Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Thromb Haemost ; 21(11): 3056-3066, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37393001

RESUMO

The hemostatic system involves an array of circulating coagulation factors that work in concert with platelets and the vascular endothelium to promote clotting in a space- and time-defined manner. Despite equal systemic exposure to circulating factors, bleeding and thrombotic diseases tend to prefer specific sites, suggesting an important role for local factors. This may be provided by endothelial heterogeneity. Endothelial cells differ not only between arteries, veins, and capillaries but also between microvascular beds from different organs, which present unique organotypic morphology and functional and molecular profiles. Accordingly, regulators of hemostasis are not uniformly distributed in the vasculature. The establishment and maintenance of endothelial diversity are orchestrated at the transcriptional level. Recent transcriptomic and epigenomic studies have provided a global picture of endothelial cell heterogeneity. In this review, we discuss the organotypic differences in the hemostatic profile of endothelial cells; we focus on 2 major endothelial regulators of hemostasis, namely von Willebrand factor and thrombomodulin, to provide examples of transcriptional mechanisms that control heterogeneity; finally, we consider some of the methodological challenges and opportunities for future studies.


Assuntos
Células Endoteliais , Hemostáticos , Humanos , Células Endoteliais/metabolismo , Hemostasia/fisiologia , Endotélio Vascular/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo , Expressão Gênica
2.
Artigo em Inglês | MEDLINE | ID: mdl-35074794

RESUMO

The Wnt/frizzled signaling pathway is one of the major regulators of endothelial biology, controlling key cellular activities. Many secreted Wnt ligands have been identified and can initiate diverse signaling via binding to a complex set of Frizzled (Fzd) transmembrane receptors and coreceptors. Roughly, Wnt signaling is subdivided into two pathways: the canonical Wnt/ß-catenin signaling pathway whose main downstream effector is the transcriptional coactivator ß-catenin, and the noncanonical Wnt signaling pathway, which is subdivided into the Wnt/Ca2+ pathway and the planar cell polarity pathway. Here, we will focus on its cross talk with other angiogenic pathways and on its role in blood-retinal- and blood-brain-barrier formation and its maintenance in a differentiated state. We will unravel how retinal vascular pathologies and neurovascular degenerative diseases result from disruption of the Wnt pathway related to vascular instability, and highlight current research into therapeutic options.


Assuntos
Barreira Hematoencefálica , Via de Sinalização Wnt , Endotélio , Receptores Frizzled , Humanos , Ligantes
3.
Nat Cardiovasc Res ; 1: 882-899, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36713285

RESUMO

Current dogma dictates that during adulthood, endothelial cells (ECs) are locked in an immutable stable homeostatic state. By contrast, herein we show that maintenance of EC fate and function are linked and active processes, which depend on the constitutive cooperativity of only two ETS-transcription factors (TFs) ERG and Fli1. While deletion of either Fli1 or ERG manifest subtle vascular dysfunction, their combined genetic deletion in adult EC results in acute vasculopathy and multiorgan failure, due to loss of EC fate and integrity, hyperinflammation, and spontaneous thrombosis, leading to death. ERG and Fli1 co-deficiency cause rapid transcriptional silencing of pan- and organotypic vascular core genes, with dysregulation of inflammation and coagulation pathways. Vascular hyperinflammation leads to impaired hematopoiesis with myeloid skewing. Accordingly, enforced ERG and FLI1 expression in adult human mesenchymal stromal cells activates vascular programs and functionality enabling engraftment of perfusable vascular network. GWAS-analysis identified vascular diseases are associated with FLI1/Erg mutations. Constitutive expression of ERG and Fli1 uphold EC fate, physiological function, and resilience in adult vasculature; while their functional loss can contribute to systemic human diseases.

6.
FASEB J ; 34(1): 1288-1303, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914666

RESUMO

Retinopathies remain major causes of visual impairment in diabetic patients and premature infants. Introduction of anti-angiogenic drugs targeting vascular endothelial growth factor (VEGF) has transformed therapy for these proliferative retinopathies. However, limitations associated with anti-VEGF medications require to unravel new pathways of vessel growth to identify potential drug targets. Here, we investigated the role of Wnt/Frizzled-7 (Fzd7) pathway in a mouse model of oxygen-induced retinopathy (OIR). Using transgenic mice, which enabled endothelium-specific and time-specific Fzd7 deletion, we demonstrated that Fzd7 controls both vaso-obliteration and neovascular phases (NV). Deletion of Fzd7 at P12, after the ischemic phase of OIR, prevented formation of aberrant neovessels into the vitreous by suppressing proliferation of endothelial cells (EC) in tufts. Next we validated in vitro two Frd7 blocking strategies: a monoclonal antibody (mAbFzd7) against Fzd7 and a soluble Fzd7 receptor (CRD). In vivo a single intravitreal microinjection of mAbFzd7 or CRD significantly attenuated retinal neovascularization (NV) in mice with OIR. Molecular analysis revealed that Fzd7 may act through the activation of Wnt/ß-catenin and Jagged1 expression to control EC proliferation in extra-retinal neovessels. We identified Fzd7/ß-catenin signaling as new regulator of pathological retinal NV. Fzd7 appears to be a potent pharmacological target to prevent or treat aberrant angiogenesis of ischemic retinopathies.


Assuntos
Retinopatia Diabética/metabolismo , Isquemia/metabolismo , Proteínas Repressoras/metabolismo , Neovascularização Retiniana/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Deleção de Genes , Isquemia/genética , Isquemia/patologia , Proteína Jagged-1/biossíntese , Proteína Jagged-1/genética , Camundongos , Camundongos Mutantes , Proteínas Repressoras/genética , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , beta Catenina/genética
7.
Blood ; 133(24): 2559-2569, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-30975637

RESUMO

During wound healing, the distribution, availability, and signaling of growth factors (GFs) are orchestrated by their binding to extracellular matrix components in the wound microenvironment. Extracellular matrix proteins have been shown to modulate angiogenesis and promote wound healing through GF binding. The hemostatic protein von Willebrand factor (VWF) released by endothelial cells (ECs) in plasma and in the subendothelial matrix has been shown to regulate angiogenesis; this function is relevant to patients in whom VWF deficiency or dysfunction is associated with vascular malformations. Here, we show that VWF deficiency in mice causes delayed wound healing accompanied by decreased angiogenesis and decreased amounts of angiogenic GFs in the wound. We show that in vitro VWF binds to several GFs, including vascular endothelial growth factor-A (VEGF-A) isoforms and platelet-derived growth factor-BB (PDGF-BB), mainly through the heparin-binding domain (HBD) within the VWF A1 domain. VWF also binds to VEGF-A and fibroblast growth factor-2 (FGF-2) in human plasma and colocalizes with VEGF-A in ECs. Incorporation of the VWF A1 HBD into fibrin matrices enables sequestration and slow release of incorporated GFs. In vivo, VWF A1 HBD-functionalized fibrin matrices increased angiogenesis and GF retention in VWF-deficient mice. Treatment of chronic skin wounds in diabetic mice with VEGF-A165 and PDGF-BB incorporated within VWF A1 HBD-functionalized fibrin matrices accelerated wound healing, with increased angiogenesis and smooth muscle cell proliferation. Therefore, the VWF A1 HBD can function as a GF reservoir, leading to effective angiogenesis and tissue regeneration.


Assuntos
Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Fator de von Willebrand/metabolismo , Animais , Diabetes Mellitus Experimental , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Domínios Proteicos
8.
Circ Res ; 124(9): 1337-1349, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30892142

RESUMO

RATIONALE: The ETS (E-26 transformation-specific) transcription factor ERG (ETS-related gene) is essential for endothelial homeostasis, driving expression of lineage genes and repressing proinflammatory genes. Loss of ERG expression is associated with diseases including atherosclerosis. ERG's homeostatic function is lineage-specific, because aberrant ERG expression in cancer is oncogenic. The molecular basis for ERG lineage-specific activity is unknown. Transcriptional regulation of lineage specificity is linked to enhancer clusters (super-enhancers). OBJECTIVE: To investigate whether ERG regulates endothelial-specific gene expression via super-enhancers. METHODS AND RESULTS: Chromatin immunoprecipitation with high-throughput sequencing in human umbilical vein endothelial cells showed that ERG binds 93% of super-enhancers ranked according to H3K27ac, a mark of active chromatin. These were associated with endothelial genes such as DLL4 (Delta-like protein 4), CLDN5 (claudin-5), VWF (von Willebrand factor), and CDH5 (VE-cadherin). Comparison between human umbilical vein endothelial cell and prostate cancer TMPRSS2 (transmembrane protease, serine-2):ERG fusion-positive human prostate epithelial cancer cell line (VCaP) cells revealed distinctive lineage-specific transcriptome and super-enhancer profiles. At a subset of endothelial super-enhancers (including DLL4 and CLDN5), loss of ERG results in significant reduction in gene expression which correlates with decreased enrichment of H3K27ac and MED (Mediator complex subunit)-1, and reduced recruitment of acetyltransferase p300. At these super-enhancers, co-occupancy of GATA2 (GATA-binding protein 2) and AP-1 (activator protein 1) is significantly lower compared with super-enhancers that remained constant following ERG inhibition. These data suggest distinct mechanisms of super-enhancer regulation in endothelial cells and highlight the unique role of ERG in controlling a core subset of super-enhancers. Most disease-associated single nucleotide polymorphisms from genome-wide association studies lie within noncoding regions and perturb transcription factor recognition sequences in relevant cell types. Analysis of genome-wide association studies data shows significant enrichment of risk variants for cardiovascular disease and other diseases, at ERG endothelial enhancers and super-enhancers. CONCLUSIONS: The transcription factor ERG promotes endothelial homeostasis via regulation of lineage-specific enhancers and super-enhancers. Enrichment of cardiovascular disease-associated single nucleotide polymorphisms at ERG super-enhancers suggests that ERG-dependent transcription modulates disease risk.


Assuntos
Elementos Facilitadores Genéticos/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Linhagem Celular Tumoral , Células Cultivadas , Claudina-5/genética , Claudina-5/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Regulador Transcricional ERG/genética
9.
J Thromb Haemost ; 17(5): 827-840, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30801958

RESUMO

Essentials To reliably study the respective roles of blood and endothelial cells in hemostasis, mouse models with a strong and specific endothelial expression of the Cre recombinase are needed. Using mT/mG reporter mice and conditional JAK2V617F/WT mice, we compared Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 with well-characterized Tie2-Cre mice. Comparison of recombination efficiency and specificity towards blood lineage reveals major differences between endothelial transgenic mice. Cre-mediated recombination occurs in a small number of adult hematopoietic stem cells in Pdgfb-iCreERT2;JAK2V617F/WT transgenic mice. SUMMARY: Background The vessel wall, and particularly blood endothelial cells (BECs), are intensively studied to better understand hemostasis and target thrombosis. To understand the specific role of BECs, it is important to have mouse models that allow specific and homogeneous expression of genes of interest in all BEC beds without concomitant expression in blood cells. Inducible Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 transgenic mice are widely used for BEC targeting. However, issues remain in terms of recombination efficiency and specificity regarding hematopoietic cells. Objectives To determine which mouse model to choose when strong expression of a transgene is required in adult BECs from various organs, without concomitant expression in hematopoietic cells. Methods Using mT/mG reporter mice to measure recombination efficiency and conditional JAK2V617F/WT mice to assess specificity regarding hematopoietic cells, we compared Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 with well-characterized Tie2-Cre mice. Results Adult Cdh5(PAC)-CreERT2 mice are endothelial specific but require a dose of 10 mg of tamoxifen to allow constant Cre expression. Pdgfb-iCreERT2 mice injected with 5 mg of tamoxifen are appropriate for most endothelial research fields except liver studies, as hepatic sinusoid ECs are not recombined. Surprisingly, 2 months after induction of Cre-mediated recombination, all Pdgfb-iCreERT2;JAK2V617F/WT mice developed a myeloproliferative neoplasm that is related to the presence of JAK2V617F in hematopoietic cells, showing for the first time that Cre-mediated recombination occurs in a small number of adult hematopoietic stem cells in Pdgfb-iCreERT2 transgenic mice. Conclusion This study provides useful guidelines for choosing the best mouse line to study the role of BECs in hemostasis and thrombosis.


Assuntos
Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Linfocinas/genética , Linfocinas/metabolismo , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Alelos , Animais , Encéfalo/metabolismo , Hemostasia , Integrases/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Reação em Cadeia da Polimerase , Retina/metabolismo , Tamoxifeno/farmacologia , Trombose/metabolismo
10.
Haematologica ; 104(1): 70-81, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30171023

RESUMO

Thrombosis is the main cause of morbidity and mortality in patients with JAK2V617F myeloproliferative neoplasms. Recent studies have reported the presence of JAK2V617F in endothelial cells of some patients with myeloproliferative neoplasms. We investigated the role of endothelial cells that express JAK2V617F in thrombus formation using an in vitro model of human endothelial cells overexpressing JAK2V617F and an in vivo model of mice with endothelial-specific JAK2V617F expression. Interestingly, these mice displayed a higher propensity for thrombus. When deciphering the mechanisms by which JAK2V617F-expressing endothelial cells promote thrombosis, we observed that they have a pro-adhesive phenotype associated with increased endothelial P-selectin exposure, secondary to degranulation of Weibel-Palade bodies. We demonstrated that P-selectin blockade was sufficient to reduce the increased propensity of thrombosis. Moreover, treatment with hydroxyurea also reduced thrombosis and decreased the pathological interaction between leukocytes and JAK2V617F-expressing endothelial cells through direct reduction of endothelial P-selectin expression. Taken together, our data provide evidence that JAK2V617F-expressing endothelial cells promote thrombosis through induction of endothelial P-selectin expression, which can be reversed by hydroxyurea. Our findings increase our understanding of thrombosis in patients with myeloproliferative neoplasms, at least those with JAK2V617F-positive endothelial cells, and highlight a new role for hydroxyurea. This novel finding provides the proof of concept that an acquired genetic mutation can affect the pro-thrombotic nature of endothelial cells, suggesting that other mutations in endothelial cells could be causal in thrombotic disorders of unknown cause, which account for 50% of recurrent venous thromboses.


Assuntos
Células Endoteliais/metabolismo , Janus Quinase 2/biossíntese , Selectina-P/biossíntese , Trombose/metabolismo , Animais , Modelos Animais de Doenças , Células Endoteliais/patologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiureia/farmacologia , Janus Quinase 2/genética , Camundongos , Camundongos Transgênicos , Selectina-P/genética , Trombose/tratamento farmacológico , Trombose/genética , Trombose/patologia
11.
Nat Commun ; 8(1): 895, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29026072

RESUMO

The role of the endothelium in protecting from chronic liver disease and TGFß-mediated fibrosis remains unclear. Here we describe how the endothelial transcription factor ETS-related gene (ERG) promotes liver homoeostasis by controlling canonical TGFß-SMAD signalling, driving the SMAD1 pathway while repressing SMAD3 activity. Molecular analysis shows that ERG binds to SMAD3, restricting its access to DNA. Ablation of ERG expression results in endothelial-to-mesenchymal transition (EndMT) and spontaneous liver fibrogenesis in EC-specific constitutive hemi-deficient (Erg cEC-Het ) and inducible homozygous deficient mice (Erg iEC-KO ), in a SMAD3-dependent manner. Acute administration of the TNF-α inhibitor etanercept inhibits carbon tetrachloride (CCL4)-induced fibrogenesis in an ERG-dependent manner in mice. Decreased ERG expression also correlates with EndMT in tissues from patients with end-stage liver fibrosis. These studies identify a pathogenic mechanism where loss of ERG causes endothelial-dependent liver fibrogenesis via regulation of SMAD2/3. Moreover, ERG represents a promising candidate biomarker for assessing EndMT in liver disease.The transcription factor ERG is key to endothelial lineage specification and vascular homeostasis. Here the authors show that ERG balances TGFß signalling through the SMAD1 and SMAD3 pathways, protecting the endothelium from endothelial-to-mesenchymal transition and consequent liver fibrosis in mice via a SMAD3-dependent mechanism.


Assuntos
Células Endoteliais/metabolismo , Cirrose Hepática Biliar/patologia , Fígado/patologia , Proteínas Oncogênicas/metabolismo , Regulador Transcricional ERG/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Regulação para Baixo , Doença Hepática Terminal/etiologia , Doença Hepática Terminal/cirurgia , Transição Epitelial-Mesenquimal , Etanercepte/farmacologia , Etanercepte/uso terapêutico , Feminino , Fibrose , Células Endoteliais da Veia Umbilical Humana , Humanos , Fígado/efeitos dos fármacos , Fígado/cirurgia , Cirrose Hepática Biliar/complicações , Cirrose Hepática Biliar/terapia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/tratamento farmacológico , Cirrose Hepática Experimental/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Oncogênicas/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Regulador Transcricional ERG/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regulação para Cima
12.
Arterioscler Thromb Vasc Biol ; 36(12): 2369-2380, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27758766

RESUMO

OBJECTIVE: Vessel formation requires precise orchestration of a series of morphometric and molecular events controlled by a multitude of angiogenic factors and morphogens. Wnt/frizzled signaling is required for proper vascular formation. In this study, we investigated the role of the Fzd7 (frizzled-7) receptor in retinal vascular development and its relationship with the Wnt/ß-catenin canonical pathway and Notch signaling. APPROACH AND RESULTS: Using transgenic mice, we demonstrated that Fzd7 is required for postnatal vascular formation. Endothelial cell (EC) deletion of fzd7 (fzd7ECKO) delayed retinal plexus formation because of an impairment in tip cell phenotype and a decrease in stalk cell proliferation. Dvl (dishevelled) proteins are a main component of Wnt signaling and play a functionally redundant role. We found that Dvl3 depletion in dvl1-/- mice mimicked the fzd7ECKO vascular phenotype and demonstrated that Fzd7 acted via ß-catenin activation by showing that LiCl treatment rescued impairment in tip and stalk cell phenotypes induced in fzd7 mutants. Deletion of fzd7 or Dvl1/3 induced a strong decrease in Wnt canonical genes and Notch partners' expression. Genetic and pharmacological rescue strategies demonstrated that Fzd7 acted via ß-catenin activation, upstream of Notch signaling to control Dll4 and Jagged1 EC expression. CONCLUSIONS: Fzd7 expressed by EC drives postnatal angiogenesis via activation of Dvl/ß-catenin signaling and can control the integrative interaction of Wnt and Notch signaling during postnatal angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica , Receptores Acoplados a Proteínas G/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio , Proliferação de Células , Células Cultivadas , Proteínas Desgrenhadas/deficiência , Proteínas Desgrenhadas/genética , Células Endoteliais/efeitos dos fármacos , Receptores Frizzled , Genótipo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-1/metabolismo , Cloreto de Lítio/farmacologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Interferência de RNA , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Notch/metabolismo , Neovascularização Retiniana/genética , Neovascularização Retiniana/fisiopatologia , Vasos Retinianos/efeitos dos fármacos , Transfecção , Via de Sinalização Wnt/efeitos dos fármacos
13.
Mol Biol Cell ; 27(6): 941-53, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26792835

RESUMO

Angiogenesis involves the coordinated growth and migration of endothelial cells (ECs) toward a proangiogenic signal. The Wnt planar cell polarity (PCP) pathway, through the recruitment of Dishevelled (Dvl) and Dvl-associated activator of morphogenesis (Daam1), has been proposed to regulate cell actin cytoskeleton and microtubule (MT) reorganization for oriented cell migration. Here we report that Kif26b--a kinesin--and Daam1 cooperatively regulate initiation of EC sprouting and directional migration via MT reorganization. First, we find that Kif26b is recruited within the Dvl3/Daam1 complex. Using a three-dimensional in vitro angiogenesis assay, we show that Kif26b and Daam1 depletion impairs tip cell polarization and destabilizes extended vascular processes. Kif26b depletion specifically alters EC directional migration and mislocalized MT organizing center (MTOC)/Golgi and myosin IIB cell rear enrichment. Therefore the cell fails to establish a proper front-rear polarity. Of interest, Kif26b ectopic expression rescues the siDaam1 polarization defect phenotype. Finally, we show that Kif26b functions in MT stabilization, which is indispensable for asymmetrical cell structure reorganization. These data demonstrate that Kif26b, together with Dvl3/Daam1, initiates cell polarity through the control of PCP signaling pathway-dependent activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Polaridade Celular , Proteínas Desgrenhadas/metabolismo , Células Endoteliais/metabolismo , Cinesinas/metabolismo , Via de Sinalização Wnt , Animais , Movimento Celular , Células Endoteliais/fisiologia , Humanos , Camundongos , Proteínas dos Microfilamentos , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neovascularização Fisiológica , Proteínas rho de Ligação ao GTP
14.
Nat Commun ; 5: 4832, 2014 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25198863

RESUMO

Development and stabilization of a vascular plexus requires the coordination of multiple signalling processes. Wnt planar cell polarity (PCP) signalling is critical in vertebrates for diverse morphogenesis events, which coordinate cell orientation within a tissue-specific plane. However, its functional role in vascular morphogenesis is not well understood. Here we identify PDZRN3, an ubiquitin ligase, and report that Pdzrn3 deficiency impairs embryonic angiogenic remodelling and postnatal retinal vascular patterning, with a loss of two-dimensional polarized orientation of the intermediate retinal plexus. Using in vitro and ex vivo Pdzrn3 loss-of-function and gain-of-function experiments, we demonstrate a key role of PDZRN3 in endothelial cell directional and coordinated extension. PDZRN3 ubiquitinates Dishevelled 3 (Dvl3), to promote endocytosis of the Frizzled/Dvl3 complex, for PCP signal transduction. These results highlight the role of PDZRN3 to direct Wnt PCP signalling, and broadly implicate this pathway in the planar orientation and highly branched organization of vascular plexuses.


Assuntos
Vasos Sanguíneos/embriologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Morfogênese/genética , Neovascularização Fisiológica/genética , Ubiquitina-Proteína Ligases/genética , Via de Sinalização Wnt/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Polaridade Celular/genética , Proteínas Desgrenhadas , Endocitose , Receptores Frizzled/metabolismo , Camundongos , Camundongos Knockout , Fosfoproteínas/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Wnt/metabolismo
15.
Cardiovasc Res ; 103(2): 291-303, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24866384

RESUMO

AIMS: Vascular permeability is essential for the health of normal tissues and is an important characteristic of many disease states. The role of the Wnt/frizzled pathway in vascular biology has recently been reported. The objectives of this study are to analyse the role of Frizzled7 (Fzd7) receptor in the control of vascular integrity. METHODS AND RESULTS: Fzd7 is expressed in endothelial cells and accumulates at the points of cell-cell contact in association with VE-cadherin and ß-catenin, two major adherens junction molecules. To selectively delete fzd7 in the vasculature, we developed gene targeting approaches using CreLox strategy in mice. Genetic fzd7 inhibition in the endothelium increases vascular permeability in basal and factor-induced conditions. On the cellular level, fzd7 knockdown or depletion leads to an increase in paracellular permeability with a loss of adherens junction organization. These impairments are associated with a decrease in both VE-Cadherin and ß-catenin expression, a decrease in their association and an increase of tyrosine phosphorylation of VE-cadherin/ß-catenin. Fzd7 transduces a Wnt/ß-catenin signalling cascade that is required to regulate ß-catenin and canonical target gene expression. Finally, LiCl, a GSK3 inhibitor, and ß-catenin overexpression rescued endothelial integrity and adherens junction organization, induced by fzd7 deletion. CONCLUSION: These findings establish that Fzd7 is a new partner of adherens junctional complex and represents a novel molecular switch for the control of vascular permeability via activation of the Wnt-canonical pathway.


Assuntos
Caderinas/metabolismo , Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Comunicação Celular , Endotélio Vascular/metabolismo , Receptores Frizzled , Quinase 3 da Glicogênio Sintase/metabolismo , Junções Intercelulares/metabolismo , Camundongos , Camundongos Transgênicos , Transdução de Sinais/fisiologia , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA