tejido Adiposo Blanco/irrigación sanguínea , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Arterias/metabolismo , Arterias/patología , Grasas de la Dieta/farmacología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/deficiencia , Vasodilatación/efectos de los fármacos , Vasodilatación/genética
7.
Exp Physiol ; 104(3): 434-442, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30633428

RESUMEN

NEW FINDINGS: What is the central question of this study? Greater large artery stiffness is associated with dysfunctional resistance artery vasodilatory responses, impaired memory and greater risk of Alzheimer's disease. However, it is unknown whether stiffer large arteries affect cerebral and skeletal muscle feed artery responses to vasoconstrictors. What is the main finding and its importance? In a mouse model with greater large artery stiffness (Eln+/- ), we find an exacerbated vasoconstrictor response to angiotensin II in cerebral arteries, but not skeletal muscle feed arteries, thus implicating altered cerebral artery angiotensin II responsiveness in the poor brain outcomes associated with greater large artery stiffness. ABSTRACT: Greater stiffness of the large elastic arteries is associated with end-organ damage and dysfunction. At the same time, resistance artery vasoconstrictor responsiveness influences vascular tone and organ blood flow. However, it is unknown whether large elastic artery stiffness modulates the responsiveness to vasoconstrictors in resistance arteries of the cerebral or skeletal muscle circulations. We previously described the elastin haploinsufficient (Eln+/- ) mouse as a model with greater aortic stiffness, but with similar cerebral and skeletal muscle feed artery stiffness to wild-type (Eln+/+ ) mice. Here, we used this model to examine the relationship between large elastic artery stiffness and resistance artery vasoconstrictor responses. In middle cerebral arteries (MCAs), vasoconstriction in response to angiotensin II (Ang II) was ∼40% greater in Eln+/- compared with Eln+/+ mice (P = 0.02), and this group difference was ameliorated by losartan, indicating a role for Ang II type 1 receptors (AT1Rs). In gastrocnemius feed arteries, Eln+/- and Eln+/+ mice did not differ in the response to Ang II. In addition, the vasoconstrictor responses to noradrenaline, endothelin-1 and potassium chloride were not different between Eln+/- and Eln+/+ mice for either MCAs or gastrocnemius feed arteries. The MCA AT1R gene expression did not differ between groups, whereas Ang II type 2 receptor gene expression was ∼50% lower in MCAs from Eln+/- versus Eln+/+ mice (P = 0.01). In conclusion, greater large elastic artery stiffness is associated with an exacerbated vasoconstriction response to Ang II in cerebral arteries, but is not associated with the responses to other vasoconstrictors in either cerebral or skeletal muscle feed arteries.


Asunto(s)
Arterias Cerebrales/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Rigidez Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Angiotensina II/farmacología , Animales , Arterias Cerebrales/metabolismo , Arterias Cerebrales/fisiopatología , Modelos Animales de Enfermedad , Endotelina-1/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Losartán/farmacología , Masculino , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Norepinefrina/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Rigidez Vascular/fisiología , Vasoconstricción/fisiología
8.
Circulation ; 135(17): 1632-1645, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28073805

RESUMEN

BACKGROUND: Cardiac recovery in response to mechanical unloading by left ventricular assist devices (LVADs) has been demonstrated in subgroups of patients with chronic heart failure (HF). Hallmarks of HF are depletion and disorganization of the transverse tubular system (t-system) in cardiomyocytes. Here, we investigated remodeling of the t-system in human end-stage HF and its role in cardiac recovery. METHODS: Left ventricular biopsies were obtained from 5 donors and 26 patients with chronic HF undergoing implantation of LVADs. Three-dimensional confocal microscopy and computational image analysis were applied to assess t-system structure, density, and distance of ryanodine receptor clusters to the sarcolemma, including the t-system. Recovery of cardiac function in response to mechanical unloading was assessed by echocardiography during turndown of the LVAD. RESULTS: The majority of HF myocytes showed remarkable t-system remodeling, particularly sheet-like invaginations of the sarcolemma. Circularity of t-system components was decreased in HF versus controls (0.37±0.01 versus 0.46±0.02; P<0.01), and the volume/length ratio was increased in HF (0.36±0.01 versus 0.25±0.02 µm2; P<0.0001). T-system density was reduced in HF, leading to increased ryanodine receptor-sarcolemma distances (0.96±0.05 versus 0.64±0.1 µm; P<0.01). Low ryanodine receptor-sarcolemma distances at the time of LVAD implantation predicted high post-LVAD left ventricular ejection fractions (P<0.01) and ejection fraction increases during unloading (P<0.01). Ejection fraction in patients with pre-LVAD ryanodine receptor-sarcolemma distances >1 µm did not improve after mechanical unloading. In addition, calcium transients were recorded in field-stimulated isolated human cardiomyocytes and analyzed with respect to local t-system density. Calcium release in HF myocytes was restricted to regions proximal to the sarcolemma. Local calcium upstroke was delayed (23.9±4.9 versus 10.3±1.7 milliseconds; P<0.05) and more asynchronous (18.1±1.5 versus 8.9±2.2 milliseconds; P<0.01) in HF cells with low t-system density versus cells with high t-system density. CONCLUSIONS: The t-system in end-stage human HF presents a characteristic novel phenotype consisting of sheet-like invaginations of the sarcolemma. Our results suggest that the remodeled t-system impairs excitation-contraction coupling and functional recovery during chronic LVAD unloading. An intact t-system at the time of LVAD implantation may constitute a precondition and predictor for functional cardiac recovery after mechanical unloading.


Asunto(s)
Acoplamiento Excitación-Contracción , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Contracción Miocárdica , Miocitos Cardíacos/patología , Función Ventricular Izquierda , Remodelación Ventricular , Adulto , Anciano , Biopsia , Estudios de Casos y Controles , Ecocardiografía , Femenino , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Microscopía Confocal , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Estudios Prospectivos , Diseño de Prótesis , Recuperación de la Función , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Factores de Tiempo , Resultado del Tratamiento
9.
Nature ; 492(7428): 252-5, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23143332

RESUMEN

The innate immune response is essential for combating infectious disease. Macrophages and other cells respond to infection by releasing cytokines, such as interleukin-1ß (IL-1ß), which in turn activate a well-described, myeloid-differentiation factor 88 (MYD88)-mediated, nuclear factor-κB (NF-κB)-dependent transcriptional pathway that results in inflammatory-cell activation and recruitment. Endothelial cells, which usually serve as a barrier to the movement of inflammatory cells out of the blood and into tissue, are also critical mediators of the inflammatory response. Paradoxically, the cytokines vital to a successful immune defence also have disruptive effects on endothelial cell-cell interactions and can trigger degradation of barrier function and dissociation of tissue architecture. The mechanism of this barrier dissolution and its relationship to the canonical NF-κB pathway remain poorly defined. Here we show that the direct, immediate and disruptive effects of IL-1ß on endothelial stability in a human in vitro cell model are NF-κB independent and are instead the result of signalling through the small GTPase ADP-ribosylation factor 6 (ARF6) and its activator ARF nucleotide binding site opener (ARNO; also known as CYTH2). Moreover, we show that ARNO binds directly to the adaptor protein MYD88, and thus propose MYD88-ARNO-ARF6 as a proximal IL-1ß signalling pathway distinct from that mediated by NF-κB. Finally, we show that SecinH3, an inhibitor of ARF guanine nucleotide-exchange factors such as ARNO, enhances vascular stability and significantly improves outcomes in animal models of inflammatory arthritis and acute inflammation.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina/metabolismo , Factor 6 de Ribosilación del ADP , Adyuvantes Inmunológicos/farmacología , Animales , Artritis/patología , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Purinas/farmacología , Transducción de Señal , Tiofenos/farmacología
10.
Proc Natl Acad Sci U S A ; 112(23): 7255-60, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26039999

RESUMEN

Yes-associated protein (YAP), a transcription coactivator, is the major downstream effector of the Hippo pathway, which plays a critical role in organ size control and cancer development. However, how YAP is regulated by extracellular stimuli in tumorigenesis remains incompletely understood. Netrin-1, a laminin-related secreted protein, displays proto-oncogenic activity in cancers. Nonetheless, the downstream signaling mediating its oncogenic effects is not well defined. Here we show that netrin-1 via its transmembrane receptors, deleted in colorectal cancer and uncoordinated-5 homolog, up-regulates YAP expression, escalating YAP levels in the nucleus and promoting cancer cell proliferation and migration. Inactivating netrin-1, deleted in colorectal cancer, or uncoordinated-5 homolog B (UNC5B) decreases YAP protein levels, abrogating cancer cell progression by netrin-1, whereas knockdown of mammalian STE20-like protein kinase 1/2 (MST1/2) or large tumor suppressor kinase 1/2 (Lats1/2), two sets of upstream core kinases of the Hippo pathway, has no effect in blocking netrin-1-induced up-regulation of YAP. Netrin-1 stimulates phosphatase 1A to dephosphorylate YAP, which leads to decreased ubiquitination and degradation, enhancing YAP accumulation and signaling. Hence, our findings support that netrin-1 exerts oncogenic activity through YAP signaling, providing a mechanism coupling extracellular signals to the nuclear YAP oncogene.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis , Factores de Crecimiento Nervioso/fisiología , Fosfoproteínas/metabolismo , Proteínas Supresoras de Tumor/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Humanos , Netrina-1 , Fosforilación , Estabilidad Proteica , Transducción de Señal , Factores de Transcripción , Regulación hacia Arriba , Proteínas Señalizadoras YAP
11.
J Physiol ; 595(22): 6869-6885, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28949006

RESUMEN

KEY POINTS: Endothelial cells employ transient receptor potential isoform 4 (TRPV4) channels to sense ambient mechanical and chemical stimuli. In retinal microvascular endothelial cells, TRPV4 channels regulate calcium homeostasis, cytoskeletal signalling and the organization of adherens junctional contacts. Intracellular calcium increases induced by TRPV4 agonists include a significant contribution from calcium release from internal stores. Activation of TRPV4 channels regulates retinal endothelial barriers in vitro and in vivo. TRPV4 sensing may provide a feedback mechanism between sensing shear flow and eicosanoid modulators, vascular permeability and contractility at the inner retinal endothelial barrier. ABSTRACT: The identity of microvascular endothelial (MVE) mechanosensors that sense blood flow in response to mechanical and chemical stimuli and regulate vascular permeability in the retina is unknown. Using immunohistochemistry, calcium imaging, electrophysiology, impedance measurements and vascular permeability assays, we show that the transient receptor potential isoform 4 (TRPV4) plays a major role in Ca2+ /cation signalling, cytoskeletal remodelling and barrier function in retinal microvasculature in vitro and in vivo. Human retinal MVE cells (HrMVECs) predominantly expressed Trpv1 and Trpv4 transcripts, and TRPV4 was broadly localized to the plasma membrane of cultured cells and intact blood vessels in the inner retina. Treatment with the selective TRPV4 agonist GSK1016790A (GSK101) activated a nonselective cation current, robustly elevated [Ca2+ ]i and reversibly increased the permeability of MVEC monolayers. This was associated with disrupted organization of endothelial F-actin, downregulated expression of occludin and remodelling of adherens contacts consisting of vascular endothelial cadherin (VE-cadherin) and ß-catenin. In vivo, GSK101 increased the permeability of retinal blood vessels in wild type but not in TRPV4 knockout mice. Agonist-evoked effects on barrier permeability and cytoskeletal reorganization were antagonized by the selective TRPV4 blocker HC 067047. Human choroidal endothelial cells expressed lower TRPV4 mRNA/protein levels and showed less pronounced agonist-evoked calcium signals compared to MVECs. These findings indicate a major role for TRPV4 in Ca2+ homeostasis and barrier function in human retinal capillaries and suggest that TRPV4 may differentially contribute to the inner vs. outer blood-retinal barrier function.


Asunto(s)
Uniones Adherentes/metabolismo , Calcio/metabolismo , Células Endoteliales/metabolismo , Vasos Retinianos/citología , Canales Catiónicos TRPV/metabolismo , Potenciales de Acción , Animales , Barrera Hematorretinal/metabolismo , Señalización del Calcio , Células Cultivadas , Células Endoteliales/citología , Retroalimentación Fisiológica , Humanos , Leucina/análogos & derivados , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Ocludina/genética , Ocludina/metabolismo , Pirroles/farmacología , Vasos Retinianos/metabolismo , Sulfonamidas , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores
12.
Biochim Biophys Acta ; 1863(7 Pt B): 1772-81, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26952936

RESUMEN

Chromatin structure is determined by nucleosome positioning, histone modifications, and DNA methylation. How chromatin modifications are coordinately altered under pathological conditions remains elusive. Here we describe a stress-activated mechanism of concerted chromatin modification in the heart. In mice, pathological stress activates cardiomyocytes to express Brg1 (nucleosome-remodeling factor), G9a/Glp (histone methyltransferase), and Dnmt3 (DNA methyltransferase). Once activated, Brg1 recruits G9a and then Dnmt3 to sequentially assemble repressive chromatin-marked by H3K9 and CpG methylation-on a key molecular motor gene (Myh6), thereby silencing Myh6 and impairing cardiac contraction. Disruption of Brg1, G9a or Dnmt3 erases repressive chromatin marks and de-represses Myh6, reducing stress-induced cardiac dysfunction. In human hypertrophic hearts, BRG1-G9a/GLP-DNMT3 complex is also activated; its level correlates with H3K9/CpG methylation, Myh6 repression, and cardiomyopathy. Our studies demonstrate a new mechanism of chromatin assembly in stressed hearts and novel therapeutic targets for restoring Myh6 and ventricular function. The stress-induced Brg1-G9a-Dnmt3 interactions and sequence of repressive chromatin assembly on Myh6 illustrates a molecular mechanism by which the heart epigenetically responds to environmental signals. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Asunto(s)
Cardiomegalia/enzimología , Cardiomiopatías/enzimología , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Helicasas/metabolismo , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Miocardio/enzimología , Cadenas Pesadas de Miosina/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Estrés Fisiológico , Factores de Transcripción/metabolismo , Adaptación Fisiológica , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Cromatina/genética , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Helicasas/deficiencia , ADN Helicasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Edad Gestacional , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Metilación , Ratones Noqueados , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Recuperación de la Función , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Función Ventricular Izquierda
14.
Circulation ; 131(3): 289-99, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25486933

RESUMEN

BACKGROUND: Cerebral cavernous malformation (CCM) is a hemorrhagic stroke disease affecting up to 0.5% of North Americans that has no approved nonsurgical treatment. A subset of patients have a hereditary form of the disease due primarily to loss-of-function mutations in KRIT1, CCM2, or PDCD10. We sought to identify known drugs that could be repurposed to treat CCM. METHODS AND RESULTS: We developed an unbiased screening platform based on both cellular and animal models of loss of function of CCM2. Our discovery strategy consisted of 4 steps: an automated immunofluorescence and machine-learning-based primary screen of structural phenotypes in human endothelial cells deficient in CCM2, a secondary screen of functional changes in endothelial stability in these same cells, a rapid in vivo tertiary screen of dermal microvascular leak in mice lacking endothelial Ccm2, and finally a quaternary screen of CCM lesion burden in these same mice. We screened 2100 known drugs and bioactive compounds and identified 2 candidates, cholecalciferol (vitamin D3) and tempol (a scavenger of superoxide), for further study. Each drug decreased lesion burden in a mouse model of CCM vascular disease by ≈50%. CONCLUSIONS: By identifying known drugs as potential therapeutics for CCM, we have decreased the time, cost, and risk of bringing treatments to patients. Each drug also prompts additional exploration of biomarkers of CCM disease. We further suggest that the structure-function screening platform presented here may be adapted and scaled to facilitate drug discovery for diverse loss-of-function genetic vascular disease.


Asunto(s)
Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos/métodos , Hemangioma Cavernoso del Sistema Nervioso Central/tratamiento farmacológico , Animales , Células Cultivadas , Neoplasias del Sistema Nervioso Central/patología , Colecalciferol/farmacología , Colecalciferol/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Resultado del Tratamiento
15.
Hum Mol Genet ; 23(23): 6223-34, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24990152

RESUMEN

Cerebral cavernous malformation (CCM) is a disease of vascular malformations known to be caused by mutations in one of three genes: CCM1, CCM2 or CCM3. Despite several studies, the mechanism of CCM lesion onset remains unclear. Using a Ccm1 knockout mouse model, we studied the morphogenesis of early lesion formation in the retina in order to provide insight into potential mechanisms. We demonstrate that lesions develop in a stereotypic location and pattern, preceded by endothelial hypersprouting as confirmed in a zebrafish model of disease. The vascular defects seen with loss of Ccm1 suggest a defect in endothelial flow response. Taken together, these results suggest new mechanisms of early CCM disease pathogenesis and provide a framework for further study.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Proto-Oncogénicas/genética , Retina/patología , Animales , Animales Modificados Genéticamente , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Humanos , Proteína KRIT1 , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pez Cebra
16.
Bioinformatics ; 31(22): 3666-72, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26209429

RESUMEN

MOTIVATION: The Cancer Genome Atlas (TCGA) RNA-Sequencing data are used widely for research. TCGA provides 'Level 3' data, which have been processed using a pipeline specific to that resource. However, we have found using experimentally derived data that this pipeline produces gene-expression values that vary considerably across biological replicates. In addition, some RNA-Sequencing analysis tools require integer-based read counts, which are not provided with the Level 3 data. As an alternative, we have reprocessed the data for 9264 tumor and 741 normal samples across 24 cancer types using the Rsubread package. We have also collated corresponding clinical data for these samples. We provide these data as a community resource. RESULTS: We compared TCGA samples processed using either pipeline and found that the Rsubread pipeline produced fewer zero-expression genes and more consistent expression levels across replicate samples than the TCGA pipeline. Additionally, we used a genomic-signature approach to estimate HER2 (ERBB2) activation status for 662 breast-tumor samples and found that the Rsubread data resulted in stronger predictions of HER2 pathway activity. Finally, we used data from both pipelines to classify 575 lung cancer samples based on histological type. This analysis identified various non-coding RNA that may influence lung-cancer histology. AVAILABILITY AND IMPLEMENTATION: The RNA-Sequencing and clinical data can be downloaded from Gene Expression Omnibus (accession number GSE62944). Scripts and code that were used to process and analyze the data are available from https://github.com/srp33/TCGA_RNASeq_Clinical. CONTACT: stephen_piccolo@byu.edu or andreab@genetics.utah.edu SUPPLEMENTARY INFORMATION: Supplementary material is available at Bioinformatics online.


Asunto(s)
Neoplasias de la Mama/genética , Genoma Humano , Análisis de Secuencia de ARN/métodos , Estadística como Asunto , Neoplasias de la Mama/clasificación , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Curva ROC , Reproducibilidad de los Resultados
17.
J Immunol ; 192(12): 6045-52, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24835390

RESUMEN

The vascular endothelium responds to infection by destabilizing endothelial cell-cell junctions to allow fluid and cells to pass into peripheral tissues, facilitating clearance of infection and tissue repair. During sepsis, endotoxin and other proinflammatory molecules induce excessive vascular leak, which can cause organ dysfunction, shock, and death. Current therapies for sepsis are limited to antibiotics and supportive care, which are often insufficient to reduce morbidity and prevent mortality. Previous attempts at blocking inflammatory cytokine responses in humans proved ineffective at reducing the pathologies associated with sepsis, highlighting the need for a new therapeutic strategy. The small GTPase ARF6 is activated by a MyD88-ARNO interaction to induce vascular leak through disruption of endothelial adherens junctions. In this study, we show that the MyD88-ARNO-ARF6-signaling axis is responsible for LPS-induced endothelial permeability and is a destabilizing convergence point used by multiple inflammatory cues. We also show that blocking ARF6 with a peptide construct of its N terminus is sufficient to reduce vascular leak and enhance survival during endotoxic shock, without inhibiting the host cytokine response. Our data highlight the therapeutic potential of blocking ARF6 and reducing vascular leak for the treatment of inflammatory conditions, such as endotoxemia.


Asunto(s)
Factores de Ribosilacion-ADP/inmunología , Uniones Adherentes/inmunología , Permeabilidad Capilar/inmunología , Células Endoteliales/inmunología , Choque Séptico/inmunología , Transducción de Señal/inmunología , Factor 6 de Ribosilación del ADP , Uniones Adherentes/patología , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Células Endoteliales/patología , Femenino , Proteínas Activadoras de GTPasa/inmunología , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/inmunología , Choque Séptico/inducido químicamente , Choque Séptico/patología , Transducción de Señal/efectos de los fármacos
18.
J Physiol ; 593(8): 1931-43, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25627876

RESUMEN

KEY POINTS: Increased large artery stiffness is a hallmark of arterial dysfunction with advancing age and is also present in other disease conditions such as diabetes. Increased large artery stiffness is correlated with resistance artery dysfunction in humans. Using a mouse model of altered arterial elastin content, this is the first study to examine the cause-and-effect relationship between large artery stiffness and peripheral resistance artery function. Our results indicate that mice with genetically greater large artery stiffness have impaired cerebral artery endothelial function, but generally preserved skeletal muscle feed artery endothelial function. The mechanisms for impaired cerebral artery endothelial function are reduced nitric oxide bioavailability and increased oxidative stress. These findings suggest that interventions that target large artery stiffness may be important to reduce disease risk associated with cerebral artery dysfunction in conditions such as advancing age. ABSTRACT: Advancing age as well as diseases such as diabetes are characterized by both increased large artery stiffness and impaired peripheral artery function. It has been hypothesized that greater large artery stiffness causes peripheral artery dysfunction; however, a cause-and-effect relationship has not previously been established. We used elastin heterozygote mice (Eln(+/-) ) as a model of increased large artery stiffness without co-morbidities unrelated to the large artery properties. Aortic stiffness, measured by pulse wave velocity, was ∼35% greater in Eln(+/-) mice than in wild-type (Eln(+/+) ) mice (P = 0.04). Endothelium-dependent dilatation (EDD), assessed by the maximal dilatation to acetylcholine, was ∼40% lower in Eln(+/-) than Eln(+/+) mice in the middle cerebral artery (MCA, P < 0.001), but was similar between groups in the gastrocnemius feed arteries (GFA, P = 0.79). In the MCA, EDD did not differ between groups after incubation with the nitric oxide (NO) synthase inhibitor N(ω) -nitro-l-arginine methyl ester (P > 0.05), indicating that lower NO bioavailability contributed to the impaired EDD in Eln(+/-) mice. Superoxide production and content of the oxidative stress marker nitrotyrosine was higher in MCAs from Eln(+/-) compared with Eln(+/+) mice (P < 0.05). In the MCA, after incubation with the superoxide scavenger TEMPOL, maximal EDD improved by ∼65% in Eln(+/-) (P = 0.002), but was unchanged in Eln(+/+) mice (P = 0.17). These results indicate that greater large artery stiffness has a more profound effect on endothelial function in cerebral arteries compared with skeletal muscle feed arteries. Greater large artery stiffness can cause cerebral artery endothelial dysfunction by reducing NO bioavailability and increasing oxidative stress.


Asunto(s)
Arterias Cerebrales/fisiopatología , Endotelio Vascular/fisiopatología , Músculo Esquelético/irrigación sanguínea , Rigidez Vascular/fisiología , Animales , Arterias Cerebrales/efectos de los fármacos , Modelos Animales de Enfermedad , Elastina/genética , Elastina/metabolismo , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Indometacina/farmacología , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología , Rigidez Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
19.
Am J Hum Genet ; 90(1): 40-8, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22226084

RESUMEN

Ocular coloboma is a developmental defect of the eye and is due to abnormal or incomplete closure of the optic fissure. This disorder displays genetic and clinical heterogeneity. Using a positional cloning approach, we identified a mutation in the ATP-binding cassette (ABC) transporter ABCB6 in a Chinese family affected by autosomal-dominant coloboma. The Leu811Val mutation was identified in seven affected members of the family and was absent in six unaffected members from three generations. A LOD score of 3.2 at θ = 0 was calculated for the mutation identified in this family. Sequence analysis was performed on the ABCB6 exons from 116 sporadic cases of microphthalmia with coloboma (MAC), isolated coloboma, and aniridia, and an additional mutation (A57T) was identified in three patients with MAC. These two mutations were not present in the ethnically matched control populations. Immunostaining of transiently transfected, Myc-tagged ABCB6 in retinal pigment epithelial (RPE) cells showed that it localized to the endoplasmic reticulum and Golgi apparatus of RPE cells. RT-PCR of ABCB6 mRNA in human cell lines and tissue indicated that ABCB6 is expressed in the retinae and RPE cells. Using zebrafish, we show that abcb6 is expressed in the eye and CNS. Morpholino knockdown of abcb6 in zebrafish produces a phenotype characteristic of coloboma and replicates the clinical phenotype observed in our index cases. The knockdown phenotype can be corrected with coinjection of the wild-type, but not mutant, ABCB6 mRNA, suggesting that the phenotypes observed in zebrafish are due to insufficient abcb6 function. Our results demonstrate that ABCB6 mutations cause ocular coloboma.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Coloboma/genética , Mutación , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Pueblo Asiatico/genética , Secuencia de Bases , Línea Celular , Sistema Nervioso Central/metabolismo , Exones , Anomalías del Ojo/genética , Femenino , Humanos , Escala de Lod , Masculino , Microftalmía/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Morfolinos/administración & dosificación , Epitelio Pigmentado de la Retina , Transfección , Pez Cebra , Proteínas de Pez Cebra/genética
20.
FASEB J ; 28(1): 265-74, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24043260

RESUMEN

Activation of Rap1 GTPase can improve the integrity of the barrier of the retina pigment epithelium (RPE) and reduce choroidal neovascularization (CNV). Inhibition of NADPH oxidase activation also reduces CNV. We hypothesize that Rap1 inhibits NADPH oxidase-generated ROS and thereby reduces CNV formation. Using a murine model of laser-induced CNV, we determined that reduced Rap1 activity in RPE/choroid occurred with CNV formation and that activation of Rap1 by 2'-O-Me-cAMP (8CPT)-reduced laser-induced CNV via inhibiting NADPH oxidase-generated ROS. In RPE, inhibition of Rap1 by Rap1 GTPase-activating protein (Rap1GAP) increased ROS generation, whereas activation of Rap1 by 8CPT reduced ROS by interfering with the assembly of NADPH oxidase membrane subunit p22phox with NOX4 or cytoplasmic subunit p47phox. Activation of NADPH oxidase with Rap1GAP reduced RPE barrier integrity via cadherin phosphorylation and facilitated choroidal EC migration across the RPE monolayer. Rap1GAP-induced ROS generation was inhibited by active Rap1a, but not Rap1b, and activation of Rap1a by 8CPT in Rap1b(-/-) mice reduced laser-induced CNV, in correlation with decreased ROS generation in RPE/choroid. These findings provide evidence that active Rap1 reduces CNV by interfering with the assembly of NADPH oxidase subunits and increasing the integrity of the RPE barrier.


Asunto(s)
Neovascularización Coroidal/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Ratones , Ratones Noqueados , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA