Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-37808725

RESUMO

In brief: The mechanisms regulating the signaling pathways involved in angiogenesis are not fully known. Ristori et al. show that Lunatic Fringe (LFng) mediates the crosstalk between Bone Morphogenic Protein 9 (Bmp9) and Notch signaling, thereby regulating the endothelial cell behavior and temporal dynamics of their identity during sprouting angiogenesis. Highlights: Bmp9 upregulates the expression of LFng in endothelial cells.LFng regulates the temporal dynamics of tip/stalk selection and rearrangement.LFng indicated to play a role in hereditary hemorrhagic telangiectasia.Bmp9 and LFng mediate the endothelial cell-pericyte crosstalk.Bone Morphogenic Protein 9 (Bmp9), whose signaling through Activin receptor-like kinase 1 (Alk1) is involved in several diseases, has been shown to independently activate Notch target genes in an additive fashion with canonical Notch signaling. Here, by integrating predictive computational modeling validated with experiments, we uncover that Bmp9 upregulates Lunatic Fringe (LFng) in endothelial cells (ECs), and thereby also regulates Notch activity in an inter-dependent, multiplicative fashion. Specifically, the Bmp9-upregulated LFng enhances Notch receptor activity creating a much stronger effect when Dll4 ligands are also present. During sprouting, this LFng regulation alters vessel branching by modulating the timing of EC phenotype selection and rearrangement. Our results further indicate that LFng can play a role in Bmp9-related diseases and in pericyte-driven vessel stabilization, since we find LFng contributes to Jag1 upregulation in Bmp9-stimulated ECs; thus, Bmp9-upregulated LFng results in not only enhanced EC Dll4-Notch1 activation, but also Jag1-Notch3 activation in pericytes.

2.
Methods Mol Biol ; 2608: 389-407, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36653719

RESUMO

The movement of groups of cells by collective cell migration requires division of labor between group members. Therefore, distinct cell identities, unique cell behaviors, and specific cellular roles are acquired by cells undergoing collective movement. A key driving force behind the acquisition of discrete cell states is the precise control of where, when, and how genes are expressed, both at the subcellular and supracellular level. Unraveling the mechanisms underpinning the spatiotemporal control of gene expression in collective cell migration requires not only suitable experimental models but also high-resolution imaging of messenger RNA and protein localization during this process. In recent times, the highly stereotyped growth of new blood vessels by sprouting angiogenesis has become a paradigm for understanding collective cell migration, and consequently this has led to the development of numerous user-friendly in vitro models of angiogenesis. In parallel, single-molecule fluorescent in situ hybridization (smFISH) has come to the fore as a powerful technique that allows quantification of both RNA number and RNA spatial distribution in cells and tissues. Moreover, smFISH can be combined with immunofluorescence to understand the precise interrelationship between RNA and protein distribution. Here, we describe methods for use of smFISH and immunofluorescence microscopy in in vitro angiogenesis models to enable the investigation of RNA and protein expression and localization during endothelial collective cell migration.


Assuntos
RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Hibridização in Situ Fluorescente/métodos , RNA/genética , Movimento Celular , Transporte Proteico
4.
Philos Trans R Soc Lond B Biol Sci ; 376(1821): 20190753, 2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-33550953

RESUMO

How do cells make efficient collective decisions during tissue morphogenesis? Humans and other organisms use feedback between movement and sensing known as 'sensorimotor coordination' or 'active perception' to inform behaviour, but active perception has not before been investigated at a cellular level within organs. Here we provide the first proof of concept in silico/in vivo study demonstrating that filopodia (actin-rich, dynamic, finger-like cell membrane protrusions) play an unexpected role in speeding up collective endothelial decisions during the time-constrained process of 'tip cell' selection during blood vessel formation (angiogenesis). We first validate simulation predictions in vivo with live imaging of zebrafish intersegmental vessel growth. Further simulation studies then indicate the effect is due to the coupled positive feedback between movement and sensing on filopodia conferring a bistable switch-like property to Notch lateral inhibition, ensuring tip selection is a rapid and robust process. We then employ measures from computational neuroscience to assess whether filopodia function as a primitive (basal) form of active perception and find evidence in support. By viewing cell behaviour through the 'basal cognitive lens' we acquire a fresh perspective on the tip cell selection process, revealing a hidden, yet vital time-keeping role for filopodia. Finally, we discuss a myriad of new and exciting research directions stemming from our conceptual approach to interpreting cell behaviour. This article is part of the theme issue 'Basal cognition: multicellularity, neurons and the cognitive lens'.


Assuntos
Morfogênese/fisiologia , Pseudópodes/fisiologia , Peixe-Zebra/fisiologia , Actinas/metabolismo , Animais , Simulação por Computador , Percepção
5.
EMBO J ; 39(21): e106003, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32946121

RESUMO

Polarised targeting of diverse mRNAs to cellular protrusions is a hallmark of cell migration. Although a widespread phenomenon, definitive functions for endogenous targeted mRNAs and their relevance to modulation of in vivo tissue dynamics remain elusive. Here, using single-molecule analysis, gene editing and zebrafish live-cell imaging, we report that mRNA polarisation acts as a molecular compass that orients motile cell polarity and spatially directs tissue movement. Clustering of protrusion-derived RNAseq datasets defined a core 192-nt localisation element underpinning precise mRNA targeting to sites of filopodia formation. Such targeting of the small GTPase RAB13 generated tight spatial coupling of mRNA localisation, translation and protein activity, achieving precise subcellular compartmentalisation of RAB13 protein function to create a polarised domain of filopodia extension. Consequently, genomic excision of this localisation element and perturbation of RAB13 mRNA targeting-but not translation-depolarised filopodia dynamics in motile endothelial cells and induced mispatterning of blood vessels in zebrafish. Hence, mRNA polarisation, not expression, is the primary determinant of the site of RAB13 action, preventing ectopic functionality at inappropriate subcellular loci and orienting tissue morphogenesis.


Assuntos
Morfogênese/genética , Morfogênese/fisiologia , RNA Mensageiro/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Movimento Celular , Polaridade Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , GTP Fosfo-Hidrolases , Edição de Genes , Pseudópodes/metabolismo , Pseudópodes/patologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/fisiologia
6.
Anal Bioanal Chem ; 411(25): 6575-6581, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31384985

RESUMO

The application of proteomic liquid chromatography mass spectrometry (LC-MS) for identifying proteins and peptides associated with human disease is rapidly growing in clinical diagnostics. However, the ability to accurately and consistently detect disease-associated peptides remains clinically uncertain. Variability in diagnostic testing occurs in part due to the absence of appropriate reference testing materials and standardised clinical guidelines for proteomic testing. In addition, multiple proteomic testing pipelines have not been fully assessed through external quality assurance (EQA). This trial was therefore devised to evaluate the performance of a small number of mass spectrometry (MS) testing facilities to (i) evaluate the EQA material for potential usage in a proteomic quality assurance program, and to (ii) identify key problem areas associated with human peptide testing. Five laboratories were sent six peptide reference testing samples formulated to contain a total of 35 peptides in differing ratios of light (natural) to heavy (labelled) peptides. Proficiency assessment of laboratory data used a modified approach to similarity and dissimilarity testing that was based on Bray-Curtis and Sorensen indices. Proficiency EQA concordant consensus values could not be derived from the assessed data since none of the laboratories correctly identified all reference testing peptides in all samples. However, the produced data may be reflective of specific inter-laboratory differences for detecting multiple peptides since no two testing pipelines used were the same for any laboratory. In addition, laboratory feedback indicated that peptide filtering of the reference material was a common key problem area prior to analysis. These data highlight the importance of an EQA programme for identifying underlying testing issues so that improvements can be made and confidence for clinical diagnostic analysis can be attained.


Assuntos
Peptídeos/análise , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia Líquida de Alta Pressão/normas , Humanos , Proteômica/métodos , Proteômica/normas , Controle de Qualidade , Padrões de Referência , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/normas
7.
Essays Biochem ; 63(5): 595-606, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31324705

RESUMO

Cell migration is a fundamental biological process involved in tissue formation and homeostasis. The correct polarization of motile cells is critical to ensure directed movement, and is orchestrated by many intrinsic and extrinsic factors. Of these, the subcellular distribution of mRNAs and the consequent spatial control of translation are key modulators of cell polarity. mRNA transport is dependent on cis-regulatory elements within transcripts, which are recognized by trans-acting proteins that ensure the efficient delivery of certain messages to the leading edge of migrating cells. At their destination, translation of localized mRNAs then participates in regional cellular responses underlying cell motility. In this review, we summarize the key findings that established mRNA targetting as a critical driver of cell migration and how the characterization of polarized mRNAs in motile cells has been expanded from just a few species to hundreds of transcripts. We also describe the molecular control of mRNA trafficking, subsequent mechanisms of local protein synthesis and how these ultimately regulate cell polarity during migration.


Assuntos
Movimento Celular/fisiologia , RNA Mensageiro/metabolismo , Actinas/metabolismo , Animais , Extensões da Superfície Celular/fisiologia , Humanos , Microtúbulos/metabolismo , Biossíntese de Proteínas/fisiologia , Transporte de RNA/fisiologia
8.
Cell Rep ; 27(11): 3139-3151.e5, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189101

RESUMO

Angiogenesis is driven by the coordinated collective branching of specialized leading "tip" and trailing "stalk" endothelial cells (ECs). While Notch-regulated negative feedback suppresses excessive tip selection, roles for positive feedback in EC identity decisions remain unexplored. Here, by integrating computational modeling with in vivo experimentation, we reveal that positive feedback critically modulates the magnitude, timing, and robustness of angiogenic responses. In silico modeling predicts that positive-feedback-mediated amplification of VEGF signaling generates an ultrasensitive bistable switch that underpins quick and robust tip-stalk decisions. In agreement, we define a positive-feedback loop exhibiting these properties in vivo, whereby Vegf-induced expression of the atypical tetraspanin, tm4sf18, amplifies Vegf signaling to dictate the speed and robustness of EC selection for angiogenesis. Consequently, tm4sf18 mutant zebrafish select fewer motile ECs and exhibit stunted hypocellular vessels with unstable tip identity that is severely perturbed by even subtle Vegfr attenuation. Hence, positive feedback spatiotemporally shapes the angiogenic switch to ultimately modulate vascular network topology.


Assuntos
Retroalimentação Fisiológica , Neovascularização Fisiológica , Animais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Receptores Notch/metabolismo , Tetraspaninas/genética , Tetraspaninas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Genes Dev ; 30(20): 2297-2309, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27898394

RESUMO

Angiogenesis, the fundamental process by which new blood vessels form from existing ones, depends on precise spatial and temporal gene expression within specific compartments of the endothelium. However, the molecular links between proangiogenic signals and downstream gene expression remain unclear. During sprouting angiogenesis, the specification of endothelial cells into the tip cells that lead new blood vessel sprouts is coordinated by vascular endothelial growth factor A (VEGFA) and Delta-like ligand 4 (Dll4)/Notch signaling and requires high levels of Notch ligand DLL4. Here, we identify MEF2 transcription factors as crucial regulators of sprouting angiogenesis directly downstream from VEGFA. Through the characterization of a Dll4 enhancer directing expression to endothelial cells at the angiogenic front, we found that MEF2 factors directly transcriptionally activate the expression of Dll4 and many other key genes up-regulated during sprouting angiogenesis in both physiological and tumor vascularization. Unlike ETS-mediated regulation, MEF2-binding motifs are not ubiquitous to all endothelial gene enhancers and promoters but are instead overrepresented around genes associated with sprouting angiogenesis. MEF2 target gene activation is directly linked to VEGFA-induced release of repressive histone deacetylases and concurrent recruitment of the histone acetyltransferase EP300 to MEF2 target gene regulatory elements, thus establishing MEF2 factors as the transcriptional effectors of VEGFA signaling during angiogenesis.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição MEF2/metabolismo , Neovascularização Fisiológica/genética , Animais , Células Cultivadas , Embrião não Mamífero , Células Endoteliais/enzimologia , Elementos Facilitadores Genéticos/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fatores de Transcrição MEF2/química , Fatores de Transcrição MEF2/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neovascularização Patológica/genética , Domínios e Motivos de Interação entre Proteínas , Retina/embriologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
11.
Nat Cell Biol ; 18(12): 1292-1301, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27870831

RESUMO

The asymmetric division of stem or progenitor cells generates daughters with distinct fates and regulates cell diversity during tissue morphogenesis. However, roles for asymmetric division in other more dynamic morphogenetic processes, such as cell migration, have not previously been described. Here we combine zebrafish in vivo experimental and computational approaches to reveal that heterogeneity introduced by asymmetric division generates multicellular polarity that drives coordinated collective cell migration in angiogenesis. We find that asymmetric positioning of the mitotic spindle during endothelial tip cell division generates daughters of distinct size with discrete 'tip' or 'stalk' thresholds of pro-migratory Vegfr signalling. Consequently, post-mitotic Vegfr asymmetry drives Dll4/Notch-independent self-organization of daughters into leading tip or trailing stalk cells, and disruption of asymmetry randomizes daughter tip/stalk selection. Thus, asymmetric division seamlessly integrates cell proliferation with collective migration, and, as such, may facilitate growth of other collectively migrating tissues during development, regeneration and cancer invasion.


Assuntos
Divisão Celular Assimétrica , Movimento Celular , Neovascularização Fisiológica , Animais , Polaridade Celular , Tamanho Celular , Simulação por Computador , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Mitose , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Notch , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais , Imagem com Lapso de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
12.
Curr Biol ; 22(19): 1789-94, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-22921365

RESUMO

Tissue branching morphogenesis requires the hierarchical organization of sprouting cells into leading "tip" and trailing "stalk" cells [1, 2]. During new blood vessel branching (angiogenesis), endothelial tip cells (TCs) lead sprouting vessels, extend filopodia, and migrate in response to gradients of the secreted ligand, vascular endothelial growth factor (Vegf) [3]. In contrast, adjacent stalk cells (SCs) trail TCs, generate the trunk of new vessels, and critically maintain connectivity with parental vessels. Here, we establish that h2.0-like homeobox-1 (Hlx1) determines SC potential, which is critical for angiogenesis during zebrafish development. By combining a novel pharmacological strategy for the manipulation of angiogenic cell behavior in vivo with transcriptomic analyses of sprouting cells, we identify the uniquely sprouting-associated gene, hlx1. Expression of hlx1 is almost entirely restricted to sprouting endothelial cells and is excluded from adjacent nonangiogenic cells. Furthermore, Hlx1 knockdown reveals its essential role in angiogenesis. Importantly, mosaic analyses uncover a cell-autonomous role for Hlx1 in the maintenance of SC identity in sprouting vessels. Hence, Hlx1-mediated maintenance of SC potential regulates angiogenesis, a finding that may have novel implications for sprouting morphogenesis of other tissues.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Proteínas de Homeodomínio/metabolismo , Neovascularização Fisiológica/fisiologia , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Embrião não Mamífero , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
13.
Nat Rev Mol Cell Biol ; 12(9): 551-64, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21860391

RESUMO

The vertebrate vasculature forms an extensive branched network of blood vessels that supplies tissues with nutrients and oxygen. During vascular development, coordinated control of endothelial cell behaviour at the levels of cell migration, proliferation, polarity, differentiation and cell-cell communication is critical for functional blood vessel morphogenesis. Recent data uncover elaborate transcriptional, post-transcriptional and post-translational mechanisms that fine-tune key signalling pathways (such as the vascular endothelial growth factor and Notch pathways) to control endothelial cell behaviour during blood vessel sprouting (angiogenesis). These emerging frameworks controlling angiogenesis provide unique insights into fundamental biological processes common to other systems, such as tissue branching morphogenesis, mechanotransduction and tubulogenesis.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Células Endoteliais/fisiologia , Morfogênese/genética , Neovascularização Fisiológica/genética , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Modelos Biológicos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
14.
Traffic ; 11(1): 161-74, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19883397

RESUMO

Vascular endothelial growth factor A (VEGF-A)-induced signaling through VEGF receptor 2 (VEGFR2) regulates both physiological and pathological angiogenesis in mammals. However, the temporal and spatial mechanism underlying VEGFR2-mediated intracellular signaling is not clear. Here, we define a pathway for VEGFR2 trafficking and proteolysis that regulates VEGF-A-stimulated signaling and endothelial cell migration. Ligand-stimulated VEGFR2 activation and ubiquitination preceded proteolysis and cytoplasmic domain removal associated with endosomes. A soluble VEGFR2 cytoplasmic domain fragment displayed tyrosine phosphorylation and activation of downstream intracellular signaling. Perturbation of endocytosis by the depletion of either clathrin heavy chain or an ESCRT-0 subunit caused differential effects on ligand-stimulated VEGFR2 proteolysis and signaling. This novel VEGFR2 proteolysis was blocked by the inhibitors of 26S proteasome activity. Inhibition of proteasome activity prolonged VEGF-A-induced intracellular signaling to c-Akt and endothelial nitric oxide synthase (eNOS). VEGF-A-stimulated endothelial cell migration was dependent on VEGFR2 and VEGFR tyrosine kinase activity. Inhibition of proteasome activity in this assay stimulated VEGF-A-mediated endothelial cell migration. VEGFR2 endocytosis, ubiquitination and proteolysis could also be stimulated by a protein kinase C-dependent pathway. Thus, removal of the VEGFR2 carboxyl terminus linked to phosphorylation, ubiquitination and trafficking is necessary for VEGF-stimulated endothelial signaling and cell migration.


Assuntos
Células Endoteliais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/enzimologia , Citoplasma/metabolismo , Eletroforese em Gel de Poliacrilamida , Endossomos/efeitos dos fármacos , Endossomos/enzimologia , Endossomos/metabolismo , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Ligantes , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Lisossomos/metabolismo , Microscopia de Fluorescência , Neovascularização Patológica/enzimologia , Neovascularização Patológica/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Transporte Proteico
15.
Science ; 326(5950): 294-8, 2009 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-19815777

RESUMO

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.


Assuntos
Artérias/embriologia , Células Endoteliais/fisiologia , Efrina-B2/metabolismo , Morfogênese , Receptor EphB4/metabolismo , Células-Tronco/fisiologia , Veias/embriologia , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/embriologia , Artérias/citologia , Movimento Celular , Células Endoteliais/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Veias/citologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
17.
J Biol Chem ; 284(9): 5784-96, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19119141

RESUMO

Release of endothelial cells from contact-inhibition and cell cycle re-entry is required for the induction of new blood vessel formation by angiogenesis. Using a combination of chemical inhibition, loss of function, and gain of function approaches, we demonstrate that endothelial cell cycle re-entry, S phase progression, and subsequent angiogenic tubule formation are dependent upon the activity of cytosolic phospholipase A(2)-alpha (cPLA(2)alpha). Inhibition of cPLA(2)alpha activity and small interfering RNA (siRNA)-mediated knockdown of endogenous cPLA(2)alpha reduced endothelial cell proliferation. In the absence of cPLA(2)alpha activity, endothelial cells exhibited retarded progression from G(1) through S phase, displayed reduced cyclin A/cdk2 expression, and generated less arachidonic acid. In quiescent endothelial cells, cPLA(2)alpha is inactivated upon its sequestration at the Golgi apparatus. Upon the stimulation of endothelial cell proliferation, activation of cPLA(2)alpha by release from the Golgi apparatus was critical to the induction of cyclin A expression and efficient cell cycle progression. Consequently, inhibition of cPLA(2)alpha was sufficient to block angiogenic tubule formation in vitro. Furthermore, the siRNA-mediated retardation of endothelial cell cycle re-entry and proliferation was reversed upon overexpression of an siRNA-resistant form of cPLA(2)alpha. Thus, activation of cPLA(2)alpha acts as a novel mechanism for the regulation of endothelial cell cycle re-entry, cell cycle progression, and angiogenesis.


Assuntos
Ciclo Celular/fisiologia , Endotélio Vascular/citologia , Fosfolipases A2 do Grupo IV/metabolismo , Neovascularização Fisiológica , Ácido Araquidônico/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Citosol , Derme/citologia , Derme/enzimologia , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/enzimologia , Citometria de Fluxo , Imunofluorescência , Complexo de Golgi/metabolismo , Fosfolipases A2 do Grupo IV/antagonistas & inibidores , Fosfolipases A2 do Grupo IV/genética , Humanos , Imunoprecipitação , RNA Interferente Pequeno/farmacologia , Veias Umbilicais/citologia , Veias Umbilicais/enzimologia
18.
Cell Biol Int ; 33(1): 83-91, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18948217

RESUMO

The association of cytosolic phospholipase A(2)-alpha (cPLA(2)alpha) with intracellular membranes is central to the generation of free arachidonic acid and thromboxane A(2) in activated platelets. Despite this, the site and nature of this membrane association has not been fully characterised upon platelet activation. High resolution imaging showed that cPLA(2)alpha was distributed in a partly structured manner throughout the resting platelet. Upon glass activation or thrombin stimulation, cPLA(2)alpha relocated to a peripheral region corresponding to the platelet plasma membrane. Upon thrombin stimulation of platelets a major pool of cPLA(2)alpha was associated with the plasma membrane in an EGTA-resistant manner. EGTA-resistant membrane binding was abolished upon de-polymerisation of actin filaments by DNase I and furthermore, cPLA(2)alpha co-immunoprecipitated with actin upon thrombin stimulation of platelets. Immunofluorescence microscopy studies revealed that, upon platelet activation, cPLA(2)alpha and actin co-localised at the plasma membrane. Thus we have identified a novel mechanism for the interaction of cPLA(2)alpha with its membrane substrate via interaction with actin.


Assuntos
Actinas/metabolismo , Plaquetas/enzimologia , Membrana Celular/enzimologia , Ácido Egtázico/farmacologia , Fosfolipases A2 do Grupo IV/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actinas/ultraestrutura , Ácido Araquidônico/metabolismo , Membrana Celular/efeitos dos fármacos , Desoxirribonuclease I/metabolismo , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Ativação Plaquetária/efeitos dos fármacos , Transporte Proteico , Trombina/farmacologia , Tromboxano A2/farmacologia , Fatores de Tempo
19.
J Biol Chem ; 282(47): 34468-78, 2007 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-17873281

RESUMO

The regulated generation of prostaglandins from endothelial cells is critical to vascular function. Here we identify a novel mechanism for the regulation of endothelial cell prostaglandin generation. Cytosolic phospholipase A(2)-alpha (cPLA(2)alpha) cleaves phospholipids in a Ca(2+)-dependent manner to yield free arachidonic acid and lysophospholipid. Arachidonic acid is then converted into prostaglandins by the action of cyclooxygenase enzymes and downstream synthases. By previously undefined mechanisms, nonconfluent endothelial cells generate greater levels of prostaglandins than confluent cells. Here we demonstrate that Ca(2+)-independent association of cPLA(2)alpha with the Golgi apparatus of confluent endothelial cells correlates with decreased prostaglandin synthesis. Golgi association blocks arachidonic acid release and prevents functional coupling between cPLA(2)alpha and COX-mediated prostaglandin synthesis. When inactivated at the Golgi apparatus of confluent endothelial cells, cPLA(2)alpha is associated with the phospholipid-binding protein annexin A1. Furthermore, the siRNA-mediated knockdown of endogenous annexin A1 significantly reverses the inhibitory effect of confluence on endothelial cell prostaglandin generation. Thus the confluence-dependent interaction of cPLA(2)alpha and annexin A1 at the Golgi acts as a novel molecular switch controlling cPLA(2)alpha activity and endothelial cell prostaglandin generation.


Assuntos
Anexina A1/metabolismo , Dinoprostona/biossíntese , Células Endoteliais/enzimologia , Complexo de Golgi/enzimologia , Fosfolipases A2 do Grupo IV/metabolismo , Anexina A1/antagonistas & inibidores , Ácido Araquidônico/metabolismo , Cálcio/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Lisofosfolipídeos/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Interferente Pequeno/farmacologia
20.
J Biol Chem ; 281(47): 35709-16, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-16966332

RESUMO

Arachidonic acid and its metabolites have been previously implicated in the regulation of endothelial cell proliferation. Arachidonic acid may be liberated from cellular phospholipids by the action of group VIA calcium-independent phospholipase A2 (iPLA2-VIA). Consequently, we tested the hypothesis that iPLA2-VIA activity is linked to the regulation of endothelial cell proliferation. Inhibition of iPLA2 activity by bromoenol lactone (BEL) was sufficient to entirely block endothelial cell growth. BEL dose-dependently inhibited endothelial cell DNA synthesis in a manner that was reversed upon the exogenous addition of arachidonic acid. DNA synthesis was inhibited by the S-isomer and not by the R-isomer of BEL, demonstrating that endothelial cell proliferation is mediated specifically by iPLA2-VIA. iPLA2-VIA activity was critical to the progression of endothelial cells through S phase and is required for the expression of the cyclin A/cdk2 complex. Thus, inhibition of iPLA2-VIA blocks S phase progression and results in exit from the cell cycle. Inhibition of iPLA2-VIA-mediated endothelial cell proliferation is sufficient to block angiogenic tubule formation in co-culture assays. Consequently, iPLA2-VIA is a novel regulator of endothelial cell S phase progression, cell cycle residence, and angiogenesis.


Assuntos
Endotélio Vascular/patologia , Fosfolipases A/metabolismo , Fosfolipases A/fisiologia , Fase S , Ácido Araquidônico/farmacologia , Ciclo Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Fosfolipases A2 do Grupo VI , Humanos , Naftalenos/farmacologia , Neovascularização Fisiológica , Inibidores de Fosfodiesterase/farmacologia , Fosfolipases A2 , Pironas/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...