Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 289(21): 14881-95, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24695740

RESUMO

Gene activation requires cooperative assembly of multiprotein transcription factor-coregulator complexes. Disruption to cooperative assemblage could underlie repression of tumor suppressor genes in leukemia cells. Mechanisms of cooperation and its disruption were therefore examined for PU.1 and RUNX1, transcription factors that cooperate to activate hematopoietic differentiation genes. PU.1 is highly expressed in leukemia cells, whereas RUNX1 is frequently inactivated by mutation or translocation. Thus, coregulator interactions of Pu.1 were examined by immunoprecipitation coupled with tandem mass spectrometry/Western blot in wild-type and Runx1-deficient hematopoietic cells. In wild-type cells, the NuAT and Baf families of coactivators coimmunoprecipitated with Pu.1. Runx1 deficiency produced a striking switch to Pu.1 interaction with the Dnmt1, Sin3A, Nurd, CoRest, and B-Wich corepressor families. Corepressors of the Polycomb family, which are frequently inactivated by mutation or deletion in myeloid leukemia, did not interact with Pu.1. The most significant gene ontology association of Runx1-Pu.1 co-bound genes was with macrophages, therefore, functional consequences of altered corepressor/coactivator exchange were examined at Mcsfr, a key macrophage differentiation gene. In chromatin immunoprecipitation analyses, high level Pu.1 binding to the Mcsfr promoter was not decreased by Runx1 deficiency. However, the Pu.1-driven shift from histone repression to activation marks at this locus, and terminal macrophage differentiation, were substantially diminished. DNMT1 inhibition, but not Polycomb inhibition, in RUNX1-translocated leukemia cells induced terminal differentiation. Thus, RUNX1 and PU.1 cooperate to exchange corepressors for coactivators, and the specific corepressors recruited to PU.1 as a consequence of RUNX1 deficiency could be rational targets for leukemia differentiation therapy.


Assuntos
Diferenciação Celular/genética , Proteínas Correpressoras/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Proteínas Correpressoras/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Espectrometria de Massas em Tandem , Transativadores/metabolismo , Células Tumorais Cultivadas
2.
Am J Pathol ; 180(4): 1702-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22322296

RESUMO

Caveolin-1 is an essential structural protein of caveolae, specialized plasma membrane organelles highly abundant in endothelial cells, where they regulate multiple functions including angiogenesis. Caveolin-1 exerts a tonic inhibition of endothelial nitric oxide synthase (eNOS) activity. Accordingly, caveolin-1 gene-disrupted mice have enhanced eNOS activity as well as increased systemic nitric oxide (NO) levels. We hypothesized that excess eNOS activity, secondary to caveolin deficiency, would mediate the decreased angiogenesis observed in caveolin-1 gene-disrupted mice. We tested tumor angiogenesis in mice lacking either one or both proteins, using in vitro, ex vivo, and in vivo assays. We show that endothelial cell migration, tube formation, cell sprouting from aortic rings, tumor growth, and angiogenesis are all significantly impaired in both caveolin-1-null and eNOS-null mice. We further show that these parameters were either partially or fully restored in double knockout mice that lack both caveolin-1 and eNOS. Furthermore, the effects of genetic ablation of eNOS are mimicked by the administration of the NOS inhibitor N-nitro-L-arginine methyl ester hydrochloride (L-NAME), including the reversal of the caveolin-1-null mouse angiogenic phenotype. This study is the first to demonstrate the detrimental effects of unregulated eNOS activity on angiogenesis, and shows that impaired tumor angiogenesis in caveolin-1-null mice is, at least in part, the result of enhanced eNOS activity.


Assuntos
Caveolina 1/deficiência , Neovascularização Patológica/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Animais , Caveolina 1/genética , Caveolina 1/fisiologia , Células Cultivadas , Quimiotaxia/fisiologia , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/prevenção & controle , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Inibidores Enzimáticos/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , NG-Nitroarginina Metil Éster/uso terapêutico , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
3.
Mol Vis ; 18: 1260-70, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22665973

RESUMO

PURPOSE: Cited2 (CBP/p300-interacting transactivators with glutamic acid (E) and aspartic acid (D)-rich tail 2) is a member of a new family of transcriptional modulators. Cited2 null embryos exhibit hyaloid hypercellularity consisting of aberrant vasculature in the eye. The purpose of the study is to address whether abnormal lenticular development is a primary defect of Cited2 deletion and whether deletion of hypoxia inducible factor (HIF)-1α or an HIF-1α target gene, vascular endothelial growth factor (VEGF), could rescue abnormal hyaloid vascular system (HVS) in Cited2 deficient adult eyes. METHODS: Le-Cre specific Cited2 knockout (Cited2(CKO)) mice with or without deletion of HIF-1α or VEGF were generated by standard Cre-Lox methods. Eyes collected from six-eight weeks old mice were characterized by Real Time PCR and immunohistological staining. RESULTS: Cited2(CKO) mice had smaller lenses, abnormal lens stalk formation, and failed regression of the HVS in the adult eye. The eye phenotype had features similar to persistent hyperplastic primary vitreous (PHPV), a human congenital eye disorder leading to abnormal lenticular development. Deletion of HIF-1α or VEGF in Cited2 knockout eyes partially rescued the abnormal HVS but had no effect on the smaller lens and abnormal lens stalk differentiation. Intravitreal injection of Topotecan (TPT), a compound that inhibits HIF-1α expression, partially eliminated HVS defects in Cited2(CKO) lenses. CONCLUSIONS: Abnormal HVS is a primary defect in Cited2 knockout mice, resulting in part from dysregulated functions of HIF-1 and VEGF. The Cited2(CKO) mouse line could be used as a novel disease model for PHPV and as an in vivo model for testing potential HIF-1 inhibitors.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Cristalino/metabolismo , Proteínas Repressoras/genética , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Cristalino/efeitos dos fármacos , Cristalino/patologia , Camundongos , Camundongos Knockout , Vítreo Primário Hiperplásico Persistente/genética , Vítreo Primário Hiperplásico Persistente/metabolismo , Vítreo Primário Hiperplásico Persistente/patologia , Ligação Proteica , Proteínas Repressoras/deficiência , Deleção de Sequência , Inibidores da Topoisomerase I/administração & dosagem , Topotecan/administração & dosagem , Transativadores/deficiência , Fator A de Crescimento do Endotélio Vascular/deficiência
4.
Am J Pathol ; 176(1): 496-503, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19948826

RESUMO

Matrix metalloproteinases (MMPs), a specialized group of enzymes capable of proteolytically degrading extracellular matrix proteins, have been postulated to play an important role in angiogenesis. It has been suggested that MMPs can regulate neovascularization using mechanisms other than simple remodeling of the capillary basement membrane. To determine the interplay between vascular endothelial growth factor (VEGF) and MMPs, we investigated the induction of angiogenesis by recombinant active MMPs and VEGF in vivo. Using a rat corneal micropocket in vivo angiogenesis assay, we observed that the active form of MMP-9 could induce neovascularization in vivo when compared with the pro- form of the enzyme as a control. This angiogenic response could be inhibited by neutralizing VEGF antibody, which suggests that MMPs acts upstream of VEGF. Additional in vitro studies using extracellular matrix loaded with radiolabeled VEGF determined that active MMPs can enzymatically release sequestered VEGF. Interestingly, in vivo angiogenesis induced by VEGF could be inhibited by MMP inhibitors, indicating that MMPs also act downstream of VEGF. In addition, inflammation plays an important role in the induction of angiogenesis mediated by both VEGF and MMPs. Our results suggest that MMPs act both upstream and downstream of VEGF and imply that potential combination therapies of VEGF and MMP inhibitors may be a useful therapeutic approach in diseases of pathological neovascularization.


Assuntos
Córnea/irrigação sanguínea , Córnea/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Córnea/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Hospedeiro Imunocomprometido , Implantes Experimentais , Metaloproteinase 2 da Matriz/farmacologia , Metaloproteinase 9 da Matriz/farmacologia , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Testes de Neutralização , Ratos , Fator A de Crescimento do Endotélio Vascular/farmacologia
5.
Nat Med ; 9(4): 407-15, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12652295

RESUMO

Tissue inhibitor of metalloproteinases-3 (TIMP3) is one of four members of a family of proteins that were originally classified according to their ability to inhibit matrix metalloproteinases (MMP). TIMP3, which encodes a potent angiogenesis inhibitor, is mutated in Sorsby fundus dystrophy, a macular degenerative disease with submacular choroidal neovascularization. In this study we demonstrate the ability of TIMP3 to inhibit vascular endothelial factor (VEGF)-mediated angiogenesis and identify the potential mechanism by which this occurs: TIMP3 blocks the binding of VEGF to VEGF receptor-2 and inhibits downstream signaling and angiogenesis. This property seems to be independent of its MMP-inhibitory activity, indicating a new function for this molecule.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfocinas/metabolismo , Neovascularização Patológica/prevenção & controle , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores Teciduais de Metaloproteinases/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Fatores de Crescimento Endotelial/farmacologia , Endotélio/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Linfocinas/farmacologia , Neuropilinas/metabolismo , Fosforilação , Proteínas Recombinantes/farmacologia , Inibidor Tecidual de Metaloproteinase-3 , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
6.
Proc Natl Acad Sci U S A ; 105(50): 19898-903, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19057008

RESUMO

Oxygen-induced retinopathy (OIR) in the mouse, like the analogous human disease retinopathy of prematurity, is an ischemic retinopathy dependent on oxygen-induced vascular obliteration. We tested the hypothesis that chemically overriding the oxygen-induced downregulation of hypoxia-inducible factor (HIF) activity would prevent vascular obliteration and subsequent pathologic neovascularization in the OIR model. Because the degradation of HIF-1alpha is regulated by prolyl hydroxylases, we examined the effect of systemic administration of a prolyl hydroxylase inhibitor, dimethyloxalylglycine, in the OIR model. Our results determine that stabilizing HIF activity in the early phase of OIR prevents the oxygen-induced central vessel loss and subsequent vascular tortuosity and tufting that is characteristic of OIR. Overall, these findings imply that simulating hypoxia chemically by stabilizing HIF activity during the causative ischemia phase (hyperoxia) of retinopathy of prematurity may be of therapeutic value in preventing progression to the proliferative stage of the disease.


Assuntos
Aminoácidos Dicarboxílicos/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Oxigênio/toxicidade , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Retinopatia da Prematuridade/prevenção & controle , Aerobiose , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Modelos Animais de Doenças , Eritropoetina/biossíntese , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/agonistas , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Recém-Nascido , Rim/metabolismo , Fígado/metabolismo , Camundongos , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Retina/metabolismo , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/enzimologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
7.
J Clin Invest ; 128(10): 4260-4279, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30015632

RESUMO

Nucleophosmin (NPM1) is among the most frequently mutated genes in acute myeloid leukemia (AML). It is not known, however, how the resulting oncoprotein mutant NPM1 is leukemogenic. To reveal the cellular machinery in which NPM1 participates in myeloid cells, we analyzed the endogenous NPM1 protein interactome by mass spectrometry and discovered abundant amounts of the master transcription factor driver of monocyte lineage differentiation PU.1 (also known as SPI1). Mutant NPM1, which aberrantly accumulates in cytoplasm, dislocated PU.1 into cytoplasm with it. CEBPA and RUNX1, the master transcription factors that collaborate with PU.1 to activate granulomonocytic lineage fates, remained nuclear; but without PU.1, their coregulator interactions were toggled from coactivators to corepressors, repressing instead of activating more than 500 granulocyte and monocyte terminal differentiation genes. An inhibitor of nuclear export, selinexor, by locking mutant NPM1/PU.1 in the nucleus, activated terminal monocytic fates. Direct depletion of the corepressor DNA methyltransferase 1 (DNMT1) from the CEBPA/RUNX1 protein interactome using the clinical drug decitabine activated terminal granulocytic fates. Together, these noncytotoxic treatments extended survival by more than 160 days versus vehicle in a patient-derived xenotransplant model of NPM1/FLT3-mutated AML. In sum, mutant NPM1 represses monocyte and granulocyte terminal differentiation by disrupting PU.1/CEBPA/RUNX1 collaboration, a transforming action that can be reversed by pharmacodynamically directed dosing of clinical small molecules.


Assuntos
Granulócitos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Monócitos/metabolismo , Mutação , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Granulócitos/patologia , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Monócitos/patologia , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Proteínas Nucleares/genética , Nucleofosmina , Células THP-1 , Fatores de Transcrição/genética
8.
Invest Ophthalmol Vis Sci ; 47(11): 4935-41, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17065510

RESUMO

PURPOSE: To test whether triamcinolone acetonide (TA) inhibits angiogenesis induced by IL-6 or VEGF and whether this inhibition is through antagonism of the IL-6 or the VEGF receptor 2. METHODS: A rat cornea micropocket assay was used to initiate IL-6- and VEGF-mediated angiogenesis. The ability of TA or neutralizing VEGF antibody to inhibit IL-6- or VEGF-mediated neovascularization was analyzed by measuring vessel length, vessel extension, and vessel area. The phosphorylation of signal transduction activator 3 (STAT3), VEGF receptor, and extracellular signal-regulated kinase 1/2 (ERK1/2) was determined by Western blot in human umbilical vein endothelial cell (HUVEC) lysates after stimulus with IL-6 or VEGF, with and without TA pretreatment. The effect of IL-6 or TA on STAT3 expression in cornea was determined by Western blot. RESULTS: IL-6 induced corneal angiogenesis in a dose-dependent manner, with 350 ng producing a peak at day 6. VEGF antibodies and TA blocked IL-6-mediated limbal neovascularization. TA also directly inhibited angiogenesis stimulated by a VEGF pellet; the glucocorticoid receptor antagonist mifepristone neutralized TA inhibition of angiogenesis. TA did not inhibit IL-6-induced STAT3 phosphorylation and did not inhibit VEGF-induced phosphorylation of the VEGF receptor 2 or of ERK1/2 in endothelial cells, but TA decreased IL-6-induced STAT3 expression in cornea. CONCLUSIONS: IL-6- and VEGF-mediated corneal neovascularization are blocked by TA through the mifepristone-sensitive steroid receptor. TA inhibits IL-6-induced STAT3 expression in cornea, but it does not inhibit activation of the IL-6 or the VEGF receptor in cultured human endothelial cells. This finding has two implications. The fact that TA directly inhibits VEGF action implies that other factors may be critical to angiogenesis and sensitive to glucocorticoids.


Assuntos
Neovascularização da Córnea/tratamento farmacológico , Glucocorticoides/farmacologia , Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Triancinolona Acetonida/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Western Blotting , Membrana Corioalantoide/irrigação sanguínea , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Interleucina-6/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Ratos , Ratos Endogâmicos F344 , Fator de Transcrição STAT3/metabolismo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
9.
FASEB J ; 19(7): 819-21, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15857892

RESUMO

Angiogenesis is critical for many physiological and pathological processes. We show here that the lipid sphingosylphosphorylcholine (SPC) induces angiogenesis in vivo and GPR4 is required for the biological effects of SPC on endothelial cells (EC). In human umbilical vein EC, down-regulation of GPR4 specifically inhibits SPC-, but not sphingosine-1-phosphate-, or vascular endothelial growth factor (VEGF)-induced tube formation. Re-introduction of GPR4 fully restores the activity of SPC. In microvascular EC, GPR4 plays a pivotal role in cell survival, growth, migration, and tube formation through both SPC-dependent and -independent pathways. The biological effects resulting from SPC/GPR4 interactions involve the activation of both phosphatidylinositol-3 kinase and Akt. Moreover, the effects of SPC on EC require SPC induced trans-phosphorylation and activation of the VEGF receptor 2. These results identify SPC and its receptor, GPR4, as critical regulators of the angiogenic potential of EC.


Assuntos
Células Endoteliais/fisiologia , Neovascularização Fisiológica/fisiologia , Fosforilcolina/análogos & derivados , Receptores Acoplados a Proteínas G/fisiologia , Esfingosina/análogos & derivados , Animais , Anticorpos/farmacologia , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Embrião de Galinha , Ativação Enzimática , Expressão Gênica/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Lisofosfolipídeos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Fragmentos de Peptídeos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilcolina/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Esfingosina/farmacologia , Veias Umbilicais , Fator A de Crescimento do Endotélio Vascular/farmacologia
10.
Exp Biol Med (Maywood) ; 239(7): 853-861, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24879424

RESUMO

Cancer chemotherapy is typically toxic. This problem could be addressed by using differences between cancer and normal cells for controlled delivery of drugs to cancer cells. One such difference is the ubiquitously elevated glutathione expression in cancer cells. We report a simple and versatile synthesis of water-soluble gold nanoparticles passivated with amine-containing molecules, which allow for controlled drug release via ligand exchange with bio-available glutathione. Taking methotrexate-passivated gold nanoparticles (Au:MTX) as an example, drug delivery and controlled release via glutathione-mediated ligand exchange was evaluated. Furthermore, the possibility of using Au:MTX to improve therapeutic index in acute myeloid leukemia (AML) models was examined in vitro and in vivo. Au:MTX exhibited cancer selectivity in vitro. Au:MTX had an elevated potency toward an AML cell line THP-1 in a dosage range of 1-5 nM, and therefore an enhanced delivery of drug, whereas normal hematopoietic stem/progenitor cell (HSPC) growth was minimally affected by Au:MTX and MTX treatments within the same range of dosage. In vivo efficacy and safety of Au:MTX was evaluated in a murine xenotransplant model of primary human AML. Au:MTX treatment, compared to control groups including MTX-only and Au nanoparticle-only treatments, produced better leukemia suppression without added toxicity, indicating an enhanced therapeutic index.

12.
PLoS One ; 8(3): e55667, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469166

RESUMO

Tissue inhibitors of metalloproteinases (TIMPs) while originally characterized as inhibitors of matrix metalloproteinases (MMPs) have recently been shown to have a wide range of functions that are independent of their MMP inhibitory properties. Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a potent inhibitor of VEGF-mediated angiogenesis and neovascularization through its ability to block the binding of VEGF to its receptor VEGFR-2. To identify and characterize the anti-angiogenic domain of TIMP-3, structure function analyses and synthetic peptide studies were performed using VEGF-mediated receptor binding, signaling, migration and proliferation. In addition, the ability of TIMP-3 peptides to inhibit CNV in a mouse model was evaluated. We demonstrate that the anti-angiogenic property resides in the COOH-terminal domain of TIMP-3 protein which can block the binding of VEGF specifically to its receptor VEGFR-2, but not to VEGFR-1 similar to the full-length wild-type protein. Synthetic peptides corresponding to putative loop 6 and tail region of TIMP-3 have anti-angiogenic properties as determined by inhibition of VEGF binding to VEGFR-2, VEGF-induced phosphorylation of VEGFR-2 and downstream signaling pathways as well as endothelial cell proliferation and migration in response to VEGF. In addition, we show that intravitreal administration of TIMP-3 peptide could inhibit the size of laser-induced choroidal neovascularization lesions in mice. Thus, we have identified TIMP-3 peptides to be efficient inhibitors of angiogenesis and have a potential to be used therapeutically in diseases with increased neovascularization.


Assuntos
Inibidores da Angiogênese/farmacologia , Lâmina Basilar da Corioide/irrigação sanguínea , Lâmina Basilar da Corioide/efeitos dos fármacos , Neovascularização de Coroide/prevenção & controle , Peptídeos/farmacologia , Inibidor Tecidual de Metaloproteinase-3/antagonistas & inibidores , Inibidor Tecidual de Metaloproteinase-3/química , Inibidores da Angiogênese/síntese química , Animais , Lâmina Basilar da Corioide/lesões , Células Cultivadas , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Endotélio Vascular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fotocoagulação a Laser/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/síntese química , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Técnicas de Síntese em Fase Sólida , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Clin Cancer Res ; 19(4): 938-48, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23287564

RESUMO

PURPOSE: The cytidine analogs 5-azacytidine and decitabine, used to treat myelodysplastic syndromes (MDS), produce a molecular epigenetic effect, depletion of DNA-methyltransferase 1 (DNMT1). This action is S-phase dependent. Hence, genetic factors that decrease the half-lives of these drugs could impact efficacy. Documentation of such impact, and elucidation of underlying mechanisms, could lead to improved clinical application. EXPERIMENTAL DESIGN: Cytidine deaminase (CDA) rapidly inactivates 5-azacytidine/decitabine. The effect of CDA SNP A79C and gender on CDA expression, enzyme activity, and drug pharmacokinetics/pharmacodynamics was examined in mice and humans, and the impact on overall survival (OS) was evaluated in 5-azacytidine/decitabine-treated patients with MDS (n = 90) and cytarabine-treated patients with acute myeloid leukemia (AML) (n = 76). RESULTS: By high-performance liquid chromatography (HPLC), plasma CDA activity was decreased as expected in individuals with the SNP A79C. Interestingly and significantly, there was an even larger decrease in females than in males. Explaining this decrease, liver CDA expression was significantly lower in female versus male mice. As expected, decitabine plasma levels, measured by mass spectrometry, were significantly higher in females. In mathematical modeling, the detrimental impact of shorter drug half-life (e.g., in males) was greater in low compared with high S-phase fraction disease (e.g., MDS vs. AML), because in high S-phase fraction disease, even a short exposure treats a major portion of cells. Accordingly, in multivariate analysis, OS was significantly worse in male versus female patients with MDS treated with 5-azacytidine/decitabine. CONCLUSIONS: Increased CDA expression/activity in males contributes to decreased cytidine analog half-life and likely contributes to worse outcomes with 5-azacytidine or decitabine therapy.


Assuntos
Citidina Desaminase/genética , DNA (Citosina-5-)-Metiltransferases/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/tratamento farmacológico , Animais , Azacitidina/análogos & derivados , Citarabina/administração & dosagem , Citidina/administração & dosagem , Citidina/análogos & derivados , Citidina Desaminase/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Polimorfismo de Nucleotídeo Único , Análise de Sobrevida , Resultado do Tratamento
14.
Oncotarget ; 3(10): 1137-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23087155

RESUMO

We document for the first time that sanctuary in an organ which expresses high levels of the enzyme cytidine deaminase (CDA) is a mechanism of cancer cell resistance to cytidine analogues. This mechanism could explain why historically, cytidine analogues have not been successful chemotherapeutics against hepatotropic cancers, despite efficacy in vitro. Importantly, this mechanism of resistance can be readily reversed, without increasing toxicity to sensitive organs, by combining a cytidine analogue with an inhibitor of cytidine deaminase (tetrahydrouridine). Specifically, CDA rapidly metabolizes cytidine analogues into inactive uridine counterparts. Hence, to determine if sheltering/protection of cancer cells in organs which express high levels of CDA (e.g., liver) is a mechanism of resistance, we utilized a murine xenotransplant model of myeloid cancer that is sensitive to epigenetic therapeutic effects of the cytidine analogue decitabine in vitro and hepato-tropic in vivo. Treatment of tumor-bearing mice with decitabine (subcutaneous 0.2mg/kg 2X/week) doubled median survival and significantly decreased extra-hepatic tumor burden, but hepatic tumor burden remained substantial, to which the animals eventually succumbed. Combining a clinically-relevant inhibitor of CDA (tetrahydrouridine) with a lower dose of decitabine (subcutaneous 0.1mg/kg 2X/week) markedly decreased liver tumor burden without blood count or bone marrow evidence of myelotoxicity, and with further improvement in survival. In conclusion, sanctuary in a CDA-rich organ is a mechanism by which otherwise susceptible cancer cells can resist the effects of decitabine epigenetic therapy. This protection can be reversed without increasing myelotoxicity by combining tetrahydrouridine with a lower dose of decitabine.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/análogos & derivados , Citidina Desaminase/metabolismo , Leucemia Mieloide/enzimologia , Neoplasias Hepáticas/enzimologia , Fígado/enzimologia , Animais , Antimetabólitos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica , Azacitidina/uso terapêutico , Decitabina , Humanos , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Fígado/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Tetra-Hidrouridina/uso terapêutico , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia
15.
Invest Ophthalmol Vis Sci ; 52(9): 6117-23, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21282576

RESUMO

PURPOSE: Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a matrix-bound inhibitor of matrix metalloproteinases (MMPs). The authors have previously determined a novel function of TIMP-3 to inhibit vascular endothelial growth factor (VEGF)-mediated angiogenesis. Here, the authors examined the in vivo angiogenic phenotype of ocular vessels in mice deficient in TIMP-3. METHODS: VEGF-mediated corneal neovascularization and laser-induced choroidal neovascularization (CNV) were examined in TIMP-3-null mice. The effects of the absence of TIMP-3 on the phosphorylation status of the VEGF-receptor-2 (VEGFR-2) and the downstream signaling pathways were evaluated biochemically. In addition, the activation state of MMPs in the retina of TIMP-3-deficient mice was examined by in situ zymography. RESULTS: The results of these studies determine an accentuation of pathologic VEGF-mediated angiogenesis in the cornea and laser-induced CNV in mice lacking TIMP-3. In the absence of the MMP inhibitor, pathophysiological changes were observed in the choroidal vasculature concomitantly with an increase in gelatinolytic activity. These results suggest that an imbalance of extracellular matrix homeostasis, together with a loss of an angiogenesis inhibitor, can prime vascular beds to be more responsive to an angiogenic stimulus. CONCLUSIONS: In light of the recent studies suggesting that genetic variants near TIMP-3 influence susceptibility to age-related macular degeneration, these results imply that TIMP-3 may regulate the development of the choroidal vasculature and is a likely contributor to increased susceptibility to choroidal neovascularization.


Assuntos
Neovascularização de Coroide/fisiopatologia , Neovascularização da Córnea/fisiopatologia , Modelos Animais de Doenças , Inibidor Tecidual de Metaloproteinase-3/deficiência , Animais , Neovascularização de Coroide/metabolismo , Neovascularização da Córnea/metabolismo , Immunoblotting , Imunoprecipitação , Fotocoagulação a Laser , Metaloproteinase 3 da Matriz/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oftalmoscopia , Fosforilação , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
16.
PLoS One ; 5(10): e13444, 2010 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-20976146

RESUMO

BACKGROUND: Diabetic maculopathy, the leading cause of vision loss in patients with type 2 diabetes, is characterized by hyper-permeability of retinal blood vessels with subsequent formation of macular edema and hard exudates. The degree of hyperglycemia and duration of diabetes have been suggested to be good predictors of retinal complications. Intervention studies have determined that while intensive treatment of diabetes reduced the development of proliferative diabetic retinopathy it was associated with a two to three-fold increased risk of severe hypoglycemia. Thus we hypothesized the need to identify downstream glycemic targets, which induce retinal vascular permeability that could be targeted therapeutically without the additional risks associated with intensive treatment of the hyperglycemia. Betacellulin is a 32 kD member of the epidermal growth factor family with mitogenic properties for the retinal pigment epithelial cells. This led us to hypothesize a role for betacellulin in the retinal vascular complications associated with diabetes. METHODS AND FINDINGS: In this study, using a mouse model of diabetes, we demonstrate that diabetic mice have accentuated retinal vascular permeability with a concomitant increased expression of a cleaved soluble form of betacellulin (s-Btc) in the retina. Intravitreal injection of soluble betacellulin induced retinal vascular permeability in normoglycemic and hyperglycemic mice. Western blot analysis of retinas from patients with diabetic retinopathy showed an increase in the active soluble form of betacellulin. In addition, an increase in the levels of A disintegrin and metalloproteinase (ADAM)-10 which plays a role in the cleavage of betacellulin was seen in the retinas of diabetic mice and humans. CONCLUSIONS: These results suggest that excessive amounts of betacellulin in the retina may contribute to the pathogenesis of diabetic macular edema.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Vasos Retinianos/efeitos dos fármacos , Animais , Betacelulina , Barreira Hematorretiniana , Eletroforese em Gel de Poliacrilamida , Humanos , Camundongos , Camundongos Endogâmicos C57BL
17.
Mol Cancer Ther ; 9(6): 1536-43, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20501800

RESUMO

The cytosine analogue decitabine alters hematopoietic differentiation. For example, decitabine treatment increases self-renewal of normal hematopoietic stem cells. The mechanisms underlying decitabine-induced shifts in differentiation are poorly understood, but likely relate to the ability of decitabine to deplete the chromatin-modifying enzyme DNA methyltransferase 1 (DNMT1), which plays a central role in transcription repression. HOXB4 is a transcription factor that promotes hematopoietic stem cell self-renewal. In hematopoietic precursors induced to differentiate by the lineage-specifying transcription factor Pu.1 or by the cytokine granulocyte-colony stimulating factor, there is rapid repression of HOXB4 and other stem cell genes. Depletion of DNMT1 using shRNA or decitabine prevents HOXB4 repression by Pu.1 or granulocyte-colony stimulating factor and maintains hematopoietic precursor self-renewal. In contrast, depletion of DNMT1 by decitabine 6 hours after the differentiation stimulus, that is, after repression of HOXB4 has occurred, augments differentiation. Therefore, DNMT1 is required for the early repression of stem cell genes, which occurs in response to a differentiation stimulus, providing a mechanistic explanation for the observation that decitabine can maintain or increase hematopoietic stem cell self-renewal in the presence of a differentiation stimulus. Using decitabine to deplete DNMT1 after this early repression phase does not impair progressive differentiation.


Assuntos
Azacitidina/análogos & derivados , Diferenciação Celular/genética , Regulação para Baixo/genética , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Azacitidina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Decitabina , Regulação para Baixo/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Transativadores/metabolismo
19.
Anat Rec (Hoboken) ; 292(3): 320-32, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19248164

RESUMO

Quantification of microvascular remodeling as a meaningful discovery tool requires mapping and measurement of site-specific changes within vascular trees and networks. Vessel density and other critical vascular parameters are often modulated by molecular regulators as determined by local vascular architecture. For example, enlargement of vessel diameter by vascular endothelial growth factor (VEGF) is restricted to specific generations of vessel branching (Parsons-Wingerter et al., Microvascular Research72: 91, 2006). The averaging of vessel diameter over many successively smaller generations is therefore not particularly useful. The newly automated, user-interactive software VESsel GENeration Analysis (VESGEN) quantifies major vessel parameters within two-dimensional (2D) vascular trees, networks, and tree-network composites. This report reviews application of VESGEN 2D to angiogenic and lymphangiogenic tissues that includes the human and murine retina, embryonic coronary vessels, and avian chorioallantoic membrane. Software output includes colorized image maps with quantification of local vessel diameter, fractal dimension, tortuosity, and avascular spacing. The density of parameters such as vessel area, length, number, and branch point are quantified according to site-specific generational branching within vascular trees. The sole user input requirement is a binary (black/white) vascular image. Future applications of VESGEN will include analysis of 3D vascular architecture and bioinformatic dimensions such as blood flow and receptor localization. Branching analysis by VESGEN has demonstrated that numerous regulators including VEGF(165), basic fibroblast growth factor, transforming growth factor beta-1, angiostatin and the clinical steroid triamcinolone acetonide induce 'fingerprint' or 'signature' changes in vascular patterning that provide unique readouts of dominant molecular signaling.


Assuntos
Linfangiogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Software , Interface Usuário-Computador , Animais , Vasos Coronários/fisiologia , Humanos , Camundongos , Codorniz , Retina/efeitos dos fármacos , Vasos Retinianos/fisiologia , Triancinolona Acetonida/farmacologia
20.
Proc Natl Acad Sci U S A ; 103(36): 13480-4, 2006 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16938854

RESUMO

Choroidal neovascularization (CNV), the advanced stage of age-related macular degeneration (AMD), accounts for >80% of vision loss in AMD. Carboxyethylpyrrole (CEP) protein modifications, uniquely generated from oxidation of docosahexaenoate-containing lipids, are more abundant in Bruch's membrane from AMD eyes. We tested the hypothesis that CEP protein adducts stimulate angiogenesis and possibly contribute to CNV in AMD. Human serum albumin (HSA) or acetyl-Gly-Lys-O-methyl ester (dipeptide) were chemically modified to yield CEP-modified HSA (CEP-HSA) or CEP-dipeptide. The in vivo angiogenic properties of CEP-HSA and CEP-dipeptide were demonstrated by using the chick chorioallantoic membrane and rat corneal micropocket assays. Low picomole amounts of CEP-HSA and CEP-dipeptide stimulated neovascularization. Monoclonal anti-CEP antibody neutralized limbal vessel growth stimulated by CEP-HSA, whereas anti-VEGF antibody was found to only partially neutralize vessel growth. Subretinal injections of CEP-modified mouse serum albumin exacerbated laser-induced CNV in mice. In vitro treatments of human retinal pigment epithelial cells with CEP-dipeptide or CEP-HSA did not induce increased VEGF secretion. Overall, these results suggest that CEP-induced angiogenesis utilizes VEGF-independent pathways and that anti-CEP therapeutic modalities might be of value in limiting CNV in AMD.


Assuntos
Neovascularização de Coroide/metabolismo , Degeneração Macular/metabolismo , Pirróis/química , Pirróis/farmacologia , Idoso de 80 Anos ou mais , Alantoide/irrigação sanguínea , Animais , Lâmina Basilar da Corioide/química , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Criança , Córion/irrigação sanguínea , Neovascularização de Coroide/etiologia , Relação Dose-Resposta a Droga , Humanos , Fotocoagulação a Laser , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neovascularização Fisiológica , Oxirredução , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/metabolismo , Pirróis/metabolismo , Ratos , Ratos Sprague-Dawley , Albumina Sérica/química , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA