Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO Mol Med ; 15(5): e16645, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36912000

RESUMO

Sphingosine-1-phosphate (S1P), the circulating HDL-bound lipid mediator that acts via S1P receptors (S1PR), is required for normal vascular development. The role of this signaling axis in vascular retinopathies is unclear. Here, we show in a mouse model of oxygen-induced retinopathy (OIR) that endothelial overexpression of S1pr1 suppresses while endothelial knockout of S1pr1 worsens neovascular tuft formation. Furthermore, neovascular tufts are increased in Apom-/- mice which lack HDL-bound S1P while they are suppressed in ApomTG mice which have more circulating HDL-S1P. These results suggest that circulating HDL-S1P activation of endothelial S1PR1 suppresses neovascular pathology in OIR. Additionally, systemic administration of ApoM-Fc-bound S1P or a small-molecule Gi-biased S1PR1 agonist suppressed neovascular tuft formation. Circulating HDL-S1P activation of endothelial S1PR1 may be a key protective mechanism to guard against neovascular retinopathies that occur not only in premature infants but also in diabetic patients and aging people.


Assuntos
Neovascularização Retiniana , Camundongos , Animais , Receptores de Esfingosina-1-Fosfato , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/agonistas , Lipoproteínas HDL , Esfingosina , Lisofosfolipídeos
2.
Elife ; 112022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36197001

RESUMO

Serine palmitoyl transferase (SPT), the rate-limiting enzyme in the de novo synthesis of sphingolipids (SL), is needed for embryonic development, physiological homeostasis, and response to stress. The functions of de novo SL synthesis in vascular endothelial cells (EC), which line the entire circulatory system, are not well understood. Here, we show that the de novo SL synthesis in EC not only regulates vascular development but also maintains circulatory and peripheral organ SL levels. Mice with an endothelial-specific gene knockout of SPTLC1 (Sptlc1 ECKO), an essential subunit of the SPT complex, exhibited reduced EC proliferation and tip/stalk cell differentiation, resulting in delayed retinal vascular development. In addition, Sptlc1 ECKO mice had reduced retinal neovascularization in the oxygen-induced retinopathy model. Mechanistic studies suggest that EC SL produced from the de novo pathway are needed for lipid raft formation and efficient VEGF signaling. Post-natal deletion of the EC Sptlc1 also showed rapid reduction of several SL metabolites in plasma, red blood cells, and peripheral organs (lung and liver) but not in the retina, part of the central nervous system (CNS). In the liver, EC de novo SL synthesis was important for acetaminophen-induced rapid ceramide elevation and hepatotoxicity. These results suggest that EC-derived SL metabolites are in constant flux between the vasculature, circulatory elements, and parenchymal cells of non-CNS organs. Taken together, our data point to the central role of the endothelial SL biosynthesis in maintaining vascular development, neovascular proliferation, non-CNS tissue metabolic homeostasis, and hepatocyte response to stress.


Assuntos
Serina C-Palmitoiltransferase , Esfingolipídeos , Animais , Camundongos , Acetaminofen , Ceramidas , Células Endoteliais/metabolismo , Homeostase , Oxigênio , Serina , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/metabolismo , Fator A de Crescimento do Endotélio Vascular
3.
Cell Rep ; 40(3): 111114, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858557

RESUMO

Hematopoietic stem cell (HSC) generation in the aorta-gonad-mesonephros region requires HSC specification signals from the surrounding microenvironment. In zebrafish, PDGF-B/PDGFRß signaling controls hematopoietic stem/progenitor cell (HSPC) generation and is required in the HSC specification niche. Little is known about murine HSPC specification in vivo and whether PDGF-B/PDGFRß is involved. Here, we show that PDGFRß is expressed in distinct perivascular stromal cell layers surrounding the mid-gestation dorsal aorta, and its deletion impairs hematopoiesis. We demonstrate that PDGFRß+ cells play a dual role in murine hematopoiesis. They act in the aortic niche to support HSPCs, and in addition, PDGFRß+ embryonic precursors give rise to a subset of HSPCs that persist into adulthood. These findings provide crucial information for the controlled production of HSPCs in vitro.


Assuntos
Mesonefro , Peixe-Zebra , Animais , Hematopoese , Células-Tronco Hematopoéticas , Camundongos , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Células Estromais
4.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33375813

RESUMO

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Assuntos
Barreira Hematoencefálica/metabolismo , Pericitos/citologia , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/patologia , Proliferação de Células , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Linfocinas/deficiência , Linfocinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Pericitos/metabolismo , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/deficiência , Fator de Crescimento Derivado de Plaquetas/genética , Análise de Célula Única , Transcriptoma
5.
J Clin Invest ; 130(8): 4055-4068, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32369453

RESUMO

Fowler syndrome is a rare autosomal recessive brain vascular disorder caused by mutation in FLVCR2 in humans. The disease occurs during a critical period of brain vascular development, is characterized by glomeruloid vasculopathy and hydrocephalus, and is almost invariably prenatally fatal. Here, we sought to gain insights into the process of brain vascularization and the pathogenesis of Fowler syndrome by inactivating Flvcr2 in mice. We showed that Flvcr2 was necessary for angiogenic sprouting in the brain, but surprisingly dispensable for maintaining the blood-brain barrier. Endothelial cells lacking Flvcr2 had altered expression of angiogenic factors, failed to adopt tip cell properties, and displayed reduced sprouting, leading to vascular malformations similar to those seen in humans with Fowler syndrome. Brain hypovascularization was associated with hypoxia and tissue infarction, ultimately causing hydrocephalus and death of mutant animals. Strikingly, despite severe vascular anomalies and brain tissue infarction, the blood-brain barrier was maintained in Flvcr2 mutant mice. Our Fowler syndrome model therefore defined the pathobiology of this disease and provided new insights into brain angiogenesis by showing uncoupling of vessel morphogenesis and blood-brain barrier formation.


Assuntos
Barreira Hematoencefálica , Malformações Vasculares do Sistema Nervoso Central , Células Endoteliais , Proteínas de Membrana Transportadoras/deficiência , Neovascularização Fisiológica , Animais , Barreira Hematoencefálica/embriologia , Barreira Hematoencefálica/patologia , Malformações Vasculares do Sistema Nervoso Central/embriologia , Malformações Vasculares do Sistema Nervoso Central/genética , Malformações Vasculares do Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Knockout
6.
Dev Cell ; 52(6): 779-793.e7, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32059774

RESUMO

Transcriptional mechanisms that drive angiogenesis and organotypic vascular endothelial cell specialization are poorly understood. Here, we show that retinal endothelial sphingosine 1-phosphate receptors (S1PRs), which restrain vascular endothelial growth factor (VEGF)-induced angiogenesis, spatially restrict expression of JunB, a member of the activator protein 1 (AP-1) family of transcription factors (TFs). Mechanistically, VEGF induces JunB expression at the sprouting vascular front while S1PR-dependent vascular endothelial (VE)-cadherin assembly suppresses JunB expression in the nascent vascular network, thus creating a gradient of this TF. Endothelial-specific JunB knockout mice showed diminished expression of neurovascular guidance genes and attenuated retinal vascular network progression. In addition, endothelial S1PR signaling is required for normal expression of ß-catenin-dependent genes such as TCF/LEF1 and ZIC3 TFs, transporters, and junctional proteins. These results show that S1PR signaling restricts JunB function to the expanding vascular front, thus creating an AP-1 gradient and enabling organotypic endothelial cell specialization of the vascular network.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Células Cultivadas , Montagem e Desmontagem da Cromatina , Células Endoteliais/citologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vasos Retinianos/citologia , Vasos Retinianos/embriologia , Fator de Transcrição AP-1/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
7.
J Cereb Blood Flow Metab ; 38(3): 456-468, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28276839

RESUMO

The establishment of a fully functional blood vascular system requires elaborate angiogenic and vascular maturation events in order to fulfill organ-specific anatomical and physiological needs. Although vascular mural cells, i.e. pericytes and vascular smooth muscle cells, are known to play fundamental roles during these processes, their characteristics during vascular development remain incompletely understood. In this report, we utilized transgenic reporter mice in which mural cells are genetically labeled to examine developing vascular mural cells in the central nervous system (CNS). We found platelet-derived growth factor receptor ß gene ( Pdgfrb)-driven EGFP reporter expression as a suitable marker for vascular mural cells at the earliest stages of mouse brain vascularization. Furthermore, the combination of Pdgfrb and NG2 gene (Cspg4) driven reporter expression increased the specificity of brain vascular mural cell labeling at later stages. The expression of other known pericyte markers revealed time-, region- and marker-specific patterns, suggesting heterogeneity in mural cell maturation. We conclude that transgenic reporter mice provide an important tool to explore the development of CNS pericytes in health and disease.


Assuntos
Vasos Sanguíneos/ultraestrutura , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Genes Reporter/genética , Animais , Antígenos/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Encéfalo/ultraestrutura , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/ultraestrutura , Desenvolvimento Embrionário , Feminino , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular , Miócitos de Músculo Liso/ultraestrutura , Pericitos/ultraestrutura , Proteoglicanas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética
8.
JCI Insight ; 1(21): e87058, 2016 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-28018969

RESUMO

Regeneration of hepatic sinusoidal vasculature is essential for non-fibrotic liver regrowth and restoration of its metabolic capacity. However, little is known about how this specialized vascular niche is regenerated. Here we show that activation of endothelial sphingosine-1-phosphate receptor-1 (S1P1) by its natural ligand bound to HDL (HDL-S1P) induces liver regeneration and curtails fibrosis. In mice lacking HDL-S1P, liver regeneration after partial hepatectomy was impeded and associated with aberrant vascular remodeling, thrombosis and peri-sinusoidal fibrosis. Notably, this "maladaptive repair" phenotype was recapitulated in mice that lack S1P1 in the endothelium. Reciprocally, enhanced plasma levels of HDL-S1P or administration of SEW2871, a pharmacological agonist specific for S1P1 enhanced regeneration of metabolically functional vasculature and alleviated fibrosis in mouse chronic injury and cholestasis models. This study shows that natural and pharmacological ligands modulate endothelial S1P1 to stimulate liver regeneration and inhibit fibrosis, suggesting that activation of this pathway may be a novel therapeutic strategy for liver fibrosis.

9.
Sci Rep ; 6: 35108, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27725773

RESUMO

Pericytes, the mural cells of blood microvessels, regulate microvascular development and function and have been implicated in many brain diseases. However, due to a paucity of defining markers, pericyte identification and functional characterization remain ambiguous and data interpretation problematic. In mice carrying two transgenic reporters, Pdgfrb-eGFP and NG2-DsRed, we found that double-positive cells were vascular mural cells, while the single reporters marked additional, but non-overlapping, neuroglial cells. Double-positive cells were isolated by fluorescence-activated cell sorting (FACS) and analyzed by RNA sequencing. To reveal defining patterns of mural cell transcripts, we compared the RNA sequencing data with data from four previously published studies. The meta-analysis provided a conservative catalogue of 260 brain mural cell-enriched gene transcripts. We validated pericyte-specific expression of two novel markers, vitronectin (Vtn) and interferon-induced transmembrane protein 1 (Ifitm1), using fluorescent in situ hybridization and immunohistochemistry. We further analyzed signaling pathways and interaction networks of the pericyte-enriched genes in silico. This work provides novel insight into the molecular composition of brain mural cells. The reported gene catalogue facilitates identification of brain pericytes by providing numerous new candidate marker genes and is a rich source for new hypotheses for future studies of brain mural cell physiology and pathophysiology.


Assuntos
Encéfalo/citologia , Perfilação da Expressão Gênica , Microvasos/citologia , Pericitos/fisiologia , Animais , Citometria de Fluxo , Camundongos , Análise de Sequência de RNA , Coloração e Rotulagem
10.
PLoS One ; 10(9): e0137949, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26394398

RESUMO

Despite its known expression in both the vascular endothelium and the lung epithelium, until recently the physiological role of the adhesion receptor Gpr116/ADGRF5 has remained elusive. We generated a new mouse model of constitutive Gpr116 inactivation, with a large genetic deletion encompassing exon 4 to exon 21 of the Gpr116 gene. This model allowed us to confirm recent results defining Gpr116 as necessary regulator of surfactant homeostasis. The loss of Gpr116 provokes an early accumulation of surfactant in the lungs, followed by a massive infiltration of macrophages, and eventually progresses into an emphysema-like pathology. Further analysis of this knockout model revealed cerebral vascular leakage, beginning at around 1.5 months of age. Additionally, endothelial-specific deletion of Gpr116 resulted in a significant increase of the brain vascular leakage. Mice devoid of Gpr116 developed an anatomically normal and largely functional vascular network, surprisingly exhibited an attenuated pathological retinal vascular response in a model of oxygen-induced retinopathy. These data suggest that Gpr116 modulates endothelial properties, a previously unappreciated function despite the pan-vascular expression of this receptor. Our results support the key pulmonary function of Gpr116 and describe a new role in the central nervous system vasculature.


Assuntos
Células Epiteliais Alveolares/metabolismo , Endotélio Vascular/metabolismo , Surfactantes Pulmonares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Permeabilidade Capilar/genética , Feminino , Expressão Gênica , Homeostase/genética , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Modelos Biológicos , Miocárdio/metabolismo , Miocárdio/patologia , Receptores Acoplados a Proteínas G/genética , Neovascularização Retiniana/genética , Neovascularização Retiniana/metabolismo , Baço/metabolismo , Baço/patologia
11.
Development ; 141(23): 4489-99, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25406396

RESUMO

Vascular development of the central nervous system and blood-brain barrier (BBB) induction are closely linked processes. The role of factors that promote endothelial sprouting and vascular leak, such as vascular endothelial growth factor A, are well described, but the factors that suppress angiogenic sprouting and their impact on the BBB are poorly understood. Here, we show that integrin αVß8 activates angiosuppressive TGFß gradients in the brain, which inhibit endothelial cell sprouting. Loss of αVß8 in the brain or downstream TGFß1-TGFBR2-ALK5-Smad3 signaling in endothelial cells increases vascular sprouting, branching and proliferation, leading to vascular dysplasia and hemorrhage. Importantly, BBB function in Itgb8 mutants is intact during early stages of vascular dysgenesis before hemorrhage. By contrast, Pdgfb(ret/ret) mice, which exhibit severe BBB disruption and vascular leak due to pericyte deficiency, have comparatively normal vascular morphogenesis and do not exhibit brain hemorrhage. Our data therefore suggest that abnormal vascular sprouting and patterning, not BBB dysfunction, underlie developmental cerebral hemorrhage.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/irrigação sanguínea , Hemorragia Cerebral/etiologia , Neovascularização Patológica/complicações , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Encéfalo/metabolismo , Contagem de Células , Células Endoteliais/fisiologia , Imuno-Histoquímica , Integrinas/metabolismo , Camundongos , Microscopia Confocal , Fator de Crescimento Transformador beta/metabolismo
12.
J Lipid Res ; 54(3): 843-851, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23296878

RESUMO

Signaling through sphingosine-1-phosphate receptor1 (S1P1) promotes blood vessel barrier function. Degradation of S1P1 results in increased vascular permeability in the lung and may explain side effects associated with administration of FTY720, a functional antagonist of the S1P1 receptor that is currently used to treat multiple sclerosis. Ulcerative colitis (UC) is characterized by an increased density of abnormal vessels. The expression or role of S1P1 in blood vessels in the colon has not been investigated. In the present study, we show that S1P1 is overexpressed in the colonic mucosa of UC patients. This increase in S1P1 levels reflects increased vascular density in the inflamed mucosa. Genetic deletion of S1pr1 in mice increases colonic vascular permeability under basal conditions and increases bleeding in experimental colitis. In contrast, neither FTY720 nor AUY954, two S1P receptor-targeting agents, increases bleeding in experimental colitis. Taken together, our findings demonstrate that S1P1 is critical to maintaining colonic vascular integrity and may play a role in UC pathogenesis.


Assuntos
Colite Ulcerativa/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Permeabilidade Capilar/efeitos dos fármacos , Colo/efeitos dos fármacos , Cloridrato de Fingolimode , Humanos , Mucosa Intestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Propilenoglicóis/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tiofenos/farmacologia , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
13.
Dev Cell ; 23(3): 600-10, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22975328

RESUMO

During angiogenesis, nascent vascular sprouts fuse to form vascular networks, enabling efficient circulation. Mechanisms that stabilize the vascular plexus are not well understood. Sphingosine 1-phosphate (S1P) is a blood-borne lipid mediator implicated in the regulation of vascular and immune systems. Here we describe a mechanism by which the G protein-coupled S1P receptor-1 (S1P1) stabilizes the primary vascular network. A gradient of S1P1 expression from the mature regions of the vascular network to the growing vascular front was observed. In the absence of endothelial S1P1, adherens junctions are destabilized, barrier function is breached, and flow is perturbed, resulting in abnormal vascular hypersprouting. Interestingly, S1P1 responds to S1P as well as laminar shear stress to transduce flow-mediated signaling in endothelial cells both in vitro and in vivo. These data demonstrate that blood flow and circulating S1P activate endothelial S1P1 to stabilize blood vessels in development and homeostasis.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Animais , Hemorreologia , Homeostase , Camundongos , Receptores de Lisoesfingolipídeo/sangue
14.
J Microbiol Biotechnol ; 17(4): 691-4, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18051286

RESUMO

Acetic acid and butyric acid were produced by the anaerobic fermentation of soil mixed with wheat or rice bran. The concentration of acetic acid produced in the wheat and rice bran-treated soil was 31.2 mM and 8 mM, respectively, whereas the concentration of butyric acid in the wheat and rice bran-treated soil was 25.0 mM and 8 mM, respectively. The minimal fungicidal concentration (MFC) for all the fungal strains was 40-60 mM acetic acid, 20-40 mM butyric acid, and 40-60 mM mixture of acetic acid: butyric acid (1:1, v/v). Consequently, the efficacy of mixing wheat-bran with soil to control soil diseases was demonstrated.


Assuntos
Ácido Acético/metabolismo , Antifúngicos/farmacologia , Ácido Butírico/metabolismo , Fermentação , Microbiologia do Solo , Ácido Acético/farmacologia , Anaerobiose , Ácido Butírico/farmacologia , Fibras na Dieta , Testes de Sensibilidade Microbiana
15.
Int J Cancer ; 120(6): 1277-83, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17187368

RESUMO

In this study, the roles of matrix metalloproteinase (MMP)-2 and MMP-9 in platelet-activating factor (PAF)-induced experimental pulmonary metastasis of the murine melanoma cell, B16F10, were investigated. An injection of PAF resulted in increases in mRNA expression, protein levels and the activities of both MMP-2 and MMP-9 in the lungs. The overall expression of MMP-9 was stronger than that of MMP-2. The increased MMP-9 expression was inhibited by both NF-kappaB and AP-1 inhibitors, whereas the increased MMP-2 expression was inhibited by only AP-1 inhibitors. Immunohistochemical analysis revealed that MMP-9 was expressed in bronchial epithelial cells as well as in the walls of blood vessels, whereas MMP-2 expression was observed only in bronchial epithelial cells. PAF significantly enhanced the pulmonary metastasis of B16F10, which was inhibited by both NF-kappaB and c-jun inhibitors. MMP-9 inhibitor, but not that of MMP-2, completely inhibited PAF-induced B16F10 metastasis. These data indicate that MMP-9, the expression of which was regulated by NF-kappaB and AP-1, plays a critical role in PAF-induced enhancement of pulmonary melanoma metastasis.


Assuntos
Metaloproteinase 9 da Matriz/fisiologia , Metástase Neoplásica , Fator de Ativação de Plaquetas/toxicidade , Animais , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/análise , Inibidores de Metaloproteinases de Matriz , Melanoma Experimental/induzido quimicamente , Melanoma Experimental/enzimologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Fator de Ativação de Plaquetas/farmacologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Fator de Transcrição AP-1/antagonistas & inibidores
16.
FEBS Lett ; 579(11): 2369-75, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15848174

RESUMO

Platelet-activating factor (PAF) augments angiogenesis by promoting the synthesis of various angiogenic factors, via the activation of NF-kappaB. In this study, we investigated the role of the matrix metalloproteinase (MMP)-9, in PAF-induced angiogenesis. PAF increased mRNA expression, protein synthesis, and MMP-9 activity in ECV304 cells, in a NF-kappaB-dependent manner. PAF increased MMP-9 promoter activity in ECV304, which was inhibited by WEB2107, and NF-kappaB inhibitors. Transfected NF-kappaB subunits, p65 or/and p50, increased luciferase activity in the reporter plasmid MMP-9, resulting in an increase not only of MMP-9 luciferase activity, but also of mRNA expression in MMP-9. MMP-9 or NF-kappaB inhibitors significantly inhibited PAF-induced angiogenesis, in a dose-dependent manner, in an in vivo mouse Matrigel implantation model. In a parallel to the Matrigel implantation study, MMP-9 or NF-kappaB inhibitors inhibited PAF-induced sprouting of porcine pulmonary arterial endothelial cells. These data indicate that NF-kappaB-dependent MMP-9 plays a key role in PAF-induced angiogenesis.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Animais , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinase 9 da Matriz/genética , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fator de Ativação de Plaquetas/antagonistas & inibidores , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos
17.
Cancer Res ; 64(18): 6482-8, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15374958

RESUMO

In this study, we investigated the molecular events involved in estrogen-induced angiogenesis. Treatment of the human endometrial adenocarcinoma cells, HEC-1A, with estrogen up-regulated mRNA expression and protein synthesis of various angiogenic factors such as tumor necrosis factor-alpha, interleukin-1, basic fibroblast growth factor, and vascular endothelial growth factor. The estrogen-dependent induction of the expression was blocked by the platelet-activating factor (PAF) antagonists, WEB 2170. Estrogen treatment caused the activation of nuclear factor (NF)-kappaB in HEC-1A cells and was also blocked by PAF antagonist. Inhibitors of NF-kappaB activation inhibited estrogen-induced mRNA expression and protein synthesis of the angiogenic factors. Estrogen led to a pronounced angiogenesis as assessed by a mouse Matrigel model in vivo and endothelial cell sprouting in vitro. PAF antagonists or NF-kappaB inhibitors significantly inhibited this estrogen-dependent angiogenesis. Estrogen caused phospholipase A2 (PLA2) gene and protein expression. Estrogen-induced vascular endothelial growth factor mRNA expression and sprouting were significantly inhibited by PLA2 inhibitors, suggesting PLA2 expression is the upstream pathway in the estrogen-induced angiogenesis. Taken together, these results suggest that estrogen induces the production of angiogenic factors via a mechanism involving PAF-mediated NF-kappaB activation.


Assuntos
Adenocarcinoma/irrigação sanguínea , Neoplasias do Endométrio/irrigação sanguínea , Congêneres do Estradiol/farmacologia , NF-kappa B/fisiologia , Neovascularização Patológica/metabolismo , Fator de Ativação de Plaquetas/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Interleucina-1/biossíntese , Interleucina-1/genética , Interleucina-8/biossíntese , Interleucina-8/genética , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/antagonistas & inibidores , Neovascularização Patológica/genética , Fosfolipases A/biossíntese , Fosfolipases A/genética , Fosfolipases A2 , Fator de Ativação de Plaquetas/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA