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.
Artículo en Inglés | MEDLINE | ID: mdl-24695740

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Proteínas Co-Represoras/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas Co-Represoras/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Espectrometría de Masas en Tándem , Transactivadores/metabolismo , Células Tumorales Cultivadas
2.
Am J Pathol ; 180(4): 1702-14, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322296

RESUMEN

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.


Asunto(s)
Caveolina 1/deficiencia , Neovascularización Patológica/metabolismo , Óxido Nítrico Sintasa de Tipo III/fisiología , Animales , Caveolina 1/genética , Caveolina 1/fisiología , Células Cultivadas , Quimiotaxis/fisiología , Neovascularización de la Córnea/inducido químicamente , Neovascularización de la Córnea/metabolismo , Neovascularización de la Córnea/prevención & control , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Inhibidores Enzimáticos/farmacología , Pulmón/citología , Pulmón/efectos de los fármacos , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , NG-Nitroarginina Metil Éster/uso terapéutico , Trasplante de Neoplasias , Neovascularización Patológica/prevención & control , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
3.
Mol Vis ; 18: 1260-70, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22665973

RESUMEN

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.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Cristalino/metabolismo , Proteínas Represoras/genética , Transactivadores/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Cristalino/efectos de los fármacos , Cristalino/patología , Ratones , Ratones Noqueados , Vítreo Primario Hiperplásico Persistente/genética , Vítreo Primario Hiperplásico Persistente/metabolismo , Vítreo Primario Hiperplásico Persistente/patología , Unión Proteica , Proteínas Represoras/deficiencia , Eliminación de Secuencia , Inhibidores de Topoisomerasa I/administración & dosificación , Topotecan/administración & dosificación , Transactivadores/deficiencia , Factor A de Crecimiento Endotelial Vascular/deficiencia
4.
Am J Pathol ; 176(1): 496-503, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19948826

RESUMEN

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.


Asunto(s)
Córnea/irrigación sanguínea , Córnea/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Córnea/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Huésped Inmunocomprometido , Implantes Experimentales , Metaloproteinasa 2 de la Matriz/farmacología , Metaloproteinasa 9 de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Pruebas de Neutralización , Ratas , Factor A de Crecimiento Endotelial Vascular/farmacología
5.
Nat Med ; 9(4): 407-15, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12652295

RESUMEN

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.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfocinas/metabolismo , Neovascularización Patológica/prevención & control , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Inhibidores Tisulares de Metaloproteinasas/fisiología , División Celular/efectos de los fármacos , Línea Celular , Factores de Crecimiento Endotelial/farmacología , Endotelio/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/farmacología , Neuropilinas/metabolismo , Fosforilación , Proteínas Recombinantes/farmacología , Inhibidor Tisular de Metaloproteinasa-3 , Inhibidores Tisulares de Metaloproteinasas/genética , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
6.
Proc Natl Acad Sci U S A ; 105(50): 19898-903, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19057008

RESUMEN

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.


Asunto(s)
Aminoácidos Dicarboxílicos/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Oxígeno/toxicidad , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Retinopatía de la Prematuridad/prevención & control , Aerobiosis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Modelos Animales de Enfermedad , Eritropoyetina/biosíntesis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Recién Nacido , Riñón/metabolismo , Hígado/metabolismo , Ratones , Procolágeno-Prolina Dioxigenasa/metabolismo , Retina/metabolismo , Retinopatía de la Prematuridad/inducido químicamente , Retinopatía de la Prematuridad/enzimología , Factor A de Crecimiento Endotelial Vascular/biosíntesis
7.
J Clin Invest ; 128(10): 4260-4279, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30015632

RESUMEN

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.


Asunto(s)
Granulocitos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Monocitos/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Granulocitos/patología , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Monocitos/patología , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Proteínas Nucleares/genética , Nucleofosmina , Células THP-1 , Factores de Transcripción/genética
8.
Invest Ophthalmol Vis Sci ; 47(11): 4935-41, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17065510

RESUMEN

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.


Asunto(s)
Neovascularización de la Córnea/tratamiento farmacológico , Glucocorticoides/farmacología , Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Triamcinolona Acetonida/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Membrana Corioalantoides/irrigación sanguínea , Neovascularización de la Córnea/inducido químicamente , Neovascularización de la Córnea/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Interleucina-6/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Ratas , Ratas Endogámicas F344 , Factor de Transcripción STAT3/metabolismo , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología
9.
FASEB J ; 19(7): 819-21, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15857892

RESUMEN

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.


Asunto(s)
Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Fosforilcolina/análogos & derivados , Receptores Acoplados a Proteínas G/fisiología , Esfingosina/análogos & derivados , Animales , Anticuerpos/farmacología , División Celular/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Embrión de Pollo , Activación Enzimática , Expresión Génica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Lisofosfolípidos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Proteína Oncogénica v-akt/metabolismo , Fragmentos de Péptidos/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilcolina/farmacología , ARN Interferente Pequeño/farmacología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Esfingosina/farmacología , Venas Umbilicales , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Exp Biol Med (Maywood) ; 239(7): 853-861, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24879424

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-23469166

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Lámina Basal de la Coroides/irrigación sanguínea , Lámina Basal de la Coroides/efectos de los fármacos , Neovascularización Coroidal/prevención & control , Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidores de la Angiogénesis/síntesis química , Animales , Lámina Basal de la Coroides/lesiones , Células Cultivadas , Neovascularización Coroidal/etiología , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Endotelio Vascular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Coagulación con Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Péptidos/síntesis química , Fosforilación , Transducción de Señal/efectos de los fármacos , Técnicas de Síntesis en Fase Sólida , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Clin Cancer Res ; 19(4): 938-48, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23287564

RESUMEN

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.


Asunto(s)
Citidina Desaminasa/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Animales , Azacitidina/análogos & derivados , Citarabina/administración & dosificación , Citidina/administración & dosificación , Citidina/análogos & derivados , Citidina Desaminasa/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Decitabina , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Polimorfismo de Nucleótido Simple , Análisis de Supervivencia , Resultado del Tratamiento
14.
Oncotarget ; 3(10): 1137-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23087155

RESUMEN

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.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Citidina Desaminasa/metabolismo , Leucemia Mieloide/enzimología , Neoplasias Hepáticas/enzimología , Hígado/enzimología , Animales , Antimetabolitos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Azacitidina/uso terapéutico , Decitabina , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Hígado/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Tetrahidrouridina/uso terapéutico , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología
15.
Invest Ophthalmol Vis Sci ; 52(9): 6117-23, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21282576

RESUMEN

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.


Asunto(s)
Neovascularización Coroidal/fisiopatología , Neovascularización de la Córnea/fisiopatología , Modelos Animales de Enfermedad , Inhibidor Tisular de Metaloproteinasa-3/deficiencia , Animales , Neovascularización Coroidal/metabolismo , Neovascularización de la Córnea/metabolismo , Immunoblotting , Inmunoprecipitación , Coagulación con Láser , Metaloproteinasa 3 de la Matriz/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Oftalmoscopía , Fosforilación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
PLoS One ; 5(10): e13444, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20976146

RESUMEN

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.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Vasos Retinianos/efectos de los fármacos , Animales , Betacelulina , Barrera Hematorretinal , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Ratones Endogámicos C57BL
17.
Mol Cancer Ther ; 9(6): 1536-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20501800

RESUMEN

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.


Asunto(s)
Azacitidina/análogos & derivados , Diferenciación Celular/genética , Regulación hacia Abajo/genética , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas Represoras/metabolismo , Animales , Azacitidina/farmacología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Decitabina , Regulación hacia Abajo/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transactivadores/metabolismo
19.
Anat Rec (Hoboken) ; 292(3): 320-32, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19248164

RESUMEN

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.


Asunto(s)
Linfangiogénesis/fisiología , Neovascularización Fisiológica/fisiología , Programas Informáticos , Interfaz Usuario-Computador , Animales , Vasos Coronarios/fisiología , Humanos , Ratones , Codorniz , Retina/efectos de los fármacos , Vasos Retinianos/fisiología , Triamcinolona Acetonida/farmacología
20.
Proc Natl Acad Sci U S A ; 103(36): 13480-4, 2006 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16938854

RESUMEN

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.


Asunto(s)
Neovascularización Coroidal/metabolismo , Degeneración Macular/metabolismo , Pirroles/química , Pirroles/farmacología , Anciano de 80 o más Años , Alantoides/irrigación sanguínea , Animales , Lámina Basal de la Coroides/química , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Embrión de Pollo , Niño , Corion/irrigación sanguínea , Neovascularización Coroidal/etiología , Relación Dosis-Respuesta a Droga , Humanos , Coagulación con Láser , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Fisiológica , Oxidación-Reducción , Epitelio Pigmentado Ocular/citología , Epitelio Pigmentado Ocular/efectos de los fármacos , Epitelio Pigmentado Ocular/metabolismo , Pirroles/metabolismo , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/química , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA