Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 28(6): 633-46, 2014 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-24656741

RESUMO

Neuropilin 1 (NRP1) modulates angiogenesis by binding vascular endothelial growth factor (VEGF) and its receptor, VEGFR2. We examined the consequences when VEGFR2 and NRP1 were expressed on the same cell (cis) or on different cells (trans). In cis, VEGF induced rapid VEGFR2/NRP1 complex formation and internalization. In trans, complex formation was delayed and phosphorylation of phospholipase Cγ (PLCγ) and extracellular regulated kinase 2 (ERK2) was prolonged, whereas ERK1 phosphorylation was reduced. Trans complex formation suppressed initiation and vascularization of NRP1-expressing mouse fibrosarcoma and melanoma. Suppression in trans required high-affinity, steady-state binding of VEGF to NRP1, which was dependent on the NRP1 C-terminal domain. Compatible with a trans effect of NRP1, quiescent vasculature in the developing retina showed continuous high NRP1 expression, whereas angiogenic sprouting occurred where NRP1 levels fluctuated between adjacent endothelial cells. Therefore, through communication in trans, NRP1 can modulate VEGFR2 signaling and suppress angiogenesis.


Assuntos
Endocitose/fisiologia , Endotélio Vascular/patologia , Fibrossarcoma/irrigação sanguínea , Melanoma Experimental/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Neuropilina-1/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Comunicação Celular , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Endotélio Vascular/metabolismo , Fibrossarcoma/metabolismo , Fibrossarcoma/prevenção & controle , Imunofluorescência , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação , Estereoisomerismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
PLoS One ; 9(1): e86273, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489709

RESUMO

ENDOGLIN (ENG) is a co-receptor for transforming growth factor-ß (TGF-ß) family members that is highly expressed in endothelial cells and has a critical function in the development of the vascular system. Mutations in Eng are associated with the vascular disease known as hereditary hemorrhagic telangiectasia type l. Using mouse embryonic stem cells we observed that angiogenic factors, including vascular endothelial growth factor (VEGF), induce vasculogenesis in embryoid bodies even when Eng deficient cells or cells depleted of Eng using shRNA are used. However, ENG is required for the stem cell-derived endothelial cells to organize effectively into tubular structures. Consistent with this finding, fetal metatarsals isolated from E17.5 Eng heterozygous mouse embryos showed reduced VEGF-induced vascular network formation. Moreover, shRNA-mediated depletion and pharmacological inhibition of ENG in human umbilical vein cells mitigated VEGF-induced angiogenesis. In summary, we demonstrate that ENG is required for efficient VEGF-induced angiogenesis.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Endoglina , Citometria de Fluxo , Imunofluorescência , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Neovascularização Fisiológica/genética
3.
Angiogenesis ; 16(4): 939-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23892628

RESUMO

GATA-binding protein 2 (GATA2) and LIM domain only 2 (Lmo2) form common transcription complexes during hematopoietic differentiation. Here we show that these two transcription factors also play a key role in endothelial cells (EC) and lymphatic EC (LEC) function. Primary EC and tumor-associated blood vessels expressed GATA2 and Lmo2. VEGF-induced sprouting angiogenesis in both differentiating embryonic stem cells (embryoid bodies) and primary EC increased GATA2 and Lmo2 levels. Conversely, silencing of GATA2 and Lmo2 expression in primary EC inhibited VEGF-induced angiogenic activity, including EC migration and sprouting in vitro, two key steps of angiogenesis in vivo. This inhibition of EC function was associated with downregulated expression of neuropilin-2 (NRP2), a co-receptor of VEGFRs for VEGF, at the protein, mRNA and promoter levels. NRP2 overexpression partially rescued the impaired angiogenic sprouting in the GATA2/Lmo2 knockdown EC, confirming that GATA2 and Lmo2 mediated EC function, at least in part, by directly regulating NRP2 gene expression. Furthermore, it was found that primary LEC expressed GATA2 and Lmo2 as well. Silencing of GATA2 and Lmo2 expression in LEC inhibited VEGF-induced LEC sprouting, also in a NRP2-dependent manner. In conclusion, our results demonstrate that GATA2 and Lmo2 cooperatively regulate VEGF-induced angiogenesis and lymphangiogenesis via NRP2.


Assuntos
Células Endoteliais/fisiologia , Fator de Transcrição GATA2/fisiologia , Proteínas com Domínio LIM/fisiologia , Linfangiogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Neuropilina-2/fisiologia , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Corpos Embrioides , Células Endoteliais/metabolismo , Feminino , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas com Domínio LIM/genética , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neuropilina-2/genética , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
4.
Exp Cell Res ; 319(9): 1264-70, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23454603

RESUMO

Blood vessels are composed of endothelial cells, mural cells (smooth muscle cells and pericytes) and their shared basement membrane. During embryonic development a multitude of signaling components orchestrate the formation of new vessels. The process is highly dependent on correct dosage, spacing and timing of these signaling molecules. As vessels mature some cascades remain active, albeit at very low levels, and may be reactivated upon demand. Members of the Transforming growth factor ß (TGF-ß) protein family are strongly engaged in developmental angiogenesis but are also regulators of vascular integrity in the adult. In humans various genetic alterations within this protein family cause vascular disorders, involving disintegration of vascular integrity. Here we summarize and discuss recent data gathered from conditional and endothelial cell specific genetic loss-of-function of members of the TGF-ß family in the mouse.


Assuntos
Proteínas da Superfamília de TGF-beta/genética , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Deleção de Genes , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Proteínas da Superfamília de TGF-beta/metabolismo , Proteínas da Superfamília de TGF-beta/fisiologia
5.
Biochim Biophys Acta ; 1831(1): 13-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23022664

RESUMO

Autotaxin (ATX) is a secreted lysophospholipase D that generates the multifunctional lipid mediator lysophosphatidic acid (LPA). LPA signals through six distinct G protein-coupled receptors, acting alone or in concert to activate multiple effector pathways. The ATX-LPA signaling axis is implicated in a remarkably wide variety of physiological and pathological processes and plays a vital role in embryonic development. Disruption of the ATX-encoding gene (Enpp2) in mice results in intrauterine death due to vascular defects in the extra-embryonic yolk sac and embryo proper. In addition, Enpp2 (-/-) embryos show impaired neural development. The observed angiogenic defects are attributable, at least in part, to loss of LPA signaling through the Gα(12/13)-linked RhoA-ROCK-actin remodeling pathway. Studies in zebrafish also have uncovered a dual role for ATX in both vascular and neural development; furthermore, they point to a key role for ATX-LPA signaling in the regulation of left-right asymmetry. Here we discuss our present understanding of the role of ATX-LPA signaling in vertebrate development. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Assuntos
Desenvolvimento Embrionário , Diester Fosfórico Hidrolases/metabolismo , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Padronização Corporal , Humanos , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo
6.
J Biol Chem ; 288(1): 510-9, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23150666

RESUMO

Autotaxin (ATX) is a secreted lysophospholipase D that generates the lipid mediator lysophosphatidic acid (LPA), playing a key role in diverse physiological and pathological processes. ATX exists in distinct splice variants, but isoform-specific functions remain elusive. Here we characterize the ATXα isoform, which differs from the canonical form (ATXß) in having a 52-residue polybasic insertion of unknown function in the catalytic domain. We find that the ATXα insertion is susceptible to cleavage by extracellular furin-like endoproteases, but cleaved ATXα remains structurally and functionally intact due to strong interactions within the catalytic domain. Through ELISA and surface plasmon resonance assays, we show that ATXα binds specifically to heparin with high affinity (K(d) ~10(-8) M), whereas ATXß does not; furthermore, heparin moderately enhanced the lysophospholipase D activity of ATXα. We further show that ATXα, but not ATXß, binds abundantly to SKOV3 carcinoma cells. ATXα binding was abolished after treating the cells with heparinase III, but not after chondroitinase treatment. Thus, the ATXα insertion constitutes a cleavable heparin-binding domain that mediates interaction with heparan sulfate proteoglycans, thereby targeting LPA production to the plasma membrane.


Assuntos
Proteoglicanas de Heparan Sulfato/química , Heparina/química , Diester Fosfórico Hidrolases/química , Sequência de Aminoácidos , Membrana Celular/metabolismo , Movimento Celular , Cristalografia por Raios X/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Células HEK293 , Humanos , Cinética , Lipídeos/química , Lisofosfolipídeos/química , Microscopia de Fluorescência/métodos , Dados de Sequência Molecular , Diester Fosfórico Hidrolases/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
7.
J Biol Chem ; 287(22): 18551-61, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22493445

RESUMO

Genetic and molecular studies suggest that activin receptor-like kinase 1 (ALK1), a transforming growth factor ß (TGF-ß) type I receptor, and endoglin, a TGF-ß co-receptor, play an essential role in vascular development and pathological angiogenesis. Several agents that interfere with ALK1 and endoglin function are currently in clinical trials for antiangiogenic activity in cancer therapy. One of these agents, PF-03446962 (anti-hALK1 antibody), shows promising results in the clinic. However, its effects on endothelial cell function and mechanism of action are unclear. Here we demonstrate that anti-hALK1 antibody selectively recognizes human ALK1. The anti-hALK1 antibody interfered with bone morphogenetic protein 9 (BMP9)-induced signaling in endothelial cells. Consistent with this notion, anti-hALK1 antibody was found to compete highly efficiently with the binding of the ALK1 ligand BMP9 and TGF-ß to ALK1. Moreover, it prevented BMP9-dependent recruitment of co-receptor endoglin into this angiogenesis-mediating signaling complex. In addition, we demonstrated that anti-hALK1 antibody inhibited endothelial cell sprouting but did not directly interfere with vascular endothelial growth factor (VEGF) signaling, VEGF-induced proliferation, and migration of endothelial cells. Finally, we demonstrated that BMP9 in serum is essential for endothelial sprouting and that anti-hALK1 antibody inhibits this potently. Our data suggest that both the VEGF/VEGF receptor and the BMP9/ALK1 pathways are essential for stimulating angiogenesis, and targeting both pathways simultaneously may be an attractive strategy to overcome resistance to antiangiogenesis therapy.


Assuntos
Receptores de Activinas Tipo II/imunologia , Endotélio Vascular/metabolismo , Fator 2 de Diferenciação de Crescimento/fisiologia , Transdução de Sinais/fisiologia , Receptores de Activinas Tipo II/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Citometria de Fluxo , Fator 2 de Diferenciação de Crescimento/metabolismo , Humanos , Reação em Cadeia da Polimerase , Ligação Proteica
8.
Cell Tissue Res ; 347(1): 177-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21866313

RESUMO

Recent evidence has demonstrated that endothelial cells can have a remarkable plasticity. By a process called Endothelial-to-Mesenchymal Transition (EndMT) endothelial cells convert to a more mesenchymal cell type that can give rise to cells such as fibroblasts, but also bone cells. EndMT is essential during embryonic development and tissue regeneration. Interestingly, it also plays a role in pathological conditions like fibrosis of organs such as the heart and kidney. In addition, EndMT contributes to the generation of cancer associated fibroblasts that are known to influence the tumor-microenvironment favorable for the tumor cells. EndMT is a form of the more widely known and studied Epithelial-to-Mesenchymal Transition (EMT). Like EMT, EndMT can be induced by transforming growth factor (TGF)-ß. Indeed many studies have pointed to the important role of TGF-ß receptor/Smad signaling and downstream targets, such as Snail transcriptional repressor in EndMT. By selective targeting of TGF-ß receptor signaling pathological EndMT may be inhibited for the therapeutic benefit of patients with cancer and fibrosis.


Assuntos
Células Endoteliais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Transdiferenciação Celular/fisiologia , Células Endoteliais/citologia , Fibrose/metabolismo , Fibrose/patologia , Humanos , Mesoderma/citologia , Mesoderma/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/fisiologia
9.
Curr Pharm Biotechnol ; 12(12): 2108-20, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21619534

RESUMO

Angiogenesis, the formation of new blood vessels is essential for diverse physiological processes such as development, but also for pathological conditions like tumor growth. Most studied in this context are tyrosine kinase signaling pathways such as those involving vascular endothelial growth factor (VEGF). There is however accumulating evidence that more pathways are as essential for angiogenesis. Knockout studies of factors in transforming growth factor ß (TGF-ß) signaling have for example showed that also this pathway is indispensable for angiogenesis. This review highlights our understanding of TGF-ß signaling in vascular development and angiogenesis. In particular, we focus on recent insights into the role of the TGF-ß type I receptor ALK1 and co-receptor endoglin in tumor angiogenesis, which provide opportunities for the development of new anti-angiogenesis therapies for treatment of cancer patients.


Assuntos
Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Endotélio Vascular/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
10.
J Lipid Res ; 52(6): 1247-1255, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21421848

RESUMO

Autotaxin (ATX) is a secreted lysophospholipase D that generates the lipid mediator lysophosphatidic acid (LPA). ATX is secreted by adipose tissue and its expression is enhanced in obese/insulin-resistant individuals. Here, we analyzed the specific contribution of adipose-ATX to fat expansion associated with nutritional obesity and its consequences on plasma LPA levels. We established ATX(F/F)/aP2-Cre (FATX-KO) transgenic mice carrying a null ATX allele specifically in adipose tissue. FATX-KO mice and their control littermates were fed either a normal or a high-fat diet (HFD) (45% fat) for 13 weeks. FATX-KO mice showed a strong decrease (up to 90%) in ATX expression in white and brown adipose tissue, but not in other ATX-expressing organs. This was associated with a 38% reduction in plasma LPA levels. When fed an HFD, FATX-KO mice showed a higher fat mass and a higher adipocyte size than control mice although food intake was unchanged. This was associated with increased expression of peroxisome proliferator-activated receptor (PPAR)γ2 and of PPAR-sensitive genes (aP2, adiponectin, leptin, glut-1) in subcutaneous white adipose tissue, as well as in an increased tolerance to glucose. These results show that adipose-ATX is a negative regulator of fat mass expansion in response to an HFD and contributes to plasma LPA levels.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Adiposidade , Modelos Animais de Doenças , Lisofosfolipídeos , Complexos Multienzimáticos , Obesidade/metabolismo , PPAR gama/metabolismo , Fosfodiesterase I , Pirofosfatases , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo Marrom/fisiopatologia , Tecido Adiposo Branco/fisiopatologia , Animais , Glicemia/análise , Tamanho Celular , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/metabolismo , Feminino , Efeito Fundador , Deleção de Genes , Teste de Tolerância a Glucose , Insulina/sangue , Lisofosfolipídeos/sangue , Masculino , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/deficiência , Complexos Multienzimáticos/genética , Obesidade/genética , Obesidade/fisiopatologia , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Fosfodiesterase I/deficiência , Fosfodiesterase I/genética , Diester Fosfórico Hidrolases , Pirofosfatases/deficiência , Pirofosfatases/genética
11.
Nat Struct Mol Biol ; 18(2): 198-204, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21240271

RESUMO

Autotaxin (ATX, also known as ectonucleotide pyrophosphatase/phosphodiesterase-2, ENPP2) is a secreted lysophospholipase D that generates the lipid mediator lysophosphatidic acid (LPA), a mitogen and chemoattractant for many cell types. ATX-LPA signaling is involved in various pathologies including tumor progression and inflammation. However, the molecular basis of substrate recognition and catalysis by ATX and the mechanism by which it interacts with target cells are unclear. Here, we present the crystal structure of ATX, alone and in complex with a small-molecule inhibitor. We have identified a hydrophobic lipid-binding pocket and mapped key residues for catalysis and selection between nucleotide and phospholipid substrates. We have shown that ATX interacts with cell-surface integrins through its N-terminal somatomedin B-like domains, using an atypical mechanism. Our results define determinants of substrate discrimination by the ENPP family, suggest how ATX promotes localized LPA signaling and suggest new approaches for targeting ATX with small-molecule therapeutic agents.


Assuntos
Integrinas/metabolismo , Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/química , Pirofosfatases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Domínio Catalítico , Linhagem Celular , Cristalografia por Raios X , Humanos , Lisofosfolipídeos/metabolismo , Dados de Sequência Molecular , Mutação , Diester Fosfórico Hidrolases/genética , Ligação Proteica , Estrutura Terciária de Proteína , Pirofosfatases/genética , Ratos , Especificidade por Substrato
12.
Artigo em Inglês | MEDLINE | ID: mdl-20823545

RESUMO

Autotaxin (ATX or ENPP2) is a secreted glycosylated mammalian enzyme that exhibits lysophospholipase D activity, hydrolyzing lysophosphatidylcholine to the signalling lipid lysophosphatidic acid. ATX is an approximately 100 kDa multi-domain protein encompassing two N-terminal somatomedin B-like domains, a central catalytic phosphodiesterase domain and a C-terminal nuclease-like domain. Protocols for the efficient expression of ATX from stably transfected mammalian HEK293 cells in amounts sufficient for crystallographic studies are reported. Purification resulted in protein that crystallized readily, but various attempts to grow crystals suitable in size for routine crystallographic structure determination were not successful. However, the available micrometre-thick plates diffracted X-rays beyond 2.0 A resolution and allowed the collection of complete diffraction data to about 2.6 A resolution. The problems encountered and the current advantages and limitations of diffraction data collection from thin crystal plates are discussed.


Assuntos
Glicoproteínas/química , Diester Fosfórico Hidrolases/química , Pirofosfatases/química , Animais , Cristalização , Cristalografia por Raios X , Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/isolamento & purificação , Células HEK293 , Humanos , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/isolamento & purificação , Pirofosfatases/genética , Pirofosfatases/isolamento & purificação , Ratos
13.
Mol Cancer Res ; 8(8): 1063-73, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20651020

RESUMO

Neuropilins (NRP1 and NRP2) are coreceptors for vascular endothelial growth factor (VEGF) and mediate angiogenesis and tumor progression. VEGF binds to the NRP1 and NRP2 B domains. Previously, it was shown that mutagenesis of the soluble NRP2 B domain (MutB-NRP2) increased affinity to VEGF by 8-fold. Here, we show that MutB-NRP2 inhibited (125)I-VEGF binding to NRP1, NRP2, and VEGFR-2. It antagonized VEGF-induced VEGFR-2/NRP2 complex formation and inhibited VEGF-induced activation of AKT, a mediator of cell survival, without affecting activation of VEGFR-2. In three-dimensional embryoid bodies, a model of VEGF-induced angiogenesis, MutB-NRP2 inhibited VEGF-induced sprouting. When overexpressed in human melanoma cells, MutB-NRP2 inhibited tumor growth compared with control tumors. Avastin (bevacizumab), a monoclonal antibody to VEGF, inhibited VEGF interactions with VEGFR-2, but not with NRPs. The combination of MutB-NRP2 and Avastin resulted in an enhanced inhibition of human melanoma tumor growth compared with MutB-NRP2 treatment only or Avastin treatment only. In conclusion, these results indicate that MutB-NRP2 is a novel antagonist of VEGF bioactivity and tumor progression.


Assuntos
Melanoma/prevenção & controle , Mutação/genética , Neuropilina-2/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Bevacizumab , Células Cultivadas , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Imunofluorescência , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Nus , Neuropilina-1/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Suínos , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
J Med Chem ; 53(13): 4958-67, 2010 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-20536182

RESUMO

Autotaxin (ATX) is an extracellular enzyme that hydrolyzes lysophosphatidylcholine (LPC) to produce the lipid mediator lysophosphatidic acid (LPA). The ATX-LPA signaling axis has been implicated in diverse physiological and pathological processes, including vascular development, inflammation, fibrotic disease, and tumor progression. Therefore, targeting ATX with small molecule inhibitors is an attractive therapeutic strategy. We recently reported that 2,4-thiazolidinediones inhibit ATX activity in the micromolar range. Interestingly, inhibitory potency was dramatically increased by introduction of a boronic acid moiety, designed to target the active site threonine in ATX. Here we report on the discovery and further optimization of boronic acid based ATX inhibitors. The most potent of these compounds inhibits ATX-mediated LPC hydrolysis in the nanomolar range (IC(50) = 6 nM). The finding that ATX can be targeted by boronic acids may aid the development of ATX inhibitors for therapeutic use.


Assuntos
Ácidos Borônicos/síntese química , Ácidos Borônicos/farmacologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Fosfodiesterase I/antagonistas & inibidores , Pirofosfatases/antagonistas & inibidores , Ácidos Borônicos/química , Ácidos Borônicos/metabolismo , Linhagem Celular , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Concentração Inibidora 50 , Lisofosfatidilcolinas/metabolismo , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Complexos Multienzimáticos/metabolismo , Fosfodiesterase I/metabolismo , Diester Fosfórico Hidrolases , Pirofosfatases/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Tiazolidinedionas/síntese química , Tiazolidinedionas/química , Tiazolidinedionas/metabolismo , Tiazolidinedionas/farmacologia
15.
Proc Natl Acad Sci U S A ; 107(16): 7257-62, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20360563

RESUMO

Autotaxin (ATX) is a secreted nucleotide pyrophosphatase/phosphodiesterase that functions as a lysophospholipase D to produce the lipid mediator lysophosphatidic acid (LPA), a mitogen, chemoattractant, and survival factor for many cell types. The ATX-LPA signaling axis has been implicated in angiogenesis, chronic inflammation, fibrotic diseases and tumor progression, making this system an attractive target for therapy. However, potent and selective nonlipid inhibitors of ATX are currently not available. By screening a chemical library, we have identified thiazolidinediones that selectively inhibit ATX-mediated LPA production both in vitro and in vivo. Inhibitor potency was approximately 100-fold increased (IC(50) approximately 30 nM) after the incorporation of a boronic acid moiety, designed to target the active-site threonine (T210) in ATX. Intravenous injection of this inhibitor into mice resulted in a surprisingly rapid decrease in plasma LPA levels, indicating that turnover of LPA in the circulation is much more dynamic than previously appreciated. Thus, boronic acid-based small molecules hold promise as candidate drugs to target ATX.


Assuntos
Ácidos Borônicos/metabolismo , Lisofosfolipídeos/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Glicoproteínas/química , Humanos , Concentração Inibidora 50 , Lipídeos/química , Masculino , Camundongos , Complexos Multienzimáticos/metabolismo , Fosfodiesterase I/metabolismo , Diester Fosfórico Hidrolases/química , Pirofosfatases/metabolismo , Transdução de Sinais , Tiazolidinedionas/química
16.
Nature ; 464(7290): 917-21, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20228789

RESUMO

The vascular endothelial growth factors (VEGFs) are major angiogenic regulators and are involved in several aspects of endothelial cell physiology. However, the detailed role of VEGF-B in blood vessel function has remained unclear. Here we show that VEGF-B has an unexpected role in endothelial targeting of lipids to peripheral tissues. Dietary lipids present in circulation have to be transported through the vascular endothelium to be metabolized by tissue cells, a mechanism that is poorly understood. Bioinformatic analysis showed that Vegfb was tightly co-expressed with nuclear-encoded mitochondrial genes across a large variety of physiological conditions in mice, pointing to a role for VEGF-B in metabolism. VEGF-B specifically controlled endothelial uptake of fatty acids via transcriptional regulation of vascular fatty acid transport proteins. As a consequence, Vegfb(-/-) mice showed less uptake and accumulation of lipids in muscle, heart and brown adipose tissue, and instead shunted lipids to white adipose tissue. This regulation was mediated by VEGF receptor 1 and neuropilin 1 expressed by the endothelium. The co-expression of VEGF-B and mitochondrial proteins introduces a novel regulatory mechanism, whereby endothelial lipid uptake and mitochondrial lipid use are tightly coordinated. The involvement of VEGF-B in lipid uptake may open up the possibility for novel strategies to modulate pathological lipid accumulation in diabetes, obesity and cardiovascular diseases.


Assuntos
Endotélio/metabolismo , Ácidos Graxos/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Núcleo Celular/genética , Células Cultivadas , Endotélio/citologia , Proteínas de Transporte de Ácido Graxo/genética , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Músculos/metabolismo , Miocárdio/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Transdução de Sinais , Transcrição Gênica , Fator B de Crescimento do Endotélio Vascular/deficiência , Fator B de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
J Biol Chem ; 284(11): 7385-94, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19139100

RESUMO

The lipid mediator lysophosphatidic acid (LPA) is a potent regulator of vascular cell function in vitro, but its physiologic role in the cardiovasculature is largely unexplored. To address the role of LPA in regulating platelet function and thrombosis, we investigated the effects of LPA on isolated murine platelets. Although LPA activates platelets from the majority of human donors, we found that treatment of isolated murine platelets with physiologic concentrations of LPA attenuated agonist-induced aggregation. Transgenic overexpression of autotaxin/lysophospholipase D (Enpp2), the enzyme necessary for production of the bulk of biologically active LPA in plasma, elevated circulating LPA levels and induced a bleeding diathesis and attenuation of thrombosis in mice. Intravascular administration of exogenous LPA recapitulated the prolonged bleeding time observed in Enpp2-Tg mice. Enpp2+/- mice, which have approximately 50% normal plasma LPA levels, were more prone to thrombosis. Plasma autotaxin associated with platelets during aggregation and concentrated in arterial thrombus, and activated but not resting platelets bound recombinant autotaxin/lysoPLD in an integrin-dependent manner. These results identify a novel pathway in which LPA production by autotaxin/lysoPLD regulates murine hemostasis and thrombosis and suggest that binding of autotaxin/lysoPLD to activated platelets may provide a mechanism to localize LPA production.


Assuntos
Hemostasia , Lisofosfolipídeos/sangue , Complexos Multienzimáticos/sangue , Fosfodiesterase I/sangue , Diester Fosfórico Hidrolases/sangue , Pirofosfatases/sangue , Trombose/sangue , Animais , Plaquetas/metabolismo , Hemorragia/sangue , Hemorragia/genética , Humanos , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Transgênicos , Complexos Multienzimáticos/genética , Fosfodiesterase I/genética , Diester Fosfórico Hidrolases/genética , Agregação Plaquetária/genética , Pirofosfatases/genética , Trombose/genética
18.
Mol Cancer Res ; 6(9): 1452-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18723828

RESUMO

Lysophosphatidic acid (LPA) is a lipid mediator of a large number of biological processes, including wound healing, brain development, vascular remodeling, and tumor progression. Its role in tumor progression is probably linked to its ability to induce cell proliferation, migration, and survival. In particular, the ascites of ovarian cancers is rich in LPA and has been implicated in growth and invasion of ovarian tumor cells. LPA binds to specific G protein-coupled receptors and thereby activates multiple signal transduction pathways, including those initiated by the small GTPases Ras, Rho, and Rac. We report here a genetic screen with retroviral cDNA expression libraries to identify genes that allow bypass of the p53-dependent replicative senescence response in mouse neuronal cells, conditionally immortalized by a temperature-sensitive mutant of SV40 large T antigen. Using this approach, we identified the LPA receptor type 2 (LPA(2)) and the Rho-specific guanine nucleotide exchange factor Dbs as potent inducers of senescence bypass. Enhanced expression of LPA(2) or Dbs also results in senescence bypass in primary mouse embryo fibroblasts in the presence of wild-type p53, in a Rho GTPase-dependent manner. Our results reveal a novel and unexpected link between LPA signaling and the p53 tumor-suppressive pathway.


Assuntos
Senescência Celular/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/fisiologia , Animais , Antígenos Transformantes de Poliomavirus , Western Blotting , Encéfalo/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Biblioteca Gênica , Proteínas de Fluorescência Verde , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Músculo Estriado/citologia , Músculo Estriado/metabolismo , Policitemia Vera/metabolismo , Policitemia Vera/patologia , Receptores de Ácidos Lisofosfatídicos/genética , Retroviridae/genética , Fatores de Troca de Nucleotídeo Guanina Rho , Proteínas rho de Ligação ao GTP/metabolismo
19.
Blood ; 112(9): 3638-49, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18664627

RESUMO

Vascular endothelial growth factor (VEGF)-A regulates vascular development and angiogenesis. VEGF isoforms differ in ability to bind coreceptors heparan sulfate (HS) and neuropilin-1 (NRP1). We used VEGF-A165 (which binds HS and NRP1), VEGF-A121 (binds neither HS nor NRP1), and parapoxvirus VEGF-E-NZ2 (binds NRP1 but not HS) to investigate the role of NRP1 in organization of endothelial cells into vascular structures. All 3 ligands induced similar level of VEGFR-2 tyrosine phosphorylation in the presence of NRP1. In contrast, sprouting angiogenesis in differentiating embryonic stem cells (embryoid bodies), formation of branching pericyte-embedded vessels in subcutaneous matrigel plugs, and sprouting of intersegmental vessels in developing zebrafish were induced by VEGF-A165 and VEGF-E-NZ2 but not by VEGF-A121. Analyses of recombinant factors with NRP1-binding gain- and loss-of-function properties supported the conclusion that NRP1 is critical for VEGF-induced sprouting and branching of endothelial cells. Signal transduction antibody arrays implicated NRP1 in VEGF-induced activation of p38MAPK. Inclusion of the p38MAPK inhibitor SB203580 in VEGF-A165-containing matrigel plugs led to attenuated angiogenesis and poor association with pericytes. Our data strongly indicate that the ability of VEGF ligands to bind NRP1 influences p38MAPK activation, and formation of functional, pericyte-associated vessels.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Animais Geneticamente Modificados , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Ligantes , Camundongos , Modelos Biológicos , Neovascularização Fisiológica , Neuropilina-1/genética , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Suínos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
20.
Cancer Lett ; 266(2): 203-8, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18378389

RESUMO

FTY720 is an immunomodulator that is phosphorylated in vivo and inhibits lymphocyte mobilization by targeting sphingosine 1-phospate receptors. At doses higher than required for immunomodulation, FTY720 inhibits tumor progression through an unknown mechanism. Here we show that FTY720-phosphate is a competitive inhibitor (Ki approximately 0.2microM) of autotaxin (ATX or NPP2), a nucleotide phosphodiesterase/pyrophosphatase (NPP) that enhances metastasis and angiogenesis and acts as a lysophospholipase D to produce the lipid mediator lysophosphatidic acid (LPA). FTY720-phosphate did no affect the activity of NPP1, the closest relative of ATX. After oral administration in mice, FTY720 (3mg/kg) significantly reduced plasma LPA levels. These results suggest that FTY720 may exert its anticancer effects, at least in part, by targeting the ATX-LPA axis.


Assuntos
Antineoplásicos/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Organofosfatos/farmacologia , Fosfodiesterase I/antagonistas & inibidores , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/antagonistas & inibidores , Esfingosina/análogos & derivados , Animais , Antineoplásicos/química , Feminino , Humanos , Lisofosfolipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multienzimáticos/metabolismo , Metástase Neoplásica , Neovascularização Patológica , Organofosfatos/química , Fosfodiesterase I/metabolismo , Inibidores de Fosfodiesterase/química , Pirofosfatases/metabolismo , Esfingosina/química , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...