Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
Oncogene ; 42(35): 2629-2640, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37500798

RESUMO

Preventing or effectively treating metastatic uveal melanoma (UM) is critical because it occurs in about half of patients and confers a very poor prognosis. There is emerging evidence that hepatocyte growth factor (HGF) and insulin-like growth factor 1 (IGF-1) promote metastasis and contribute to the striking metastatic hepatotropism observed in UM metastasis. However, the molecular mechanisms by which HGF and IGF-1 promote UM liver metastasis have not been elucidated. ASAP1, which acts as an effector for the small GTPase ARF6, is highly expressed in the subset of uveal melanomas most likely to metastasize. Here, we found that HGF and IGF-1 hyperactivate ARF6, leading to its interaction with ASAP1, which then acts as an effector to induce nuclear localization and transcriptional activity of NFAT1. Inhibition of any component of this pathway impairs cellular invasiveness. Additionally, knocking down ASAP1 or inhibiting NFAT signaling reduces metastasis in a xenograft mouse model of UM. The discovery of this signaling pathway represents not only an advancement in our understanding of the biology of uveal melanoma metastasis but also identifies a novel pathway that could be targeted to treat or prevent metastatic uveal melanoma.


Assuntos
Melanoma , Neoplasias Uveais , Humanos , Animais , Camundongos , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Melanoma/patologia , Neoplasias Uveais/metabolismo , Modelos Animais de Doenças , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
3.
Neuron ; 110(19): 3106-3120.e7, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-35961320

RESUMO

Breakdown of the blood-central nervous system barrier (BCNSB) is a hallmark of many neuroinflammatory disorders, such as multiple sclerosis (MS). Using a mouse model of MS, experimental autoimmune encephalomyelitis (EAE), we show that endothelial-to-mesenchymal transition (EndoMT) occurs in the CNS before the onset of clinical symptoms and plays a major role in the breakdown of BCNSB function. EndoMT can be induced by an IL-1ß-stimulated signaling pathway in which activation of the small GTPase ADP ribosylation factor 6 (ARF6) leads to crosstalk with the activin receptor-like kinase (ALK)-SMAD1/5 pathway. Inhibiting the activation of ARF6 both prevents and reverses EndoMT, stabilizes BCNSB function, reduces demyelination, and attenuates symptoms even after the establishment of severe EAE, without immunocompromising the host. Pan-inhibition of ALKs also reduces disease severity in the EAE model. Therefore, multiple components of the IL-1ß-ARF6-ALK-SMAD1/5 pathway could be targeted for the treatment of a variety of neuroinflammatory disorders.


Assuntos
Encefalomielite Autoimune Experimental , Proteínas Monoméricas de Ligação ao GTP , Esclerose Múltipla , Receptores de Ativinas/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Doenças Neuroinflamatórias , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
4.
Cancer Res ; 79(11): 2892-2908, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31048499

RESUMO

Melanoma has an unusual capacity to spread in early-stage disease, prompting aggressive clinical intervention in very thin primary tumors. Despite these proactive efforts, patients with low-risk, low-stage disease can still develop metastasis, indicating the presence of permissive cues for distant spread. Here, we show that constitutive activation of the small GTPase ARF6 (ARF6Q67L) is sufficient to accelerate metastasis in mice with BRAFV600E/Cdkn2aNULL melanoma at a similar incidence and severity to Pten loss, a major driver of PI3K activation and melanoma metastasis. ARF6Q67L promoted spontaneous metastasis from significantly smaller primary tumors than PTENNULL, implying an enhanced ability of ARF6-GTP to drive distant spread. ARF6 activation increased lung colonization from circulating melanoma cells, suggesting that the prometastatic function of ARF6 extends to late steps in metastasis. Unexpectedly, ARF6Q67L tumors showed upregulation of Pik3r1 expression, which encodes the p85 regulatory subunit of PI3K. Tumor cells expressing ARF6Q67L displayed increased PI3K protein levels and activity, enhanced PI3K distribution to cellular protrusions, and increased AKT activation in invadopodia. ARF6 is necessary and sufficient for activation of both PI3K and AKT, and PI3K and AKT are necessary for ARF6-mediated invasion. We provide evidence for aberrant ARF6 activation in human melanoma samples, which is associated with reduced survival. Our work reveals a previously unknown ARF6-PI3K-AKT proinvasive pathway, it demonstrates a critical role for ARF6 in multiple steps of the metastatic cascade, and it illuminates how melanoma cells can acquire an early metastatic phenotype in patients. SIGNIFICANCE: These findings reveal a prometastatic role for ARF6 independent of tumor growth, which may help explain how melanoma spreads distantly from thin, early-stage primary tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2892/F1.large.jpg.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Melanoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Cutâneas/patologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Guanosina Trifosfato/metabolismo , Humanos , Neoplasias Pulmonares/secundário , Melanoma/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos Mutantes , Camundongos SCID , Metástase Neoplásica , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/metabolismo
5.
J Clin Invest ; 127(12): 4569-4582, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29058688

RESUMO

The devastating sequelae of diabetes mellitus include microvascular permeability, which results in retinopathy. Despite clinical and scientific advances, there remains a need for new approaches to treat retinopathy. Here, we have presented a possible treatment strategy, whereby targeting the small GTPase ARF6 alters VEGFR2 trafficking and reverses signs of pathology in 4 animal models that represent features of diabetic retinopathy and in a fifth model of ocular pathological angiogenesis. Specifically, we determined that the same signaling pathway utilizes distinct GEFs to sequentially activate ARF6, and these GEFs exert distinct but complementary effects on VEGFR2 trafficking and signal transduction. ARF6 activation was independently regulated by 2 different ARF GEFs - ARNO and GEP100. Interaction between VEGFR2 and ARNO activated ARF6 and stimulated VEGFR2 internalization, whereas a VEGFR2 interaction with GEP100 activated ARF6 to promote VEGFR2 recycling via coreceptor binding. Intervening in either pathway inhibited VEGFR2 signal output. Finally, using a combination of in vitro, cellular, genetic, and pharmacologic techniques, we demonstrated that ARF6 is pivotal in VEGFR2 trafficking and that targeting ARF6-mediated VEGFR2 trafficking has potential as a therapeutic approach for retinal vascular diseases such as diabetic retinopathy.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Retinopatia Diabética/metabolismo , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Linhagem Celular , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Transporte Proteico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
6.
Nat Cell Biol ; 19(9): 1027-1036, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28812582

RESUMO

Most differentiated cells convert glucose to pyruvate in the cytosol through glycolysis, followed by pyruvate oxidation in the mitochondria. These processes are linked by the mitochondrial pyruvate carrier (MPC), which is required for efficient mitochondrial pyruvate uptake. In contrast, proliferative cells, including many cancer and stem cells, perform glycolysis robustly but limit fractional mitochondrial pyruvate oxidation. We sought to understand the role this transition from glycolysis to pyruvate oxidation plays in stem cell maintenance and differentiation. Loss of the MPC in Lgr5-EGFP-positive stem cells, or treatment of intestinal organoids with an MPC inhibitor, increases proliferation and expands the stem cell compartment. Similarly, genetic deletion of the MPC in Drosophila intestinal stem cells also increases proliferation, whereas MPC overexpression suppresses stem cell proliferation. These data demonstrate that limiting mitochondrial pyruvate metabolism is necessary and sufficient to maintain the proliferation of intestinal stem cells.


Assuntos
Proliferação de Células , Drosophila melanogaster/metabolismo , Glicólise , Mucosa Intestinal/metabolismo , Mitocôndrias/metabolismo , Ácido Pirúvico/metabolismo , Células-Tronco/metabolismo , Acrilatos/farmacologia , Animais , Proteínas de Transporte de Ânions/antagonistas & inibidores , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Genótipo , Humanos , Intestinos/citologia , Intestinos/efeitos dos fármacos , Ácido Láctico/metabolismo , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Mitocondriais/metabolismo , Transportadores de Ácidos Monocarboxílicos , Fenótipo , Interferência de RNA , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Fatores de Tempo , Técnicas de Cultura de Tecidos , Transfecção
7.
Cancer Cell ; 29(6): 889-904, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27265506

RESUMO

Activating mutations in Gαq proteins, which form the α subunit of certain heterotrimeric G proteins, drive uveal melanoma oncogenesis by triggering multiple downstream signaling pathways, including PLC/PKC, Rho/Rac, and YAP. Here we show that the small GTPase ARF6 acts as a proximal node of oncogenic Gαq signaling to induce all of these downstream pathways as well as ß-catenin signaling. ARF6 activates these diverse pathways through a common mechanism: the trafficking of GNAQ and ß-catenin from the plasma membrane to cytoplasmic vesicles and the nucleus, respectively. Blocking ARF6 with a small-molecule inhibitor reduces uveal melanoma cell proliferation and tumorigenesis in a mouse model, confirming the functional relevance of this pathway and suggesting a therapeutic strategy for Gα-mediated diseases.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Melanoma/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/administração & dosagem , Neoplasias Uveais/tratamento farmacológico , beta Catenina/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/antagonistas & inibidores , Fatores de Ribosilação do ADP/genética , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Citoplasma/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , Transplante de Neoplasias , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Neoplasias Uveais/genética , Neoplasias Uveais/metabolismo
8.
Sci Signal ; 6(265): ra14, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23462101

RESUMO

ß-Catenin has a dual function in cells: fortifying cadherin-based adhesion at the plasma membrane and activating transcription in the nucleus. We found that in melanoma cells, WNT5A stimulated the disruption of N-cadherin and ß-catenin complexes by activating the guanosine triphosphatase adenosine diphosphate ribosylation factor 6 (ARF6). Binding of WNT5A to the Frizzled 4-LRP6 (low-density lipoprotein receptor-related protein 6) receptor complex activated ARF6, which liberated ß-catenin from N-cadherin, thus increasing the pool of free ß-catenin, enhancing ß-catenin-mediated transcription, and stimulating invasion. In contrast to WNT5A, the guidance cue SLIT2 and its receptor ROBO1 inhibited ARF6 activation and, accordingly, stabilized the interaction of N-cadherin with ß-catenin and reduced transcription and invasion. Thus, ARF6 integrated competing signals in melanoma cells, thereby enabling plasticity in the response to external cues. Moreover, small-molecule inhibition of ARF6 stabilized adherens junctions, blocked ß-catenin signaling and invasiveness of melanoma cells in culture, and reduced spontaneous pulmonary metastasis in mice, suggesting that targeting ARF6 may provide a means of inhibiting WNT/ß-catenin signaling in cancer.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Melanoma/patologia , Invasividade Neoplásica , Metástase Neoplásica , Proteínas Proto-Oncogênicas/fisiologia , Ativação Transcricional/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Fator 6 de Ribosilação do ADP , Inativação Gênica , Humanos , Transdução de Sinais , Proteína Wnt-5a , beta Catenina/metabolismo
9.
Nature ; 492(7428): 252-5, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23143332

RESUMO

The innate immune response is essential for combating infectious disease. Macrophages and other cells respond to infection by releasing cytokines, such as interleukin-1ß (IL-1ß), which in turn activate a well-described, myeloid-differentiation factor 88 (MYD88)-mediated, nuclear factor-κB (NF-κB)-dependent transcriptional pathway that results in inflammatory-cell activation and recruitment. Endothelial cells, which usually serve as a barrier to the movement of inflammatory cells out of the blood and into tissue, are also critical mediators of the inflammatory response. Paradoxically, the cytokines vital to a successful immune defence also have disruptive effects on endothelial cell-cell interactions and can trigger degradation of barrier function and dissociation of tissue architecture. The mechanism of this barrier dissolution and its relationship to the canonical NF-κB pathway remain poorly defined. Here we show that the direct, immediate and disruptive effects of IL-1ß on endothelial stability in a human in vitro cell model are NF-κB independent and are instead the result of signalling through the small GTPase ADP-ribosylation factor 6 (ARF6) and its activator ARF nucleotide binding site opener (ARNO; also known as CYTH2). Moreover, we show that ARNO binds directly to the adaptor protein MYD88, and thus propose MYD88-ARNO-ARF6 as a proximal IL-1ß signalling pathway distinct from that mediated by NF-κB. Finally, we show that SecinH3, an inhibitor of ARF guanine nucleotide-exchange factors such as ARNO, enhances vascular stability and significantly improves outcomes in animal models of inflammatory arthritis and acute inflammation.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores de Interleucina/metabolismo , Fator 6 de Ribosilação do ADP , Adjuvantes Imunológicos/farmacologia , Animais , Artrite/patologia , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Interleucina-1beta/farmacologia , NF-kappa B/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Purinas/farmacologia , Transdução de Sinais , Tiofenos/farmacologia
10.
Ann Biomed Eng ; 39(1): 337-46, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20737290

RESUMO

Elastin is an essential component of arteries which provides structural integrity and instructs smooth muscle cells to adopt a quiescent state. Despite interaction of endothelial cells with elastin in the internal elastic lamina, the potential for exploiting this interaction therapeutically has not been explored in detail. In this study, we show that tropoelastin (a precursor of elastin) stimulates endothelial cell migration and adhesion more than smooth muscle cells. The biological activity of tropoelastin on endothelial cells is contained in the VGVAPG domain and in the carboxy-terminal 17-amino acids. We show that the effects of the carboxy-terminal 17 amino acids, but not those of VGVAPG, are mediated by integrin α(V)ß(3). We demonstrate that tropoelastin covalently linked to stainless steel disks promotes adhesion of endothelial progenitor cells and endothelial cells to the metal surfaces. The adherent cells on the tropoelastin-coated metal surfaces form monolayers that can withstand and respond to arterial shear stress. Because of the unique effects of tropoelastin on endothelial and smooth muscle cells, coating intravascular devices with tropoelastin may stimulate their endothelialization, inhibit smooth muscle hyperplasia, and improve device performance.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Tropoelastina/administração & dosagem , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos
11.
Proc Natl Acad Sci U S A ; 107(23): 10520-5, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20498081

RESUMO

Formation of the vascular system within organs requires the balanced action of numerous positive and negative factors secreted by stromal and epithelial cells. Here, we used a genetic approach to determine the role of SLITs in regulating the growth and organization of blood vessels in the mammary gland. We demonstrate that vascularization of the gland is not affected by loss of Slit expression in the epithelial compartment. Instead, we identify a stromal source of SLIT, mural cells encircling blood vessels, and show that loss of Slit in the stroma leads to elevated blood vessel density and complexity. We examine candidate SLIT receptors, Robo1 and Robo4, and find that increased vessel angiogenesis is phenocopied by loss of endothelial-specific Robo4, as long as it is combined with the presence of an angiogenic stimulus such as preneoplasia or pregnancy. In contrast, loss of Robo1 does not affect blood vessel growth. The enhanced growth of blood vessels in Robo4(-/-) endothelium is due to activation of vascular endothelial growth factor (VEGF)-R2 signaling through the Src and FAK kinases. Thus, our studies present a genetic dissection of SLIT/ROBO signaling during organ development. We identify a stromal, rather than epithelial, source of SLITs that inhibits blood vessel growth by signaling through endothelial ROBO4 to down-regulate VEGF/VEGFR2 signaling.


Assuntos
Glândulas Mamárias Animais/irrigação sanguínea , Glândulas Mamárias Animais/metabolismo , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Receptores de Superfície Celular , Receptores Imunológicos/deficiência , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Roundabout
12.
Sci Transl Med ; 2(23): 23ra19, 2010 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-20375003

RESUMO

The innate immune system provides a first line of defense against invading pathogens by releasing multiple inflammatory cytokines, such as interleukin-1beta and tumor necrosis factor-alpha, which directly combat the infectious agent and recruit additional immune responses. This exuberant cytokine release paradoxically injures the host by triggering leakage from capillaries, tissue edema, organ failure, and shock. Current medical therapies target individual pathogens with antimicrobial agents or directly either blunt or boost the host's immune system. We explored a third approach: activating with the soluble ligand Slit an endothelium-specific, Robo4-dependent signaling pathway that strengthens the vascular barrier, diminishing deleterious aspects of the host's response to the pathogen-induced cytokine storm. This approach reduced vascular permeability in the lung and other organs and increased survival in animal models of bacterial endotoxin exposure, polymicrobial sepsis, and H5N1 influenza. Thus, enhancing the resilience of the host vascular system to the host's innate immune response may provide a therapeutic strategy for treating multiple infectious agents.


Assuntos
Citocinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Infecções por Orthomyxoviridae/imunologia , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Sepse/imunologia , Transdução de Sinais , Animais , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Cateninas/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/patologia , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Sepse/complicações , Sepse/patologia , Transdução de Sinais/efeitos dos fármacos , delta Catenina
13.
Nat Cell Biol ; 11(11): 1325-31, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19855388

RESUMO

Slit-Roundabout (Robo) signalling has a well-understood role in axon guidance. Unlike in the nervous system, however, Slit-dependent activation of an endothelial-specific Robo, Robo4, does not initiate a guidance program. Instead, Robo4 maintains the barrier function of the mature vascular network by inhibiting neovascular tuft formation and endothelial hyperpermeability induced by pro-angiogenic factors. In this study, we used cell biological and biochemical techniques to elucidate the molecular mechanism underlying the maintenance of vascular stability by Robo4. Here, we demonstrate that Robo4 mediates Slit2-dependent suppression of cellular protrusive activity through direct interaction with the intracellular adaptor protein paxillin and its paralogue, Hic-5. Formation of a Robo4-paxillin complex at the cell surface blocks activation of the small GTPase Arf6 and, consequently, Rac by recruitment of Arf-GAPs (ADP-ribosylation factor- directed GTPase-activating proteins) such as GIT1. Consistent with these in vitro studies, inhibition of Arf6 activity in vivo phenocopies Robo4 activation by reducing pathologic angiogenesis in choroidal and retinal vascular disease and VEGF-165 (vascular endothelial growth factor-165)-induced retinal hyperpermeability. These data reveal that a Slit2-Robo4-paxillin-GIT1 network inhibits the cellular protrusive activity underlying neovascularization and vascular leak, and identify a new therapeutic target for ameliorating diseases involving the vascular system.


Assuntos
Fatores de Ribosilação do ADP/antagonistas & inibidores , Vasos Sanguíneos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Fator 6 de Ribosilação do ADP , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Linhagem Celular , Movimento Celular , Cricetinae , Humanos , Camundongos
14.
PLoS Genet ; 4(5): e1000073, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18483557

RESUMO

In eutherian mammals, embryonic growth and survival is dependent on the formation of the placenta, an organ that facilitates the efficient exchange of oxygen, nutrients, and metabolic waste between the maternal and fetal blood supplies. Key to the placenta's function is the formation of its vascular labyrinth, a series of finely branched vessels whose molecular ontogeny remains largely undefined. In this report, we demonstrate that HOXA13 plays an essential role in labyrinth vessel formation. In the absence of HOXA13 function, placental endothelial cell morphology is altered, causing a loss in vessel wall integrity, edema of the embryonic blood vessels, and mid-gestational lethality. Microarray analysis of wild-type and mutant placentas revealed significant changes in endothelial gene expression profiles. Notably, pro-vascular genes, including Tie2 and Foxf1, exhibited reduced expression in the mutant endothelia, which also exhibited elevated expression of genes normally expressed in lymphatic or sinusoidal endothelia. ChIP analysis of HOXA13-DNA complexes in the placenta confirmed that HOXA13 binds the Tie2 and Foxf1 promoters in vivo. In vitro, HOXA13 binds sequences present in the Tie2 and Foxf1 promoters with high affinity (K(d) = 27-42 nM) and HOXA13 can use these bound promoter regions to direct gene expression. Taken together, these findings demonstrate that HOXA13 directly regulates Tie2 and Foxf1 in the placental labyrinth endothelia, providing a functional explanation for the mid-gestational lethality exhibited by Hoxa13 mutant embryos as well as a novel transcriptional program necessary for the specification of the labyrinth vascular endothelia.


Assuntos
Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Placenta/irrigação sanguínea , Animais , Sequência de Bases , Endotélio Vascular/embriologia , Endotélio Vascular/crescimento & desenvolvimento , Fatores de Transcrição Forkhead/genética , Genes Reporter , Proteínas de Homeodomínio/genética , Homozigoto , Técnicas In Vitro , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neovascularização Fisiológica , Análise de Sequência com Séries de Oligonucleotídeos , Placenta/embriologia , Placenta/metabolismo , Regiões Promotoras Genéticas , Receptor TIE-2/genética , Trofoblastos/fisiologia
15.
Development ; 135(4): 659-67, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18223200

RESUMO

There is emerging evidence that the canonical neural guidance factor netrin can also direct the growth of blood vessels. We deleted the gene encoding UNC5B, a receptor for the netrin family of guidance molecules, specifically within the embryonic endothelium of mice. The result is a profound structural and functional deficiency in the arterioles of the placental labyrinth, which leads first to flow reversal in the umbilical artery and ultimately to embryonic death. As this is the only detectable site of vascular abnormality in the mutant embryos, and because the phenotype cannot be rescued by a wild-type trophectoderm, we propose that UNC5B-mediated signaling is a specific and autonomous component of fetal-placental angiogenesis. Disruption of UNC5B represents a unique example of a mutation that acts solely within the fetal-placental vasculature and one that faithfully recapitulates the structural and physiological characteristics of clinical uteroplacental insufficiency. This pro-angiogenic, but spatially restricted requirement for UNC5B is not unique to murine development, as the knock-down of the Unc5b ortholog in zebrafish similarly results in the specific and highly penetrant absence of the parachordal vessel, the precursor to the lymphatic system.


Assuntos
Vasos Sanguíneos/embriologia , Neovascularização Fisiológica , Receptores de Superfície Celular/metabolismo , Animais , Arteríolas/anormalidades , Arteríolas/patologia , Vasos Sanguíneos/metabolismo , Perda do Embrião , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/patologia , Embrião não Mamífero/citologia , Endotélio/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipóxia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Netrina , Especificidade de Órgãos , Fenótipo , Placenta/metabolismo , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Fluxo Sanguíneo Regional , Transdução de Sinais , Cordão Umbilical/irrigação sanguínea , Peixe-Zebra/embriologia
16.
Science ; 313(5787): 640-4, 2006 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-16809490

RESUMO

Axonal guidance and vascular patterning share several guidance cues, including proteins in the netrin family. We demonstrate that netrins stimulate proliferation, migration, and tube formation of human endothelial cells in vitro and that this stimulation is independent of known netrin receptors. Suppression of netrin1a messenger RNA in zebrafish inhibits vascular sprouting, implying a proangiogenic role for netrins during vertebrate development. We also show that netrins accelerate neovascularization in an in vivo model of ischemia and that they reverse neuropathy and vasculopathy in a diabetic murine model. We propose that the attractive vascular and neural guidance functions of netrins offer a unique therapeutic potential.


Assuntos
Células Endoteliais/fisiologia , Neovascularização Fisiológica , Fatores de Crescimento Neural/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Indutores da Angiogênese , Animais , Linhagem Celular , Movimento Celular , Quimiotaxia , DNA Complementar , Angiopatias Diabéticas/terapia , Neuropatias Diabéticas/terapia , Embrião não Mamífero , Endotélio Vascular/citologia , Terapia Genética , Humanos , Isquemia/tratamento farmacológico , Camundongos , Músculo Esquelético/irrigação sanguínea , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/farmacologia , Receptores de Netrina , Netrina-1 , Netrinas , Condução Nervosa , Receptores de Superfície Celular/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/farmacologia , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Peixe-Zebra
17.
Proc Natl Acad Sci U S A ; 101(46): 16210-5, 2004 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-15520390

RESUMO

Blood vessels and nerves often follow parallel trajectories, suggesting that distal targets use common cues that induce vascularization and innervation. Netrins are secreted by the floor plate and attract commissural axons toward the midline of the neural tube. Here, we show that Netrin-1 is also a potent vascular mitogen. Netrin-1 stimulates proliferation, induces migration, and promotes adhesion of endothelial cells and vascular smooth muscle cells with a specific activity comparable to vascular endothelial growth factor and platelet-derived growth factor. Our evidence indicates that the netrin receptor, Neogenin, mediates netrin signaling in vascular smooth muscle cells, but suggests that an unidentified receptor mediates the proangiogenic effects of Netrin-1 on endothelial cells. Netrin-1 also stimulates angiogenesis in vivo and augments the response to vascular endothelial growth factor. Thus, we demonstrate that Netrin-1 is a secreted neural guidance cue with the unique ability to attract both blood vessels and axons, and suggest that other cues may also function as vascular endothelial growth factors.


Assuntos
Proteínas Angiogênicas/fisiologia , Axônios/fisiologia , Fatores de Crescimento Neural/fisiologia , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/farmacologia , Animais , Sequência de Bases , Bovinos , DNA/genética , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Técnicas In Vitro , Masculino , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/farmacologia , Netrina-1 , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Proteínas Supressoras de Tumor , Fator A de Crescimento do Endotélio Vascular/fisiologia
18.
Dev Biol ; 261(1): 235-50, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12941632

RESUMO

Several characteristic morphological and functional differences distinguish arteries from veins. It was thought that hemodynamic forces shaped these differences; however, increasing evidence suggests that morphogenetic programs play a central role in blood vessel differentiation. Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia characterized by the inappropriate fusion of arterioles with venules. The genes implicated in this disease, ALK1 and endoglin, may be involved in defining the fundamental boundaries between arteries and veins. We previously showed that mice lacking Alk1 lost structural, molecular, and functional distinctions between arteries and veins. Here, we report that mice lacking endoglin develop arterial-venous malformations and fail to confine intraembryonic hematopoiesis to arteries. In contrast to Alk1 mutants, endoglin mutants do not show profound vessel dilation or downregulation of arterial ephrinB2. Finally, our data indicate that a failure in cardiac cushion formation observed in both strains may be secondary to the peripheral vasculature defect. The phenotypic similarities, yet reduced severity, implicates endoglin as an accessory coreceptor that specifically modulates Alk1 signaling. We propose that endoglin and Alk1 are necessary for the maintenance of distinct arterial-venous vascular beds and that attenuation of the Alk1 signaling pathway is the precipitating event in the etiology of HHT.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Vasos Sanguíneos/embriologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Molécula 1 de Adesão de Célula Vascular/fisiologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Antígenos CD , Artérias/embriologia , Malformações Arteriovenosas/embriologia , Malformações Arteriovenosas/genética , Sequência de Bases , DNA/genética , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/genética , Endoglina , Efrina-B2/genética , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptores de Superfície Celular , Telangiectasia Hemorrágica Hereditária/embriologia , Telangiectasia Hemorrágica Hereditária/genética , Molécula 1 de Adesão de Célula Vascular/genética , Veias/embriologia
19.
Dev Biol ; 261(1): 251-67, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12941633

RESUMO

Guidance and patterning of axons are orchestrated by cell-surface receptors and ligands that provide directional cues. Interactions between the Robo receptor and Slit ligand families of proteins initiate signaling cascades that repel axonal outgrowth. Although the vascular and nervous systems grow as parallel networks, the mechanisms by which the vascular endothelial cells are guided to their appropriate positions remain obscure. We have identified a putative Robo homologue, Robo4, based on its differential expression in mutant mice with defects in vascular sprouting. In contrast to known neuronal Robo family members, the arrangement of the extracellular domains of Robo4 diverges significantly from that of all other Robo family members. Moreover, Robo4 is specifically expressed in the vascular endothelium during murine embryonic development. We show that Robo4 binds Slit and inhibits cellular migration in a heterologous expression system, analogous to the role of known Robo receptors in the nervous system. Immunoprecipitation studies indicate that Robo4 binds to Mena, a known effector of Robo-Slit signaling. Finally, we show that Robo4 is the only Robo family member expressed in primary endothelial cells and that application of Slit inhibits their migration. These data demonstrate that Robo4 is a bona fide member of the Robo family and may provide a repulsive cue to migrating endothelial cells during vascular development.


Assuntos
Endotélio Vascular/embriologia , Receptores Imunológicos/fisiologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Receptores de Activinas Tipo II , Sequência de Aminoácidos , Animais , Linhagem Celular , Movimento Celular , Mapeamento Cromossômico , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Imunológicos/química , Receptores Imunológicos/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Peixe-Zebra , Proteínas Roundabout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA