Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 9(21): eade7280, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37235659

RESUMEN

Mechanisms underlying arteriovenous malformations (AVMs) are poorly understood. Using mice with endothelial cell (EC) expression of constitutively active Notch4 (Notch4*EC), we show decreased arteriolar tone in vivo during brain AVM initiation. Reduced vascular tone is a primary effect of Notch4*EC, as isolated pial arteries from asymptomatic mice exhibited reduced pressure-induced arterial tone ex vivo. The nitric oxide (NO) synthase (NOS) inhibitor NG-nitro-l-arginine (L-NNA) corrected vascular tone defects in both assays. L-NNA treatment or endothelial NOS (eNOS) gene deletion, either globally or specifically in ECs, attenuated AVM initiation, assessed by decreased AVM diameter and delayed time to moribund. Administering nitroxide antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl also attenuated AVM initiation. Increased NOS-dependent production of hydrogen peroxide, but not NO, superoxide, or peroxynitrite was detected in isolated Notch4*EC brain vessels during AVM initiation. Our data suggest that eNOS is involved in Notch4*EC-mediated AVM formation by up-regulating hydrogen peroxide and reducing vascular tone, thereby permitting AVM initiation and progression.


Asunto(s)
Malformaciones Arteriovenosas , Peróxido de Hidrógeno , Óxido Nítrico Sintasa de Tipo III , Animales , Ratones , Arterias/metabolismo , Peróxido de Hidrógeno/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Nitroarginina/farmacología
2.
J Exp Med ; 220(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36441145

RESUMEN

Upregulation of Notch signaling is associated with brain arteriovenous malformation (bAVM), a disease that lacks pharmacological treatments. Tetracycline (tet)-regulatable endothelial expression of constitutively active Notch4 (Notch4*tetEC) from birth induced bAVMs in 100% of mice by P16. To test whether targeting downstream signaling, while sustaining the causal Notch4*tetEC expression, induces AVM normalization, we deleted Rbpj, a mediator of Notch signaling, in endothelium from P16, by combining tet-repressible Notch4*tetEC with tamoxifen-inducible Rbpj deletion. Established pathologies, including AV connection diameter, AV shunting, vessel tortuosity, intracerebral hemorrhage, tissue hypoxia, life expectancy, and arterial marker expression were improved, compared with Notch4*tetEC mice without Rbpj deletion. Similarly, Rbpj deletion from P21 induced advanced bAVM regression. After complete AVM normalization induced by repression of Notch4*tetEC, virtually no bAVM relapsed, despite Notch4*tetEC re-expression in adults. Thus, inhibition of endothelial Rbpj halted Notch4*tetEC bAVM progression, normalized bAVM abnormalities, and restored microcirculation, providing proof of concept for targeting a downstream mediator to treat AVM pathologies despite a sustained causal molecular lesion.


Asunto(s)
Malformaciones Arteriovenosas , Encefalopatías , Malformaciones del Sistema Nervioso , Animales , Ratones , Antibacterianos , Malformaciones Arteriovenosas/genética , Encéfalo , Endotelio , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Tetraciclina , Receptor Notch4/metabolismo
3.
Science ; 378(6623): eabo5503, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36454848

RESUMEN

Monitoring of cell-cell communication in multicellular organisms is fundamental to understanding diverse biological processes such as embryogenesis and tumorigenesis. To track cell-cell contacts in vivo, we developed an intercellular genetic technology to monitor cell-cell contact and to trace cell contact histories by permanently marking contacts between cells. In mice, we engineered an artificial Notch ligand into one cell (the sender cell) and an artificial receptor into another cell (the receiver cell). Contact between the sender and receiver cells triggered a synthetic Notch signaling that activated downstream transcriptional programs in the receiver cell, thereby transiently or permanently labeling it. In vivo cell-cell contact was observed during development, tissue homeostasis, and tumor growth. This technology may be useful for studying dynamic in vivo cell-cell contacts and cell fate plasticity.


Asunto(s)
Comunicación Celular , Perfilación de la Expresión Génica , Receptores Notch , Animales , Ratones , Carcinogénesis , Plasticidad de la Célula , Transducción de Señal , Receptores Notch/genética , Perfilación de la Expresión Génica/métodos
4.
Gastroenterology ; 157(1): 193-209.e9, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30872106

RESUMEN

BACKGROUND & AIMS: Mechanical forces contribute to portal hypertension (PHTN) and fibrogenesis. We investigated the mechanisms by which forces are transduced by liver sinusoidal endothelial cells (LSECs) into pressure and matrix changes. METHODS: We isolated primary LSECs from mice and induced mechanical stretch with a Flexcell device, to recapitulate the pulsatile forces induced by congestion, and performed microarray and RNA-sequencing analyses to identify gene expression patterns associated with stretch. We also performed studies with C57BL/6 mice (controls), mice with deletion of neutrophil elastase (NE-/-) or peptidyl arginine deiminase type IV (Pad4-/-) (enzymes that formation of neutrophil extracellular traps [NETs]), and mice with LSEC-specific deletion of Notch1 (Notch1iΔEC). We performed partial ligation of the suprahepatic inferior vena cava (pIVCL) to simulate congestive hepatopathy-induced portal hypertension in mice; some mice were given subcutaneous injections of sivelestat or underwent bile-duct ligation. Portal pressure was measured using a digital blood pressure analyzer and we performed intravital imaging of livers of mice. RESULTS: Expression of the neutrophil chemoattractant CXCL1 was up-regulated in primary LSECs exposed to mechanical stretch, compared with unexposed cells. Intravital imaging of livers in control mice revealed sinusoidal complexes of neutrophils and platelets and formation of NETs after pIVCL. NE-/- and Pad4-/- mice had lower portal pressure and livers had less fibrin compared with control mice after pIVCL and bile-duct ligation; neutrophil recruitment into sinusoidal lumen of liver might increase portal pressure by promoting sinusoid microthrombi. RNA-sequencing of LSECs identified proteins in mechanosensitive signaling pathways that are altered in response to mechanical stretch, including integrins, Notch1, and calcium signaling pathways. Mechanical stretch of LSECs increased expression of CXCL1 via integrin-dependent activation of transcription factors regulated by Notch and its interaction with the mechanosensitive piezo calcium channel. CONCLUSIONS: In studies of LSECs and knockout mice, we identified mechanosensitive angiocrine signals released by LSECs which promote PHTN by recruiting sinusoidal neutrophils and promoting formation of NETs and microthrombi. Strategies to target these pathways might be developed for treatment of PHTN. RNA-sequencing accession number: GSE119547.


Asunto(s)
Capilares/metabolismo , Quimiocina CXCL1/metabolismo , Células Endoteliales/metabolismo , Hipertensión Portal/metabolismo , Hígado/irrigación sanguínea , Infiltración Neutrófila , Estrés Mecánico , Trombosis/metabolismo , Animales , Señalización del Calcio , Capilares/citología , Trampas Extracelulares , Hidrolasas/genética , Técnicas In Vitro , Integrinas/metabolismo , Elastasa de Leucocito/genética , Ligadura , Hígado/metabolismo , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presión Portal , Arginina Deiminasa Proteína-Tipo 4 , Receptor Notch1/genética , Vena Cava Inferior/cirugía
5.
Sci Rep ; 7(1): 11965, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28931948

RESUMEN

The functions of blood flow in the morphogenesis of mammalian arteries and veins are not well understood. We examined the development of the dorsal aorta (DA) and the cardinal vein (CV) in Ncx1 -/- mutants, which lack blood flow due to a deficiency in a sodium calcium ion exchanger expressed specifically in the heart. The mutant DA and CV were abnormally connected. The endothelium of the Ncx1 -/- mutant DA lacked normal expression of the arterial markers ephrin-B2 and Connexin-40. Notch1 activation, known to promote arterial specification, was decreased in mutant DA endothelial cells (ECs), which ectopically expressed the venous marker Coup-TFII. These findings suggest that flow has essential functions in the DA by promoting arterial and suppressing venous marker expression. In contrast, flow plays a lesser role in the CV, because expression of arterial-venous markers in CV ECs was not as dramatically affected in Ncx1 -/- mutants. We propose a molecular mechanism by which blood flow mediates DA and CV morphogenesis, by regulating arterial-venous specification of DA ECs to ensure proper separation of the developing DA and CV.


Asunto(s)
Circulación Sanguínea , Vasos Sanguíneos/embriología , Morfogénesis , Animales , Aorta/patología , Conexinas/análisis , Células Endoteliales/patología , Efrina-B2/análisis , Ratones , Ratones Noqueados , Receptor Notch1/análisis , Intercambiador de Sodio-Calcio/genética , Venas/patología , Proteína alfa-5 de Unión Comunicante
6.
Hepatology ; 64(4): 1302-1316, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27362333

RESUMEN

UNLABELLED: Liver vasculature is crucial for adequate hepatic functions. Global deletion of Notch signaling in mice results in liver vascular pathologies. However, whether Notch in endothelium is essential for hepatic vascular structure and function remains unknown. To uncover the function of endothelial Notch in the liver, we deleted Rbpj, a transcription factor mediating all canonical Notch signaling, or Notch1 from the endothelium of postnatal mice. We investigated the hepatic vascular defects in these mutants. The liver was severely affected within 2 weeks of endothelial deletion of Rbpj from birth. Two-week old mutant mice had enlarged vessels on the liver surface, abnormal vascular architecture, and dilated sinusoids. Vascular casting and fluorosphere passage experiments indicated the presence of porto-systemic shunts. These mutant mice presented with severely necrotic liver parenchyma and significantly larger hypoxic areas, likely resulting from vascular shunts. We also found elevated levels of VEGF receptor 3 together with reduced levels of ephrin-B2, suggesting a possible contribution of these factors to the generation of hepatic vascular abnormalities. Deletion of Rbpj from the adult endothelium also led to dilated sinusoids, vascular shunts, and necrosis, albeit milder than that observed in mice with deletion from birth. Similar to deletion of Rbpj, loss of endothelial Notch1 from birth led to similar hepatic vascular malformations within 2 weeks. CONCLUSIONS: Endothelial Notch signaling is essential for the development and maintenance of proper hepatic vascular architecture and function. These findings may elucidate the molecular pathogenesis of hepatic vascular malformation and the safety of therapeutics inhibiting Notch. (Hepatology 2016;64:1302-1316).


Asunto(s)
Hígado/irrigación sanguínea , Receptor Notch1/fisiología , Malformaciones Vasculares/etiología , Animales , Endotelio Vascular , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Ratones , Transducción de Señal
8.
Proc Natl Acad Sci U S A ; 111(50): 18007-12, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25468970

RESUMEN

Arteriovenous (AV) malformation (AVM) is a devastating condition characterized by focal lesions of enlarged, tangled vessels that shunt blood from arteries directly to veins. AVMs can form anywhere in the body and can cause debilitating ischemia and life-threatening hemorrhagic stroke. The mechanisms that underlie AVM formation remain poorly understood. Here, we examined the cellular and hemodynamic changes at the earliest stages of brain AVM formation by time-lapse two-photon imaging through cranial windows of mice expressing constitutively active Notch4 (Notch4*). AVMs arose from enlargement of preexisting microvessels with capillary diameter and blood flow and no smooth muscle cell coverage. AV shunting began promptly after Notch4* expression in endothelial cells (ECs), accompanied by increased individual EC areas, rather than increased EC number or proliferation. Alterations in Notch signaling in ECs of all vessels, but not arteries alone, affected AVM formation, suggesting that Notch functions in the microvasculature and/or veins to induce AVM. Increased Notch signaling interfered with the normal biological control of hemodynamics, permitting a positive feedback loop of increasing blood flow and vessel diameter and driving focal AVM growth from AV connections with higher blood velocity at the expense of adjacent AV connections with lower velocity. Endothelial expression of constitutively active Notch1 also led to brain AVMs in mice. Our data shed light on cellular and hemodynamic mechanisms underlying AVM pathogenesis elicited by increased Notch signaling in the endothelium.


Asunto(s)
Capilares/patología , Malformaciones Arteriovenosas Intracraneales/metabolismo , Malformaciones Arteriovenosas Intracraneales/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Animales , Bromodesoxiuridina , Capilares/metabolismo , Células Endoteliales/metabolismo , Citometría de Flujo , Malformaciones Arteriovenosas Intracraneales/etiología , Ratones , Receptor Notch4 , Flujo Sanguíneo Regional/fisiología , Transducción de Señal/fisiología , Microtomografía por Rayos X
9.
Development ; 141(19): 3782-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25209249

RESUMEN

Arteriovenous malformations (AVMs) are tortuous vessels characterized by arteriovenous (AV) shunts, which displace capillaries and shunt blood directly from artery to vein. Notch signaling regulates embryonic AV specification by promoting arterial, as opposed to venous, endothelial cell (EC) fate. To understand the essential role of endothelial Notch signaling in postnatal AV organization, we used inducible Cre-loxP recombination to delete Rbpj, a mediator of canonical Notch signaling, from postnatal ECs in mice. Deletion of endothelial Rbpj from birth resulted in features of AVMs by P14, including abnormal AV shunting and tortuous vessels in the brain, intestine and heart. We further analyzed brain AVMs, as they pose particular health risks. Consistent with AVM pathology, we found cerebral hemorrhage, hypoxia and necrosis, and neurological deficits. AV shunts originated from capillaries (and possibly venules), with the earliest detectable morphological abnormalities in AV connections by P8. Prior to AV shunt formation, alterations in EC gene expression were detected, including decreased Efnb2 and increased Pai1, which encodes a downstream effector of TGFß signaling. After AV shunts had formed, whole-mount immunostaining showed decreased Efnb2 and increased Ephb4 expression within AV shunts, suggesting that ECs were reprogrammed from arterial to venous identity. Deletion of Rbpj from adult ECs led to tortuosities in gastrointestinal, uterine and skin vascular beds, but had mild effects in the brain. Our results demonstrate a temporal requirement for Rbpj in postnatal ECs to maintain proper artery, capillary and vein organization and to prevent abnormal AV shunting and AVM pathogenesis.


Asunto(s)
Malformaciones Arteriovenosas/genética , Malformaciones Arteriovenosas/patología , Endotelio Vascular/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Eliminación de Gen , Perfilación de la Expresión Génica , Procesamiento de Imagen Asistido por Computador , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor EphB4/metabolismo
10.
Microcirculation ; 21(7): 578-86, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24673722

RESUMEN

OBJECTIVE: The cell surface protein ephrin-B2 is expressed in arterial and not venous ECs throughout development and adulthood. Endothelial ephrin-B2 is required for vascular development and angiogenesis, but its role in established arteries is currently unknown. We investigated the physiological role of ephrin-B2 signaling in adult endothelium. METHODS: We generated adult conditional knockout mice lacking the Efnb2 gene specifically in ECs and evaluated the vasodilation responses to blood flow increase and ACh in the cremaster muscle preparation by intravital microscope and in carotid artery by in vivo ultrasound. RESULTS: We found that the Efnb2 conditional knockout mice were defective in acute arterial dilation. Vasodilation was impaired in cremaster arterioles in response to either increased flow or ACh, and in the carotid arteries in response to increased flow. Levels of cGMP, an effector of NO, were diminished in mutant arteries following ACh stimulation. GSNO, a donor for the vasodilator NO, alleviated the vasodilatory defects in the mutants. Immunostaining showed that a subset of ephrin-B2 proteins colocalized with caveolin-1, a negative regulator of eNOS. CONCLUSIONS: Our data suggest that endothelial ephrin-B2 is required for endothelial-dependent arterial dilation and NO signaling in adult endothelium.


Asunto(s)
Arterias/fisiología , Células Endoteliales/fisiología , Efrina-B2/fisiología , Vasodilatación/fisiología , Acetilcolina/farmacología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Arteria Carótida Común/fisiología , Caveolina 1/análisis , GMP Cíclico/metabolismo , Células Endoteliales/efectos de los fármacos , Femenino , Genes Reporteros , Hemorreología , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Óxido Nítrico/fisiología , Donantes de Óxido Nítrico/farmacología , Especificidad de Órganos , S-Nitrosoglutatión/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
11.
Development ; 141(5): 1120-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24550118

RESUMEN

Coordinated arterial-venous differentiation is crucial for vascular development and function. The origin of the cardinal vein (CV) in mammals is unknown, while conflicting theories have been reported in chick and zebrafish. Here, we provide the first molecular characterization of endothelial cells (ECs) expressing venous molecular markers, or venous-fated ECs, within the emergent dorsal aorta (DA). These ECs, expressing the venous molecular markers Coup-TFII and EphB4, cohabited the early DA with ECs expressing the arterial molecular markers ephrin B2, Notch and connexin 40. These mixed ECs in the early DA expressed either the arterial or venous molecular marker, but rarely both. Subsequently, the DA exhibited uniform arterial markers. Real-time imaging of mouse embryos revealed EC movement from the DA to the CV during the stage when venous-fated ECs occupied the DA. We analyzed mutants for EphB4, which encodes a receptor tyrosine kinase for the ephrin B2 ligand, as we hypothesized that ephrin B2/EphB4 signaling may mediate the repulsion of venous-fated ECs from the DA to the CV. Using an EC quantification approach, we discovered that venous-fated ECs increased in the DA and decreased in the CV in the mutants, whereas the rest of the ECs in each vessel were unaffected. This result suggests that the venous-fated ECs were retained in the DA and missing in the CV in the EphB4 mutant, and thus that ephrin B2/EphB4 signaling normally functions to clear venous-fated ECs from the DA to the CV by cell repulsion. Therefore, our cellular and molecular evidence suggests that the DA harbors venous progenitors that move to participate in CV formation, and that ephrin B2/EphB4 signaling regulates this aortic contribution to the mammalian CV.


Asunto(s)
Aorta/citología , Células Madre/citología , Venas/citología , Animales , Células Endoteliales/citología , Células Endoteliales/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Células Madre/metabolismo
12.
Vasc Cell ; 5(1): 7, 2013 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-23601498

RESUMEN

BACKGROUND: Notch4 is a member of the Notch family of receptors that is primarily expressed in the vascular endothelial cells. Genetic deletion of Notch4 does not result in an overt phenotype in mice, thus the function of Notch4 remains poorly understood. METHODS: We examined the requirement for Notch4 in the development of breast cancer vasculature. Orthotopic transplantation of mouse mammary tumor cells wild type for Notch4 into Notch4 deficient hosts enabled us to delineate the contribution of host Notch4 independent of its function in the tumor cell compartment. RESULTS: Here, we show that Notch4 expression is required for tumor onset and early tumor perfusion in a mouse model of breast cancer. We found that Notch4 expression is upregulated in mouse and human mammary tumor vasculature. Moreover, host Notch4 deficiency delayed the onset of MMTV-PyMT tumors, wild type for Notch4, after transplantation. Vessel perfusion was decreased in tumors established in Notch4-deficient hosts. Unlike in inhibition of Notch1 or Dll4, vessel density and branching in tumors developed in Notch4-deficient mice were unchanged. However, final tumor size was similar between tumors grown in wild type and Notch4 null hosts. CONCLUSION: Our results suggest a novel role for Notch4 in the establishment of tumor colonies and vessel perfusion of transplanted mammary tumors.

13.
PLoS One ; 7(6): e38590, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761686

RESUMEN

BACKGROUND: The ability to measure blood velocities is critical for studying vascular development, physiology, and pathology. A key challenge is to quantify a wide range of blood velocities in vessels deep within living specimens with concurrent diffraction-limited resolution imaging of vascular cells. Two-photon laser scanning microscopy (TPLSM) has shown tremendous promise in analyzing blood velocities hundreds of micrometers deep in animals with cellular resolution. However, current analysis of TPLSM-based data is limited to the lower range of blood velocities and is not adequate to study faster velocities in many normal or disease conditions. METHODOLOGY/PRINCIPAL FINDINGS: We developed line-scanning particle image velocimetry (LS-PIV), which used TPLSM data to quantify peak blood velocities up to 84 mm/s in live mice harboring brain arteriovenous malformation, a disease characterized by high flow. With this method, we were able to accurately detect the elevated blood velocities and exaggerated pulsatility along the abnormal vascular network in these animals. LS-PIV robustly analyzed noisy data from vessels as deep as 850 µm below the brain surface. In addition to analyzing in vivo data, we validated the accuracy of LS-PIV up to 800 mm/s using simulations with known velocity and noise parameters. CONCLUSIONS/SIGNIFICANCE: To our knowledge, these blood velocity measurements are the fastest recorded with TPLSM. Partnered with transgenic mice carrying cell-specific fluorescent reporters, LS-PIV will also enable the direct in vivo correlation of cellular, biochemical, and hemodynamic parameters in high flow vascular development and diseases such as atherogenesis, arteriogenesis, and vascular anomalies.


Asunto(s)
Malformaciones Arteriovenosas/patología , Velocidad del Flujo Sanguíneo/fisiología , Encefalopatías/patología , Eritrocitos/patología , Microscopía Confocal , Reología , Animales , Encéfalo/citología , Rastreo Celular , Efrina-B2/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Hemodinámica , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/fisiología , Receptor Notch4 , Receptores Notch/fisiología
14.
Sci Transl Med ; 4(117): 117ra8, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22261032

RESUMEN

Abnormally enlarged blood vessels underlie many life-threatening disorders including arteriovenous (AV) malformations (AVMs). The core defect in AVMs is high-flow AV shunts, which connect arteries directly to veins, "stealing" blood from capillaries. Here, we studied mouse brain AV shunts caused by up-regulation of Notch signaling in endothelial cells (ECs) through transgenic expression of constitutively active Notch4 (Notch4*). Using four-dimensional two-photon imaging through a cranial window, we found that normalizing Notch signaling by repressing Notch4* expression converted large-caliber, high-flow AV shunts to capillary-like vessels. The structural regression of the high-flow AV shunts returned blood to capillaries, thus reversing tissue hypoxia. This regression was initiated by vessel narrowing without the loss of ECs and required restoration of EphB4 receptor expression by venous ECs. Normalization of Notch signaling resulting in regression of high-flow AV shunts, and a return to normal blood flow suggests that targeting the Notch pathway may be useful therapeutically for treating diseases such as AVMs.


Asunto(s)
Malformaciones Arteriovenosas/metabolismo , Vasos Sanguíneos/patología , Proteínas Proto-Oncogénicas/fisiología , Receptores Notch/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Capilares , Células Endoteliales/citología , Regulación de la Expresión Génica , Hipoxia , Ratones , Ratones Transgénicos , Modelos Cardiovasculares , Fotones , Proteínas Proto-Oncogénicas/biosíntesis , Receptor EphB4/metabolismo , Receptor Notch4 , Receptores Notch/biosíntesis , Transducción de Señal
15.
Am J Physiol Cell Physiol ; 298(5): C1087-99, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181926

RESUMEN

The bone morphogenetic protein (BMP) pathway is known to be involved in limb myogenesis during development, but whether it is involved in postnatal muscle regeneration is unclear. We have found that adult inhibitor of differentiation (Id)-mutant (Id1(+/-)Id3(-/-)) mice display delayed and reduced skeletal muscle regeneration after injury compared with either wild-type littermates or Id3-null mice. Immunoblotting of wild-type muscle lysates revealed that, not only were Id1 and Id3 highly upregulated within 24 h after injury, but other upstream components of the BMP pathway were as well, including the BMP receptor type II and phosphorylated Smad1/5/8 (pSmad1/5/8). Inhibition of BMP signaling in injured skeletal muscle by Noggin injection reduced pSmad1/5/8, Id1, and Id3 protein levels. The mouse myoblast-derived cell line C2C12 also expressed Id1, Id3, BMP receptor type II, and pSmad1/5/8 during proliferation, but all were reduced upon differentiation into myotubes. In addition, these cells secreted mature BMP-4, and BMP signaling could be inhibited with exogenous Noggin, causing a reduction in pSmad1/5/8, Id1, and Id3 levels. Confocal immunofluorescence microscopy revealed that activated Pax7(+) myoblasts coexpressed nuclear pSmad1/5/8, Id1, and Id3 in injured mouse skeletal muscle sections. Although we did not observe differences in the numbers of quiescent Pax7(+) satellite cells in adult uninjured hindlimb muscles, we did observe a significant reduction in the number of proliferating Pax7(+) cells in the Id-mutant mice after muscle injury compared with either wild-type or Id3-null mice. These data suggest a model in which BMP signaling regulates Id1 and Id3 in muscle satellite cells, which directs their proper proliferation before terminal myogenic differentiation after skeletal muscle injury in postnatal animals.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Músculo Esquelético/lesiones , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Mutación , Regeneración , Células Satélite del Músculo Esquelético , Transducción de Señal
16.
Am J Physiol Lung Cell Mol Physiol ; 298(2): L169-77, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19933399

RESUMEN

Lung arteriovenous (AV) shunts or malformations cause significant morbidity and mortality in several distinct clinical syndromes. For most patients with lung AV shunts, there is still no optimal treatment. The underlying molecular and cellular etiology for lung AV shunts remains elusive, and currently described animal models have insufficiently addressed this problem. Using a tetracycline-repressible system, we expressed constitutively active Notch4 (Notch4*) specifically in the endothelium of adult mice. More than 90% of mice developed lung hemorrhages and respiratory insufficiency and died by 6-7 wk after gene expression began. Vascular casting and fluorescent microsphere analysis showed evidence of lung AV shunts in affected mice. Cessation of Notch4* expression reversed these pathophysiological effects. Assessment of the vascular morphology revealed enlarged, tortuous vessels in the lungs that resembled arteriovenous malformations. By using whole lung organ culture, we demonstrated the effects of constitutively active Notch4 on the lung vasculature to be a primary lung phenomenon. Together, our results indicate the importance of Notch signaling in maintaining the lung vasculature and offer a new, reliable model with which to study the pathobiology of lung arteriovenous shunts and malformations.


Asunto(s)
Malformaciones Arteriovenosas/metabolismo , Endotelio Vascular/metabolismo , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Animales , Malformaciones Arteriovenosas/patología , Embrión de Mamíferos/anatomía & histología , Humanos , Pulmón/embriología , Pulmón/patología , Ratones , Modelos Animales , Mutación , Proteínas Proto-Oncogénicas/genética , Receptor Notch4 , Receptores Notch/genética , Transducción de Señal/fisiología , Técnicas de Cultivo de Tejidos
17.
Science ; 326(5950): 294-8, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19815777

RESUMEN

Blood vessels form de novo (vasculogenesis) or upon sprouting of capillaries from preexisting vessels (angiogenesis). With high-resolution imaging of zebrafish vascular development, we uncovered a third mode of blood vessel formation whereby the first embryonic artery and vein, two unconnected blood vessels, arise from a common precursor vessel. The first embryonic vein formed by selective sprouting of progenitor cells from the precursor vessel, followed by vessel segregation. These processes were regulated by the ligand EphrinB2 and its receptor EphB4, which are expressed in arterial-fated and venous-fated progenitors, respectively, and interact to orient the direction of progenitor migration. Thus, directional control of progenitor migration drives arterial-venous segregation and generation of separate parallel vessels from a single precursor vessel, a process essential for vascular development.


Asunto(s)
Arterias/embriología , Células Endoteliales/fisiología , Efrina-B2/metabolismo , Morfogénesis , Receptor EphB4/metabolismo , Células Madre/fisiología , Venas/embriología , Animales , Animales Modificados Genéticamente , Aorta/citología , Aorta/embriología , Arterias/citología , Movimiento Celular , Células Endoteliales/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Venas/citología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
18.
Lab Invest ; 89(9): 971-82, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19546852

RESUMEN

Brain arteriovenous malformations (BAVMs) can cause lethal hemorrhagic stroke and have no effective treatment. The cellular and molecular basis for this disease is largely unknown. We have previously shown that expression of constitutively-active Notch4 receptor in the endothelium elicits and maintains the hallmarks of BAVM in mice, thus establishing a mouse model of the disease. Our work suggested that Notch pathway could be a critical molecular mediator of BAVM pathogenesis. Here, we investigated the hypothesis that upregulated Notch activation contributes to the pathogenesis of human BAVM. We examined the expression of the canonical Notch downstream target Hes1 in the endothelium of human BAVMs by immunofluorescence, and showed increased levels relative to either autopsy or surgical biopsy controls. We then analyzed receptor activity using an antibody to the activated form of the Notch1 receptor, and found increased levels of activity. These findings suggest that Notch activation may promote the development and even maintenance of BAVM. We also detected increases in Hes1 and activated Notch1 expression in our mouse model of BAVM induced by constitutively active Notch4, demonstrating molecular similarity between the mouse model and the human disease. Our work suggests that activation of Notch signaling is an important molecular candidate in BAVM pathogenesis and further validates that our animal model provides a platform to study the progression as well as the regression of the disease.


Asunto(s)
Endotelio Vascular/metabolismo , Malformaciones Arteriovenosas Intracraneales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch1/biosíntesis , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Proteínas de Homeodominio/metabolismo , Humanos , Malformaciones Arteriovenosas Intracraneales/patología , Ratones , Ratones Mutantes , Microscopía Fluorescente , Neovascularización Patológica , Receptor Notch4 , Factor de Transcripción HES-1 , Regulación hacia Arriba
19.
Development ; 135(22): 3755-64, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18952909

RESUMEN

A mutual coordination of size between developing arteries and veins is essential for establishing proper connections between these vessels and, ultimately, a functional vasculature; however, the cellular and molecular regulation of this parity is not understood. Here, we demonstrate that the size of the developing dorsal aorta and cardinal vein is reciprocally balanced. Mouse embryos carrying gain-of-function Notch alleles show enlarged aortae and underdeveloped cardinal veins, whereas those with loss-of-function mutations show small aortae and large cardinal veins. Notch does not affect the overall number of endothelial cells but balances the proportion of arterial to venous endothelial cells, thereby modulating the relative sizes of both vessel types. Loss of ephrin B2 or its receptor EphB4 also leads to enlarged aortae and underdeveloped cardinal veins; however, endothelial cells with venous identity are mislocalized in the aorta, suggesting that ephrin B2/EphB4 signaling functions distinctly from Notch by sorting arterial and venous endothelial cells into their respective vessels. Our findings provide mechanistic insight into the processes underlying artery and vein size equilibration during angiogenesis.


Asunto(s)
Arterias/metabolismo , Efrina-B2/metabolismo , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas/metabolismo , Receptor EphB4/metabolismo , Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Venas/metabolismo , Animales , Arterias/anatomía & histología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Efrina-B2/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas/genética , Receptor EphB4/genética , Receptor Notch1/deficiencia , Receptor Notch1/genética , Receptor Notch4 , Receptores Notch/genética , Venas/anatomía & histología
20.
Proc Natl Acad Sci U S A ; 105(31): 10901-6, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18667694

RESUMEN

Brain arteriovenous malformations (BAVMs) can cause devastating stroke in young people and contribute to half of all hemorrhagic stroke in children. Unfortunately, the pathogenesis of BAVMs is unknown. In this article we show that activation of Notch signaling in the endothelium during brain development causes BAVM in mice. We turned on constitutively active Notch4 (int3) expression in endothelial cells from birth by using the tetracycline-regulatable system. All mutants developed hallmarks of BAVMs, including cerebral arteriovenous shunting and vessel enlargement, by 3 weeks of age and died by 5 weeks of age. Twenty-five percent of the mutants showed signs of neurological dysfunction, including ataxia and seizure. Affected mice exhibited hemorrhage and neuronal cell death within the cerebral cortex and cerebellum. Strikingly, int3 repression resolved ataxia and reversed the disease progression, demonstrating that int3 is not only sufficient to induce, but also required to sustain the disease. We show that int3 expression results in widespread enlargement of the microvasculature, which coincided with a reduction in capillary density, linking vessel enlargement to Notch's known function of inhibiting vessel sprouting. Our data suggest that the Notch pathway is a molecular regulator of BAVM pathogenesis in mice, and offer hope that their regression might be possible by targeting the causal molecular lesion.


Asunto(s)
Endotelio Vascular/metabolismo , Malformaciones Arteriovenosas Intracraneales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Inmunohistoquímica , Malformaciones Arteriovenosas Intracraneales/etiología , Malformaciones Arteriovenosas Intracraneales/patología , Ratones , Receptor Notch4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...