Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
APL Bioeng ; 8(1): 016108, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38352162

RESUMEN

Cerebral cavernous malformations (CCMs) are vascular lesions that predominantly form in blood vessels of the central nervous system upon loss of the CCM multimeric protein complex. The endothelial cells within CCM lesions are characterized by overactive MEKK3 kinase and KLF2/4 transcription factor signaling, leading to pathological changes such as increased endothelial cell spreading and reduced junctional integrity. Concomitant to aberrant endothelial cell signaling, non-autonomous signals from the extracellular matrix (ECM) have also been implicated in CCM lesion growth and these factors might explain why CCM lesions mainly develop in the central nervous system. Here, we adapted a three-dimensional microfluidic system to examine CCM1 deficient human micro-vessels in distinctive extracellular matrices. We validate that pathological hallmarks are maintained in this model. We further show that key genes responsible for homeostasis of hyaluronic acid, a major extracellular matrix component of the central nervous system, are dysregulated in CCM. Supplementing the matrix in our model with distinct forms of hyaluronic acid inhibits pathological cell spreading and rescues barrier function. Hyaluronic acid acts by dampening cell-matrix adhesion signaling in CCM, either downstream or in parallel of KLF2/4. This study provides a proof-of-principle that ECM embedded 3D microfluidic models are ideally suited to identify how changes in ECM structure and signaling impact vascular malformations.

2.
Sci Signal ; 16(782): eabq1366, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37098119

RESUMEN

Macrophages are key cellular contributors to the pathogenesis of COVID-19, the disease caused by the virus SARS-CoV-2. The SARS-CoV-2 entry receptor ACE2 is present only on a subset of macrophages at sites of SARS-CoV-2 infection in humans. Here, we investigated whether SARS-CoV-2 can enter macrophages, replicate, and release new viral progeny; whether macrophages need to sense a replicating virus to drive cytokine release; and, if so, whether ACE2 is involved in these mechanisms. We found that SARS-CoV-2 could enter, but did not replicate within, ACE2-deficient human primary macrophages and did not induce proinflammatory cytokine expression. By contrast, ACE2 overexpression in human THP-1-derived macrophages permitted SARS-CoV-2 entry, processing and replication, and virion release. ACE2-overexpressing THP-1 macrophages sensed active viral replication and triggered proinflammatory, antiviral programs mediated by the kinase TBK-1 that limited prolonged viral replication and release. These findings help elucidate the role of ACE2 and its absence in macrophage responses to SARS-CoV-2 infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/genética , Citocinas , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Macrófagos/metabolismo , Virión/metabolismo
3.
EMBO J ; 42(11): e112590, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36912146

RESUMEN

During development, the lymphatic vasculature forms as a second network derived chiefly from blood vessels. The transdifferentiation of embryonic venous endothelial cells (VECs) into lymphatic endothelial cells (LECs) is a key step in this process. Specification, differentiation and maintenance of LEC fate are all driven by the transcription factor Prox1, yet the downstream mechanisms remain to be elucidated. We here present a single-cell transcriptomic atlas of lymphangiogenesis in zebrafish, revealing new markers and hallmarks of LEC differentiation over four developmental stages. We further profile single-cell transcriptomic and chromatin accessibility changes in zygotic prox1a mutants that are undergoing a LEC-VEC fate shift. Using maternal and zygotic prox1a/prox1b mutants, we determine the earliest transcriptomic changes directed by Prox1 during LEC specification. This work altogether reveals new downstream targets and regulatory regions of the genome controlled by Prox1 and presents evidence that Prox1 specifies LEC fate primarily by limiting blood vascular and haematopoietic fate. This extensive single-cell resource provides new mechanistic insights into the enigmatic role of Prox1 and the control of LEC differentiation in development.


Asunto(s)
Vasos Linfáticos , Pez Cebra , Animales , Pez Cebra/genética , Proteínas de Homeodominio/genética , Proteínas Supresoras de Tumor/genética , Células Endoteliales , Células Cultivadas , Diferenciación Celular , Linfangiogénesis/genética , Factores de Transcripción/genética , Análisis de la Célula Individual
4.
Front Oncol ; 13: 1101522, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36776301

RESUMEN

Brain tumors represent the leading cause of disease-related mortality and morbidity in children, with effective treatments urgently required. One factor limiting the effectiveness of systemic therapy is the blood-brain-barrier (BBB), which limits the brain penetration of many anticancer drugs. BBB integrity is often compromised in tumors, referred to as the blood-brain-tumor-barrier (BBTB), and the impact of a compromised BBTB on the therapeutic sensitivity of brain tumors has been clearly shown for a few selected agents. However, the heterogeneity of barrier alteration observed within a single tumor and across distinct pediatric tumor types represents an additional challenge. Herein, we discuss what is known regarding the heterogeneity of tumor-associated vasculature in pediatric brain tumors. We discuss innovative and complementary preclinical model systems that will facilitate real-time functional analyses of BBTB for all pediatric brain tumor types. We believe a broader use of these preclinical models will enable us to develop a greater understanding of the processes underlying tumor-associated vasculature formation and ultimately more efficacious treatment options.

5.
PLoS Pathog ; 18(4): e1010389, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35446924

RESUMEN

Meningitis caused by infectious pathogens is associated with vessel damage and infarct formation, however the physiological cause is often unknown. Cryptococcus neoformans is a human fungal pathogen and causative agent of cryptococcal meningitis, where vascular events are observed in up to 30% of patients, predominantly in severe infection. Therefore, we aimed to investigate how infection may lead to vessel damage and associated pathogen dissemination using a zebrafish model that permitted noninvasive in vivo imaging. We find that cryptococcal cells become trapped within the vasculature (dependent on their size) and proliferate there resulting in vasodilation. Localised cryptococcal growth, originating from a small number of cryptococcal cells in the vasculature was associated with sites of dissemination and simultaneously with loss of blood vessel integrity. Using a cell-cell junction tension reporter we identified dissemination from intact blood vessels and where vessel rupture occurred. Finally, we manipulated blood vessel tension via cell junctions and found increased tension resulted in increased dissemination. Our data suggest that global vascular vasodilation occurs following infection, resulting in increased vessel tension which subsequently increases dissemination events, representing a positive feedback loop. Thus, we identify a mechanism for blood vessel damage during cryptococcal infection that may represent a cause of vascular damage and cortical infarction during cryptococcal meningitis.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Meningitis Criptocócica , Animales , Criptococosis/microbiología , Humanos , Pez Cebra
6.
Dev Dyn ; 251(2): 336-349, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34174014

RESUMEN

BACKGROUND: Lymphatic vascular development is regulated by well-characterized signaling and transcriptional pathways. These pathways regulate lymphatic endothelial cell (LEC) migration, motility, polarity, and morphogenesis. Canonical and non-canonical WNT signaling pathways are known to control LEC polarity and development of lymphatic vessels and valves. PKD1, encoding Polycystin-1, is the most commonly mutated gene in polycystic kidney disease but has also been shown to be essential in lymphatic vascular morphogenesis. The mechanism by which Pkd1 acts during lymphangiogenesis remains unclear. RESULTS: Here we find that loss of non-canonical WNT signaling components Wnt5a and Ryk phenocopy lymphatic defects seen in Pkd1 knockout mice. To investigate genetic interaction, we generated Pkd1;Wnt5a double knockout mice. Loss of Wnt5a suppressed phenotypes seen in the lymphatic vasculature of Pkd1-/- mice and Pkd1 deletion suppressed phenotypes observed in Wnt5a-/- mice. Thus, we report mutually suppressive roles for Pkd1 and Wnt5a, with developing lymphatic networks restored to a more wild type state in double mutant mice. This genetic interaction between Pkd1 and the non-canonical WNT signaling pathway ultimately controls LEC polarity and the morphogenesis of developing vessel networks. CONCLUSION: Our work suggests that Pkd1 acts at least in part by regulating non-canonical WNT signaling during the formation of lymphatic vascular networks.


Asunto(s)
Vasos Linfáticos , Enfermedades Renales Poliquísticas , Animales , Vasos Linfáticos/metabolismo , Ratones , Ratones Noqueados , Morfogénesis/genética , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Proteína Quinasa C , Proteínas Tirosina Quinasas Receptoras/metabolismo , Vía de Señalización Wnt/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
7.
Curr Biol ; 31(6): 1326-1336.e5, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33581074

RESUMEN

Epithelia must eliminate apoptotic cells to preserve tissue barriers and prevent inflammation.1 Several different mechanisms exist for apoptotic clearance, including efferocytosis2,3 and apical extrusion.4,5 We found that extrusion was the first-line response to apoptosis in cultured monolayers and in zebrafish epidermis. During extrusion, the apoptotic cell elicited active lamellipodial protrusions and assembly of a contractile extrusion ring in its neighbors. Depleting E-cadherin compromised both the contractile ring and extrusion, implying that a cadherin-dependent pathway allows apoptotic cells to engage their neighbors for extrusion. We identify RhoA as the cadherin-dependent signal in the neighbor cells and show that it is activated in response to contractile tension from the apoptotic cell. This mechanical stimulus is conveyed by a myosin-VI-dependent mechanotransduction pathway that is necessary both for extrusion and to preserve the epithelial barrier when apoptosis was stimulated. Earlier studies suggested that release of sphingosine-1-phosphate (S1P) from apoptotic cells might define where RhoA was activated. However, we found that, although S1P is necessary for extrusion, its contribution does not require a localized source of S1P in the epithelium. We therefore propose a unified view of how RhoA is stimulated to engage neighbor cells for apoptotic extrusion. Here, tension-sensitive mechanotransduction is the proximate mechanism that activates RhoA specifically in the immediate neighbors of apoptotic cells, but this also must be primed by S1P in the tissue environment. Together, these elements provide a coincidence detection system that confers robustness on the extrusion response.


Asunto(s)
Apoptosis , Células Epiteliales/citología , Mecanotransducción Celular , Pez Cebra , Proteína de Unión al GTP rhoA/fisiología , Animales , Cadherinas/genética , Lisofosfolípidos , Esfingosina/análogos & derivados
8.
Neuro Oncol ; 23(5): 732-742, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33258962

RESUMEN

BACKGROUND: Novel targeted therapies for children diagnosed with medulloblastoma (MB), the most common malignant pediatric brain tumor, are urgently required. A major hurdle in the development of effective therapies is the impaired delivery of systemic therapies to tumor cells due to a specialized endothelial blood-brain barrier (BBB). Accordingly, the integrity of the BBB is an essential consideration in any preclinical model used for assessing novel therapeutics. This study sought to assess the functional integrity of the BBB in several preclinical mouse models of MB. METHODS: Dynamic contrast enhancement magnetic resonance imaging (MRI) was used to evaluate blood-brain-tumor barrier (BBTB) permeability in a murine genetically engineered mouse model (GEMM) of Sonic Hedgehog (SHH) MB, patient-derived orthotopic xenograft models of MB (SHH and Gp3), and orthotopic transplantation of GEMM tumor cells, enabling a comparison of the direct effects of transplantation on the integrity of the BBTB. Immunofluorescence analysis was performed to compare the structural and subcellular features of tumor-associated vasculature in all models. RESULTS: Contrast enhancement was observed in all transplantation models of MB. No contrast enhancement was observed in the GEMM despite significant tumor burden. Cellular analysis of BBTB integrity revealed aberrancies in all transplantation models, correlating to the varying levels of BBTB permeability observed by MRI in these models. CONCLUSIONS: These results highlight functional differences in the integrity of the BBTB and tumor vessel phenotype between commonly utilized preclinical models of MB, with important implications for the preclinical evaluation of novel therapeutic agents for MB.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Animales , Barrera Hematoencefálica , Línea Celular Tumoral , Niño , Proteínas Hedgehog , Xenoinjertos , Humanos , Ratones
9.
Mol Biol Cell ; 31(23): 2557-2569, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32903148

RESUMEN

Epithelia can eliminate apoptotic cells by apical extrusion. This is a complex morphogenetic event where expulsion of the apoptotic cell is accompanied by rearrangement of its immediate neighbors to form a rosette. A key mechanism for extrusion is constriction of an actomyosin network that neighbor cells form at their interface with the apoptotic cell. Here we report a complementary process of cytoskeletal relaxation that occurs when cortical contractility is down-regulated at the junctions between those neighbor cells themselves. This reflects a mechanosensitive Src family kinase (SFK) signaling pathway that is activated in neighbor cells when the apoptotic cell relaxes shortly after injury. Inhibiting SFK signaling blocks both the expulsion of apoptotic cells and the rosette formation among their neighbor cells. This reveals the complex pattern of spatially distinct contraction and relaxation that must be established in the neighboring epithelium for apoptotic cells to be extruded.


Asunto(s)
Uniones Adherentes/metabolismo , Apoptosis/fisiología , Familia-src Quinasas/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Uniones Adherentes/fisiología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/fisiología , Humanos , Células MCF-7 , Morfogénesis , Transducción de Señal , Familia-src Quinasas/fisiología
10.
Development ; 147(18)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32839180

RESUMEN

The lymphatic vasculature develops primarily from pre-existing veins. A pool of lymphatic endothelial cells (LECs) first sprouts from cardinal veins followed by migration and proliferation to colonise embryonic tissues. Although much is known about the molecular regulation of LEC fate and sprouting during early lymphangiogenesis, we know far less about the instructive and permissive signals that support LEC migration through the embryo. Using a forward genetic screen, we identified mbtps1 and sec23a, components of the COP-II protein secretory pathway, as essential for developmental lymphangiogenesis. In both mutants, LECs initially depart the cardinal vein but then fail in their ongoing migration. A key cargo that failed to be secreted in both mutants was a type II collagen (Col2a1). Col2a1 is normally secreted by notochord sheath cells, alongside which LECs migrate. col2a1a mutants displayed defects in the migratory behaviour of LECs and failed lymphangiogenesis. These studies thus identify Col2a1 as a key cargo secreted by notochord sheath cells and required for the migration of LECs. These findings combine with our current understanding to suggest that successive cell-to-cell and cell-matrix interactions regulate the migration of LECs through the embryonic environment during development.


Asunto(s)
Movimiento Celular/fisiología , Colágeno Tipo II/metabolismo , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Pez Cebra/metabolismo , Animales , Comunicación Celular/fisiología , Proliferación Celular/fisiología , Linfangiogénesis/fisiología , Morfogénesis/fisiología , Venas/metabolismo
11.
Dev Cell ; 49(2): 279-292.e5, 2019 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-31014480

RESUMEN

The correct assignment of cell fate within fields of multipotent progenitors is essential for accurate tissue diversification. The first lymphatic vessels arise from pre-existing veins after venous endothelial cells become specified as lymphatic progenitors. Prox1 specifies lymphatic fate and labels these progenitors; however, the mechanisms restricting Prox1 expression and limiting the progenitor pool remain unknown. We identified a zebrafish mutant that displayed premature, expanded, and prolonged lymphatic specification. The gene responsible encodes the regulator of alternative splicing, Nova2. In zebrafish and human endothelial cells, Nova2 selectively regulates pre-mRNA splicing for components of signaling pathways and phosphoproteins. Nova2-deficient endothelial cells display increased Mapk/Erk signaling, and Prox1 expression is dynamically controlled by Erk signaling. We identify a mechanism whereby Nova2-regulated splicing constrains Erk signaling, thus limiting lymphatic progenitor cell specification. This identifies the capacity of a factor that tunes mRNA splicing to control assignment of cell fate during vascular differentiation.


Asunto(s)
Vasos Linfáticos/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Empalme Alternativo , Animales , Diferenciación Celular , Linaje de la Célula , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Linfangiogénesis , Vasos Linfáticos/citología , Masculino , Antígeno Ventral Neuro-Oncológico , Proteínas Supresoras de Tumor/metabolismo , Venas/citología , Venas/metabolismo , Pez Cebra
12.
Development ; 146(2)2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30642834

RESUMEN

Mural cells (MCs) are essential for blood vessel stability and function; however, the mechanisms that regulate MC development remain incompletely understood, in particular those involved in MC specification. Here, we investigated the first steps of MC formation in zebrafish using transgenic reporters. Using pdgfrb and abcc9 reporters, we show that the onset of expression of abcc9, a pericyte marker in adult mice and zebrafish, occurs almost coincidentally with an increment in pdgfrb expression in peri-arterial mesenchymal cells, suggesting that these transcriptional changes mark the specification of MC lineage cells from naïve pdgfrblow mesenchymal cells. The emergence of peri-arterial pdgfrbhigh MCs required Notch signaling. We found that pdgfrb-positive cells express notch2 in addition to notch3, and although depletion of notch2 or notch3 failed to block MC emergence, embryos depleted of both notch2 and notch3 lost mesoderm- as well as neural crest-derived pdgfrbhigh MCs. Using reporters that read out Notch signaling and Notch2 receptor cleavage, we show that Notch activation in the mesenchyme precedes specification into pdgfrbhigh MCs. Taken together, these results show that Notch signaling is necessary for peri-arterial MC specification.


Asunto(s)
Arterias/citología , Arterias/embriología , Tipificación del Cuerpo , Mesodermo/embriología , Receptores Notch/metabolismo , Transducción de Señal , Pez Cebra/embriología , Animales , Biomarcadores/metabolismo , Endotelio Vascular/metabolismo , Mesodermo/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Imagen de Lapso de Tiempo , Factor de Crecimiento Transformador beta/metabolismo
13.
Nat Commun ; 9(1): 3545, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30171187

RESUMEN

Angiogenesis and vascular remodeling are driven by extensive endothelial cell movements. Here, we present in vivo evidence that endothelial cell movements are associated with oscillating lamellipodia-like structures, which emerge from cell junctions in the direction of cell movements. High-resolution time-lapse imaging of these junction-based lamellipodia (JBL) shows dynamic and distinct deployment of junctional proteins, such as F-actin, VE-cadherin and ZO1, during JBL oscillations. Upon initiation, F-actin and VE-cadherin are broadly distributed within JBL, whereas ZO1 remains at cell junctions. Subsequently, a new junction is formed at the front of the JBL, which then merges with the proximal junction. Rac1 inhibition interferes with JBL oscillations and disrupts cell elongation-similar to a truncation in ve-cadherin preventing VE-cad/F-actin interaction. Taken together, our observations suggest an oscillating ratchet-like mechanism, which is used by endothelial cells to move over each other and thus provides the physical means for cell rearrangements.


Asunto(s)
Actinas/metabolismo , Antígenos CD/fisiología , Cadherinas/fisiología , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Seudópodos/fisiología , Animales , Animales Modificados Genéticamente , Comunicación Celular/fisiología , Embrión no Mamífero , Uniones Intercelulares/fisiología , Proteínas de Pez Cebra/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
14.
Development ; 145(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752386

RESUMEN

Atrial natriuretic peptide (nppa/anf) and brain natriuretic peptide (nppb/bnp) form a gene cluster with expression in the chambers of the developing heart. Despite restricted expression, a function in cardiac development has not been demonstrated by mutant analysis. This is attributed to functional redundancy; however, their genomic location in cis has impeded formal analysis. Using genome editing, we have generated mutants for nppa and nppb, and found that single mutants were indistinguishable from wild type, whereas nppa/nppb double mutants displayed heart morphogenesis defects and pericardial oedema. Analysis of atrioventricular canal (AVC) markers show expansion of bmp4, tbx2b, has2 and versican expression into the atrium of double mutants. This expanded expression correlates with increased extracellular matrix in the atrium. Using a biosensor for hyaluronic acid to measure the cardiac jelly (cardiac extracellular matrix), we confirmed cardiac jelly expansion in nppa/nppb double mutants. Finally, bmp4 knockdown rescued the expansion of has2 expression and cardiac jelly in double mutants. This definitively shows that nppa and nppb function redundantly during cardiac development to restrict gene expression to the AVC, preventing excessive cardiac jelly synthesis in the atrial chamber.


Asunto(s)
Factor Natriurético Atrial/genética , Corazón/embriología , Péptido Natriurético Encefálico/genética , Receptores del Factor Natriurético Atrial/genética , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Edición Génica , Cardiopatías Congénitas/genética , Hialuronano Sintasas/metabolismo , Proteínas de Dominio T Box/metabolismo , Versicanos/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Trends Cell Biol ; 28(3): 173-175, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29305160

RESUMEN

Vascular barrier function is controlled at cell-cell junctions in response to blood flow, but how vascular endothelial cells sense and respond to flow remains to be understood. A recent study describes a flow-sensing pathway involving non-canonical Notch and cadherin signaling that sheds new light on mechanisms controlling the endothelial barrier.


Asunto(s)
Uniones Adherentes , Antígenos CD , Cadherinas , Células Cultivadas , Células Endoteliales , Endotelio Vascular , Uniones Intercelulares
16.
Nat Neurosci ; 20(6): 774-783, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28459441

RESUMEN

Mural cells of the vertebrate brain maintain vascular integrity and function, play roles in stroke and are involved in maintenance of neural stem cells. However, the origins, diversity and roles of mural cells remain to be fully understood. Using transgenic zebrafish, we identified a population of isolated mural lymphatic endothelial cells surrounding meningeal blood vessels. These meningeal mural lymphatic endothelial cells (muLECs) express lymphatic endothelial cell markers and form by sprouting from blood vessels. In larvae, muLECs develop from a lymphatic endothelial loop in the midbrain into a dispersed, nonlumenized mural lineage. muLEC development requires normal signaling through the Vegfc-Vegfd-Ccbe1-Vegfr3 pathway. Mature muLECs produce vascular growth factors and accumulate low-density lipoproteins from the bloodstream. We find that muLECs are essential for normal meningeal vascularization. Together, these data identify an unexpected lymphatic lineage and developmental mechanism necessary for establishing normal meningeal blood vasculature.


Asunto(s)
Células Endoteliales/fisiología , Meninges/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Factores de Crecimiento Endotelial Vascular/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra , Animales , Animales Modificados Genéticamente , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Femenino , Lipoproteínas LDL/metabolismo , Masculino , Meninges/crecimiento & desarrollo , Meninges/metabolismo , Meninges/fisiología , Transducción de Señal/fisiología , Factores de Crecimiento Endotelial Vascular/biosíntesis , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
18.
Dev Cell ; 40(2): 123-136, 2017 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28118600

RESUMEN

Angiogenesis is responsible for tissue vascularization during development, as well as in pathological contexts, including cancer and ischemia. Vascular endothelial growth factors (VEGFs) regulate angiogenesis by acting through VEGF receptors to induce endothelial cell signaling. VEGF is processed in the extracellular matrix (ECM), but the complexity of ECM control of VEGF signaling and angiogenesis remains far from understood. In a forward genetic screen, we identified angiogenesis defects in tmem2 zebrafish mutants that lack both arterial and venous Vegf/Vegfr/Erk signaling. Strikingly, tmem2 mutants display increased hyaluronic acid (HA) surrounding developing vessels. Angiogenesis in tmem2 mutants was rescued, or restored after failed sprouting, by degrading this increased HA. Furthermore, oligomerized HA or overexpression of Vegfc rescued angiogenesis in tmem2 mutants. Based on these data, and the known structure of Tmem2, we find that Tmem2 regulates HA turnover to promote normal Vegf signaling during developmental angiogenesis.


Asunto(s)
Embrión no Mamífero/metabolismo , Ácido Hialurónico/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Arterias/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Membrana/química , Mutación/genética , Neovascularización Fisiológica , Fenotipo , Polimerizacion , Torso/irrigación sanguínea , Venas/metabolismo , Proteínas de Pez Cebra/química
19.
Sci Rep ; 6: 24658, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27145952

RESUMEN

Polycomb group (PcG) proteins are transcriptional repressors of numerous genes, many of which regulate cell cycle progression or developmental processes. We used zebrafish to study Enhancer of zeste homolog 2 (Ezh2), the PcG protein responsible for placing the transcriptional repressive H3K27me3 mark. We identified a nonsense mutant of ezh2 and generated maternal zygotic (MZ) ezh2 mutant embryos. In contrast to knockout mice for PcG proteins, MZezh2 mutant embryos gastrulate seemingly normal, but die around 2 days post fertilization displaying pleiotropic phenotypes. Expression analyses indicated that genes important for early development are not turned off properly, revealing a regulatory role for Ezh2 during zygotic gene expression. In addition, we suggest that Ezh2 regulates maternal mRNA loading of zygotes. Analyses of tissues arising later in development, such as heart, liver, and pancreas, indicated that Ezh2 is required for maintenance of differentiated cell fates. Our data imply that the primary role of Ezh2 is to maintain tissues after tissue specification. Furthermore, our work indicates that Ezh2 is essential to sustain tissue integrity and to set up proper maternal mRNA contribution, and presents a novel and powerful tool to study how PcG proteins contribute to early vertebrate development.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas de Pez Cebra/genética , Animales , Diferenciación Celular , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Desarrollo Embrionario/fisiología , Proteína Potenciadora del Homólogo Zeste 2/deficiencia , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Tracto Gastrointestinal/crecimiento & desarrollo , Expresión Génica , Genotipo , Corazón/crecimiento & desarrollo , Histonas/genética , Histonas/metabolismo , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Hibridación Fluorescente in Situ , Miocardio/metabolismo , ARN Mensajero/metabolismo , Imagen de Lapso de Tiempo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo , Cigoto/metabolismo
20.
Genes Dev ; 29(15): 1618-30, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26253536

RESUMEN

The lymphatic vasculature plays roles in tissue fluid balance, immune cell trafficking, fatty acid absorption, cancer metastasis, and cardiovascular disease. Lymphatic vessels form by lymphangiogenesis, the sprouting of new lymphatics from pre-existing vessels, in both development and disease contexts. The apical signaling pathway in lymphangiogenesis is the VEGFC/VEGFR3 pathway, yet how signaling controls cellular transcriptional output remains unknown. We used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for lymphatic vessel development. We found that mafba is required for the migration of lymphatic precursors after their initial sprouting from the posterior cardinal vein. mafba expression is enriched in sprouts emerging from veins, and we show that mafba functions cell-autonomously during lymphatic vessel development. Mechanistically, Vegfc signaling increases mafba expression to control downstream transcription, and this regulatory relationship is dependent on the activity of SoxF transcription factors, which are essential for mafba expression in venous endothelium. Here we identify an indispensable Vegfc-SoxF-Mafba pathway in lymphatic development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Linfangiogénesis/genética , Vasos Linfáticos/embriología , Factor de Transcripción MafB/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Movimiento Celular/genética , Embrión no Mamífero , Factor de Transcripción MafB/genética , Mutación , Proteínas del Tejido Nervioso/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...