Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Angiogenesis ; 25(3): 411-434, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35320450

RESUMO

The small monomeric GTPase RHOA acts as a master regulator of signal transduction cascades by activating effectors of cellular signaling, including the Rho-associated protein kinases ROCK1/2. Previous in vitro cell culture studies suggest that RHOA can regulate many critical aspects of vascular endothelial cell (EC) biology, including focal adhesion, stress fiber formation, and angiogenesis. However, the specific in vivo roles of RHOA during vascular development and homeostasis are still not well understood. In this study, we examine the in vivo functions of RHOA in regulating vascular development and integrity in zebrafish. We use zebrafish RHOA-ortholog (rhoaa) mutants, transgenic embryos expressing wild type, dominant negative, or constitutively active forms of rhoaa in ECs, pharmacological inhibitors of RHOA and ROCK1/2, and Rock1 and Rock2a/b dgRNP-injected zebrafish embryos to study the in vivo consequences of RHOA gain- and loss-of-function in the vascular endothelium. Our findings document roles for RHOA in vascular integrity, developmental angiogenesis, and vascular morphogenesis in vivo, showing that either too much or too little RHOA activity leads to vascular dysfunction.


Assuntos
Peixe-Zebra , Proteína rhoA de Ligação ao GTP , Animais , Animais Geneticamente Modificados , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Transdução de Sinais , Peixe-Zebra/genética , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
2.
Development ; 144(11): 2070-2081, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28506987

RESUMO

The lymphatic vascular system is a hierarchically organized complex network essential for tissue fluid homeostasis, immune trafficking and absorption of dietary fats in the human body. Despite its importance, the assembly of the lymphatic network is still not fully understood. The zebrafish is a powerful model organism that enables study of lymphatic vessel development using high-resolution imaging and sophisticated genetic and experimental manipulation. Although several studies have described early lymphatic development in the fish, lymphatic development at later stages has not been completely elucidated. In this study, we generated a new Tg(mrc1a:egfp)y251 transgenic zebrafish that uses a mannose receptor, C type 1 (mrc1a) promoter to drive strong EGFP expression in lymphatic vessels at all stages of development and in adult zebrafish. We used this line to describe the assembly of the major vessels of the trunk lymphatic vascular network, including the later-developing collateral cardinal, spinal, superficial lateral and superficial intersegmental lymphatics. Our results show that major trunk lymphatic vessels are conserved in the zebrafish, and provide a thorough and complete description of trunk lymphatic vessel assembly.


Assuntos
Sistema Linfático/crescimento & desenvolvimento , Sistema Linfático/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Vasos Linfáticos/metabolismo , Transgenes , Veias/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Cell Rep ; 17(6): 1595-1606, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27806298

RESUMO

All mature blood cell types in the adult animal arise from hematopoietic stem and progenitor cells (HSPCs). However, the developmental cues regulating HSPC ontogeny are incompletely understood. In particular, the details surrounding a requirement for Wnt/ß-catenin signaling in the development of mature HSPCs are controversial and difficult to consolidate. Using zebrafish, we demonstrate that Wnt signaling is required to direct an amplification of HSPCs in the aorta. Wnt9a is specifically required for this process and cannot be replaced by Wnt9b or Wnt3a. This proliferative event occurs independently of initial HSPC fate specification, and the Wnt9a input is required prior to aorta formation. HSPC arterial amplification occurs prior to seeding of secondary hematopoietic tissues and proceeds, in part, through the cell cycle regulator myca (c-myc). Our results support a general paradigm, in which early signaling events, including Wnt, direct later HSPC developmental processes.


Assuntos
Aorta/citologia , Aorta/embriologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Contagem de Células , Ciclo Celular , Proliferação de Células , Hemangioblastos/metabolismo , Via de Sinalização Wnt
4.
Arterioscler Thromb Vasc Biol ; 36(4): 655-62, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26821951

RESUMO

OBJECTIVE: Understanding the mechanisms regulating normal and pathological angiogenesis is of great scientific and clinical interest. In this report, we show that mutations in 2 different aminoacyl-transfer RNA synthetases, threonyl tRNA synthetase (tars(y58)) or isoleucyl tRNA synthetase (iars(y68)), lead to similar increased branching angiogenesis in developing zebrafish. APPROACH AND RESULTS: The unfolded protein response pathway is activated by aminoacyl-transfer RNA synthetase deficiencies, and we show that unfolded protein response genes atf4, atf6, and xbp1, as well as the key proangiogenic ligand vascular endothelial growth factor (vegfaa), are all upregulated in tars(y58) and iars(y68) mutants. Finally, we show that the protein kinase RNA-like endoplasmic reticulum kinase-activating transcription factor 4 arm of the unfolded protein response pathway is necessary for both the elevated vegfaa levels and increased angiogenesis observed in tars(y58) mutants. CONCLUSIONS: Our results suggest that endoplasmic reticulum stress acts as a proangiogenic signal via unfolded protein response pathway-dependent upregulation of vegfaa.


Assuntos
Isoleucina-tRNA Ligase/deficiência , Neovascularização Fisiológica , Treonina-tRNA Ligase/deficiência , Resposta a Proteínas não Dobradas , Proteínas de Peixe-Zebra/deficiência , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Isoleucina-tRNA Ligase/genética , Mutação , Fenótipo , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais , Treonina-tRNA Ligase/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína 1 de Ligação a X-Box , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
5.
Dev Biol ; 390(2): 116-25, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24699544

RESUMO

Initial embryonic determination of artery or vein identity is regulated by genetic factors that work in concert to specify the endothelial cell׳s (EC) fate, giving rise to two structurally unique components of the circulatory loop. The Shh/VEGF/Notch pathway is critical for arterial specification, while the orphan receptor nr2f2 (COUP-TFII) has been implicated in venous specification. Studies in mice have shown that nr2f2 is expressed in venous but not arterial ECs, and that it preferentially induces markers of venous cell fate. We have examined the role of nr2f2 during early arterial-venous development in the zebrafish trunk. We show that expression of a subset of markers of venous endothelial identity requires nr2f2, while the expression of nr2f2 itself requires sox7 and sox18 gene function. However, while sox7 and sox18 are expressed in both the cardinal vein and the dorsal aorta during early trunk development, nr2f2 is expressed only in the cardinal vein. We show that Notch signaling activity present in the dorsal aorta suppresses expression of nr2f2, restricting nr2f2-dependent promotion of venous differentiation to the cardinal vein.


Assuntos
Vasos Sanguíneos/embriologia , Fator II de Transcrição COUP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores Notch/metabolismo , Fatores de Transcrição SOXF/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Fator II de Transcrição COUP/genética , Diferenciação Celular/fisiologia , Clonagem Molecular , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Hibridização In Situ , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXF/genética , Transgenes/genética , Veias/citologia , Veias/embriologia , Proteínas de Peixe-Zebra/genética
6.
Development ; 138(22): 4875-86, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22007135

RESUMO

Here, we show that a novel Rspo1-Wnt-Vegfc-Vegfr3 signaling pathway plays an essential role in developmental angiogenesis. A mutation in R-spondin1 (rspo1), a Wnt signaling regulator, was uncovered during a forward-genetic screen for angiogenesis-deficient mutants in the zebrafish. Embryos lacking rspo1 or the proposed rspo1 receptor kremen form primary vessels by vasculogenesis, but are defective in subsequent angiogenesis. Endothelial cell-autonomous inhibition of canonical Wnt signaling also blocks angiogenesis in vivo. The pro-angiogenic effects of Rspo1/Wnt signaling are mediated by Vegfc/Vegfr3(Flt4) signaling. Vegfc expression is dependent on Rspo1 and Wnt, and Vegfc and Vegfr3 are necessary to promote angiogenesis downstream from Rspo1-Wnt. As all of these molecules are expressed by the endothelium during sprouting stages, these results suggest that Rspo1-Wnt-VegfC-Vegfr3 signaling plays a crucial role as an endothelial-autonomous permissive cue for developmental angiogenesis.


Assuntos
Neovascularização Fisiológica/genética , Fator C de Crescimento do Endotélio Vascular/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Via de Sinalização Wnt/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Modelos Biológicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Trombospondinas , Regulação para Cima/genética , Regulação para Cima/fisiologia , Fator C de Crescimento do Endotélio Vascular/genética , Fator C 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 , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Circ Res ; 104(5): 576-88, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19286613

RESUMO

The major arteries and veins of the vertebrate circulatory system are formed early in embryonic development, before the onset of circulation, following de novo aggregation of "angioblast" progenitors in a process called vasculogenesis. Initial embryonic determination of artery or vein identity is regulated by variety of genetic factors that work in concert to specify endothelial cell fate, giving rise to 2 distinct components of the circulatory loop possessing unique structural characteristics. Work in multiple in vivo animal model systems has led to a detailed examination of the interacting partners that determine arterial and venous specification. We discuss the hierarchical arrangement of many signaling molecules, including Hedgehog (Hh), vascular endothelial growth factor (VEGF), Notch, and chicken ovalbumin upstream-transcription factor II (COUP-TFII) that promote or inhibit divergent pathways of endothelial cell fate. Elucidation of the functional role of these genetic determinants of blood vessel specification together with the epigenetic factors involved in subsequent modification of arterial-venous identity will allow for potential new therapeutic targets for vascular disorders.


Assuntos
Artérias/embriologia , Padronização Corporal , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Veias/embriologia , Animais , Artérias/enzimologia , Artérias/metabolismo , Padronização Corporal/genética , Fator II de Transcrição COUP/metabolismo , Diferenciação Celular , Células Endoteliais/enzimologia , Efrina-B2/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neovascularização Fisiológica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor EphB4/metabolismo , Receptores Notch/metabolismo , Fatores de Transcrição SOX/metabolismo , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Veias/enzimologia , Veias/metabolismo
8.
J Biol Chem ; 283(42): 28329-37, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18669637

RESUMO

Fibroblast growth factor-binding protein 1 (FGF-BP1 is BP1) is involved in the regulation of embryonic development, tumor growth, and angiogenesis by mobilizing endogenous FGFs from their extracellular matrix storage. Here we describe a new member of the FGF-BP family, human BP3. We show that the hBP3 protein is secreted from cells, binds to FGF2 in vitro and in intact cells, and inhibits FGF2 binding to heparin. To determine the function of hBP3 in vivo, hBP3 was transiently expressed in chicken embryos and resulted in > 50% lethality within 24 h because of vascular leakage. The onset of vascular permeability was monitored by recording the extravasation kinetics of FITC-labeled 40-kDa dextran microperfused into the vitelline vein of 3-day-old embryos. Vascular permeability increased as early as 8 h after expression of hBP3. The increased vascular permeability caused by hBP3 was prevented by treatment of embryos with PD173074, a selective FGFR kinase inhibitor. Interestingly, a C-terminal 66-amino acid fragment (C66) of hBP3, which contains the predicted FGF binding domain, still inhibited binding of FGF2 to heparin similar to full-length hBP3. However, expression of the C66 fragment did not increase vascular permeability on its own, but required the administration of exogenous FGF2 protein. We conclude that the FGF binding domain and the heparin binding domain are necessary for the hBP3 interaction with endogenous FGF and the activation of FGFR signaling in vivo.


Assuntos
Proteínas de Transporte/metabolismo , Sequência de Aminoácidos , Animais , Permeabilidade Capilar , Embrião de Galinha , Galinhas , Cromatografia Líquida/métodos , Humanos , Cinética , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA , Homologia de Sequência de Aminoácidos , Transdução de Sinais
9.
Cancer Res ; 66(2): 1191-8, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424058

RESUMO

The activity of growth factors is crucial for tumor progression. We previously characterized a secreted fibroblast growth factor-binding protein (FGF-BP1) as a chaperone molecule, which enhances the biological functions of FGFs by releasing FGFs from the extracellular matrix. Here, we characterize the frequency and pattern of FGF-BP1 expression during the malignant progression of pancreas and colorectal carcinoma. For this, we generated monoclonal antibodies that detect FGF-BP1 protein in formalin-fixed, paraffin-embedded tissues and applied in situ hybridization to detect FGF-BP1 mRNA in adjacent tissue sections. FGF-BP1 protein and mRNA were found up-regulated (>70% positive) in parallel (r = 0.70, P < 0.0001) in colon adenoma (n = 9) as well as primary (n = 46) and metastatic (n = 71) colorectal cancers relative to normal colon epithelia (all P < 0.0001, versus normal). Similarly, pancreatitis (n = 17), pancreatic intraepithelial neoplasia (n = 80), and pancreatic adenocarcinoma (n = 67) showed a significant up-regulation of FGF-BP1 compared with normal pancreas (n = 42; all P < 0.0001, relative to normal). Furthermore, the biological activity of FGF-BP1 is neutralized by one of the antibodies, suggesting the potential for antibody-based therapeutic targeting. We propose that the up-regulation of the secreted FGF-BP1 protein during initiation of pancreas and colon neoplasia could make this protein a possible serum marker indicating the presence of high-risk premalignant lesions.


Assuntos
Adenocarcinoma/genética , Proteínas de Transporte/biossíntese , Neoplasias do Colo/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/patologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Proteínas de Transporte/fisiologia , Transformação Celular Neoplásica , Neoplasias do Colo/patologia , Progressão da Doença , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Neoplasias Pancreáticas/patologia , Pancreatite/genética , Pancreatite/patologia , Pancrelipase/fisiologia , RNA Mensageiro/biossíntese , Medição de Risco , Células Tumorais Cultivadas , Regulação para Cima
10.
J Biol Chem ; 281(2): 1137-44, 2006 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-16257968

RESUMO

Fibroblast growth factor-binding proteins (FGF-BP) are secreted carrier proteins that release fibroblast growth factors (FGFs) from the extracellular matrix storage and thus enhance FGF activity. Here we have mapped the interaction domain between human FGF-BP1 and FGF-2. For this, we generated T7 phage display libraries of N-terminally and C-terminally truncated FGF-BP1 fragments that were then panned against immobilized FGF-2. From this panning, a C-terminal fragment of FGF-BP1 (amino acids 193-234) was identified as the minimum binding domain for FGF. As a recombinant protein, this C-terminal fragment binds to FGF-2 and enhances FGF-2-induced signaling in NIH-3T3 fibroblasts and GM7373 endothelial cells, as well as mitogenesis and chemotaxis of NIH-3T3 cells. The FGF interaction domain in FGF-BP1 is distinct from the heparin-binding domain (amino acids 110-143), and homology modeling supports the notion of a distinct domain in the C terminus that is conserved across different species. This domain also contains conserved positioning of cysteine residues with the Cys-214/Cys-222 positions in the human protein predicted to participate in disulfide bridge formation. Phage display of a C214A mutation of FGF-BP1 reduced binding to FGF-2, indicating the functional significance of this disulfide bond. We concluded that the FGF interaction domain is contained in the C terminus of FGF-BP1.


Assuntos
Proteínas de Transporte/química , Fatores de Crescimento de Fibroblastos/química , Sequência de Aminoácidos , Animais , Aorta/metabolismo , Ligação Competitiva , Western Blotting , Bovinos , Proliferação de Células , Células Cultivadas , Quimiotaxia , Cisteína/química , DNA Complementar/metabolismo , Dissulfetos/química , Relação Dose-Resposta a Droga , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Células NIH 3T3 , Fases de Leitura Aberta , Biblioteca de Peptídeos , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Receptores de Fatores de Crescimento de Fibroblastos/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...