Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 6(1): 447, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185301

RESUMEN

Efficacy of immune checkpoint inhibitors in cancers can be limited by CD8 T cell dysfunction or HLA-I down-regulation. Tumor control mechanisms independent of CD8/HLA-I axis would overcome these limitations. Here, we report potent CD4 T cell-mediated tumor regression and memory responses in humanized immune system (HIS) mice implanted with HT-29 colorectal tumors. The regressing tumors showed increased CD4 cytotoxic T lymphocyte (CTL) infiltration and enhanced tumor HLA-II expression compared to progressing tumors. The intratumoral CD4 T cell subset associated with tumor regression expressed multiple cytotoxic markers and exhibited clonal expansion. Notably, tumor control was abrogated by depletion of CD4 but not CD8 T cells. CD4 T cells derived from tumor-regressing mice exhibited HLA-II-dependent and tumor-specific killing ex vivo. Taken together, our study demonstrates a critical role of human CD4 CTLs in mediating tumor clearance independent of CD8 T cells and provides a platform to study human anti-tumor immunity in vivo.


Asunto(s)
Neoplasias , Linfocitos T Citotóxicos , Humanos , Ratones , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Neoplasias/metabolismo
2.
Mol Cancer Ther ; 18(11): 2051-2062, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31395688

RESUMEN

In the tumor microenvironment, multiple inhibitory checkpoint receptors can suppress T-cell function, thereby enabling tumor immune evasion. Blockade of one of these checkpoint receptors, PD-1, with therapeutic antibodies has produced positive clinical responses in various cancers; however, the efficacy of this approach can be further improved. Simultaneously targeting multiple inhibitory checkpoint receptors has emerged as a promising therapeutic strategy. Here, we report the development and characterization of REGN3767, a fully human IgG4 antibody targeting LAG-3, another inhibitory receptor on T cells. REGN3767 binds human and monkey LAG-3 with high affinity and specificity and blocks the interaction of LAG-3 with its ligand, MHC class II. In an engineered T-cell/antigen-presenting cell bioassay, REGN3767 alone, or in combination with cemiplimab (REGN2810, human anti-PD-1 antibody), blocked inhibitory signaling to T cells mediated by hLAG-3/MHCII in the presence of PD-1/PD-L1. To test the in vivo activity of REGN3767 alone or in combination with cemiplimab, we generated human PD-1xLAG-3 knockin mice, in which the extracellular domains of mouse Pdcd1 and Lag3 were replaced with their human counterparts. In these humanized mice, treatment with cemiplimab and REGN3767 showed increased efficacy in a mouse tumor model and enhanced the secretion of proinflammatory cytokines by tumor-specific T cells. The favorable pharmacokinetics and toxicology of REGN3767 in nonhuman primates, together with enhancement of antitumor efficacy of anti-PD-1 antibody in preclinical tumor models, support its clinical development.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antígenos CD/química , Antígenos CD/genética , Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/genética , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/metabolismo , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Técnicas de Sustitución del Gen , Haplorrinos , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Ratones , Neoplasias/genética , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína del Gen 3 de Activación de Linfocitos
3.
Mol Cancer Ther ; 16(5): 861-870, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28265006

RESUMEN

The Programmed Death-1 (PD-1) receptor delivers inhibitory checkpoint signals to activated T cells upon binding to its ligands PD-L1 and PD-L2 expressed on antigen-presenting cells and cancer cells, resulting in suppression of T-cell effector function and tumor immune evasion. Clinical antibodies blocking the interaction between PD-1 and PD-L1 restore the cytotoxic function of tumor antigen-specific T cells, yielding durable objective responses in multiple cancers. This report describes the preclinical characterization of REGN2810, a fully human hinge-stabilized IgG4(S228P) high-affinity anti-PD-1 antibody that potently blocks PD-1 interactions with PD-L1 and PD-L2. REGN2810 was characterized in a series of binding, blocking, and functional cell-based assays, and preclinical in vivo studies in mice and monkeys. In cell-based assays, REGN2810 reverses PD-1-dependent attenuation of T-cell receptor signaling in engineered T cells and enhances responses of human primary T cells. To test the in vivo activity of REGN2810, which does not cross-react with murine PD-1, knock-in mice were generated to express a hybrid protein containing the extracellular domain of human PD-1, and transmembrane and intracellular domains of mouse PD-1. In these mice, REGN2810 binds the humanized PD-1 receptor and inhibits growth of MC38 murine tumors. As REGN2810 binds to cynomolgus monkey PD-1 with high affinity, pharmacokinetic and toxicologic assessment of REGN2810 was performed in cynomolgus monkeys. High doses of REGN2810 were well tolerated, without adverse immune-related effects. These preclinical studies validate REGN2810 as a potent and promising candidate for cancer immunotherapy. Mol Cancer Ther; 16(5); 861-70. ©2017 AACR.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/genética , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Línea Celular Tumoral , Técnicas de Sustitución del Gen , Humanos , Inmunoterapia , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
4.
Cancer Res ; 76(8): 2327-39, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26921327

RESUMEN

Anti-VEGF therapies benefit several cancer types, but drug resistance that limits therapeutic response can emerge. We generated cell lines from anti-VEGF-resistant tumor xenografts to investigate the mechanisms by which resistance develops. Of all tumor cells tested, only A431 (A431-V) epidermoid carcinoma cells developed partial resistance to the VEGF inhibitor aflibercept. Compared with the parental tumors, A431-V tumors secreted greater amounts of IL6 and exhibited higher levels of phospho-STAT3. Notably, combined blockade of IL6 receptor (IL6R) and VEGF resulted in enhanced activity against A431-V tumors. Similarly, inhibition of IL6R enhanced the antitumor effects of aflibercept in DU145 prostate tumor cells that displays high endogenous IL6R activity. In addition, post hoc stratification of data obtained from a clinical trial investigating aflibercept efficacy in ovarian cancer showed poorer survival in patients with high levels of circulating IL6. These results suggest that the activation of the IL6/STAT3 pathway in tumor cells may provide a survival advantage during anti-VEGF treatment, suggesting its utility as a source of response biomarkers and as a therapeutic target to heighten efficacious results. Cancer Res; 76(8); 2327-39. ©2016 AACR.


Asunto(s)
Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Xenoinjertos , Humanos , Interleucina-6/antagonistas & inhibidores , Ratones , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/farmacología
5.
Int J Oncol ; 34(1): 79-87, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19082480

RESUMEN

Approval of the anti-vascular endothelial growth factor (VEGF) antibody bevacizumab by the FDA in 2004 reflected the success of this vascular targeting strategy in extending survival in patients with advanced cancers. However, consistent with previous reports that experimental tumors can grow or recur during VEGF blockade, it has become clear that many patients treated with VEGF inhibitors will ultimately develop progressive disease. Previous studies have shown that disruption of VEGF signaling in tumors induces remodeling in surviving vessels, and link increased expression of angiopoietin-1 (Ang-1) with this process. However, overexpression of Ang-1 in different tumors has yielded divergent results, restricting angiogenesis in some systems while promoting it in others. These data raise the possibility that effects of Ang-1/Tie-2 may be context-dependent. Expression of an Ang-1 construct (Ang1*) did not significantly change tumor growth in our model prior to treatment, although vessels exhibited changes consistent with increased Tie-2 signaling. During inhibition of VEGF, however, both overexpression of Ang1* and administration of an engineered Ang-1 agonist (Bow-Ang1) strikingly protected tumors and vasculature from regression. In this context, Ang-1/Tie-2 activation limited tumor hypoxia, increased vessel caliber, and promoted recruitment of mural cells. Thus, these studies support a model in which activation of Tie-2 is important for tumor and vessel survival when VEGF-dependent vasculature is stressed. Understanding such mechanisms of adaptation to this validated form of therapy may be important in designing regimens that make the best use of this approach.


Asunto(s)
Angiopoyetina 1/genética , Regulación de la Expresión Génica/fisiología , Neoplasias Renales/irrigación sanguínea , Neovascularización Patológica/patología , Receptor TIE-2/genética , Sarcoma de Ewing/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoprecipitación , Neoplasias Renales/patología , Ratones , Ratones Desnudos , Fosforilación , Reacción en Cadena de la Polimerasa , Sarcoma de Ewing/patología , Transfección , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Kidney Int ; 74(3): 300-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18480750

RESUMEN

The loss of interstitial capillaries is a feature of several experimental models of renal disease and this contributes to secondary kidney injury. Angiopoietin-1 is a secreted growth factor which binds to Tie-2 present on endothelia to enhance cell survival thereby stabilizing capillary architecture in-vitro. Previous studies showed that angiopoietin-1 prevented renal capillary and interstitial lesions following experimental ureteric obstruction. We tested here the effect of angiopoietin-1 treatment on capillary loss and associated tubulointerstitial damage known to follow recovery from folic acid-induced tubular necrosis and acute renal injury. We found that delivery of angiopoietin-1 by adenoviral vectors stabilized peritubular capillaries in folic acid nephropathy but this was accompanied by profibrotic and inflammatory effects. These results suggest that the use of endothelial growth factor therapy for kidney disease may have varying outcomes that depend on the disease model tested.


Asunto(s)
Angiopoyetina 1/efectos adversos , Fibrosis/inducido químicamente , Inflamación/inducido químicamente , Necrosis Tubular Aguda/tratamiento farmacológico , Adenoviridae/genética , Angiopoyetina 1/administración & dosificación , Angiopoyetina 1/uso terapéutico , Animales , Modelos Animales de Enfermedad , Ácido Fólico/efectos adversos , Vectores Genéticos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Necrosis Tubular Aguda/inducido químicamente , Necrosis Tubular Aguda/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Circulación Renal/efectos de los fármacos
7.
Proc Natl Acad Sci U S A ; 103(42): 15491-6, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17030814

RESUMEN

Angiopoietin (Ang)-2, a context-dependent agonist/antagonist for the vascular-specific Tie2 receptor, is highly expressed by endothelial cells at sites of normal and pathologic angiogenesis. One prevailing model suggests that in these settings, Ang-2 acts as an autocrine Tie2 blocker, inhibiting the stabilizing influence of the Tie2 activator Ang-1, thereby promoting vascular remodeling. However, the effects of endogenous Ang-2 on cells that are actively producing it have not been studied in detail. Here, we demonstrate that Ang-2 expression is rapidly induced in endothelial cells by the transcription factor FOXO1 after inhibition of the phosphatidylinositol 3-kinase/Akt pathway. We employ RNAi and blocking antibodies to show that in this setting, Ang-2 unexpectedly functions as a Tie2 agonist, bolstering Akt activity so as to provide negative feedback on FOXO1-regulated transcription and apoptosis. In addition, we show that Ang-2, like Ang-1, activates Tie2/Akt signaling in vivo, thereby inhibiting the expression of FOXO1 target genes. Consistent with a role for Ang-2 as a Tie2 activator, we demonstrate that Ang-2 inhibits vascular leak. Our data suggests a model in which Ang-2 expression is induced in stressed endothelial cells, where it acts as an autocrine Tie2 agonist and protective factor.


Asunto(s)
Angiopoyetina 2/metabolismo , Comunicación Autocrina , Células Endoteliales/fisiología , Estrés Oxidativo , Androstadienos/metabolismo , Angiopoyetina 2/genética , Animales , Apoptosis/fisiología , Células Cultivadas , Células Endoteliales/citología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Activación Transcripcional , Wortmanina
8.
J Invest Dermatol ; 126(10): 2316-22, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16741507

RESUMEN

Hemangioma of infancy is the most common neoplasm of childhood. While hemangiomas are classic examples of angiogenesis, the angiogenic factors responsible for hemangiomas are not fully understood. Previously, we demonstrated that malignant endothelial tumors arise in the setting of autocrine loops involving vascular endothelial growth factor (VEGF) and its major mitogenic receptor vascular endothelial growth factor receptor 2. Hemangiomas of infancy differ from malignant endothelial tumors in that they usually regress, or can be induced to regress by pharmacologic means, suggesting that angiogenesis in hemangiomas differs fundamentally from that of malignant endothelial tumors. Here, we demonstrate constitutive activation of the endothelial tie-2 receptor in human hemangioma of infancy and, using a murine model of hemangioma, bEnd.3 cells; we show that bEnd.3 hemangiomas produce both angiopoietin-2 (ang-2) and its receptor, tie-2, in vivo. We also demonstrate that inhibition of tie-2 signaling with a soluble tie-2 receptor decreases bEnd.3 hemangioma growth in vivo. The efficacy of tie-2 blockade suggests that either tie-2 activation or ang-2 may be required for in vivo growth. To address this issue, we used tie-2-deficient bEnd.3 hemangioma cells, which, surprisingly, were fully proficient in in vivo growth. Previous studies from our laboratory and others have implicated reactive oxygen-generating nox enzymes in the angiogenic switch, so we examined the effect of nox inhibitors on ang-2 production in vitro and on bEnd.3 tumor growth in vivo. We then inhibited ang-2 production pharmacologically using novel inhibitors of nox enzymes and found that this treatment nearly abolished bEnd.3 hemangioma growth in vivo. Signal-transduction blockade targeting ang-2 production may be useful in the treatment of human hemangiomas in vivo.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Hemangioma/tratamiento farmacológico , Angiopoyetina 2/biosíntesis , Angiopoyetina 2/genética , Animales , Violeta de Genciana/farmacología , Hemangioma/etiología , Hemangioma/patología , Humanos , Ratones , NADPH Oxidasas/antagonistas & inhibidores , Compuestos de Amonio Cuaternario/farmacología , ARN Mensajero/análisis , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/fisiología , Transducción de Señal
10.
Genes Dev ; 18(9): 1060-71, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15132996

RESUMEN

Despite genetic evidence establishing angiopoietin-1 (Ang-1) as an essential regulator of vascular development, the molecular mechanisms underlying Ang-1 function are almost completely uncharacterized. In this report, we demonstrate that Ang-1, via Akt activation, is a potent inhibitor of the forkhead transcription factor FKHR (FOXO1), identifying for the first time a nuclear signaling pathway through which Ang-1 modulates gene expression. We use microarray analysis to show that FKHR, whose function in endothelial cells has not previously been elucidated, regulates many genes associated with vascular destabilization and remodeling (including angiopoietin-2, an Ang-1 antagonist) and endothelial cell apoptosis (e.g., survivin, TRAIL). Ang-1 inhibits FKHR-mediated changes in gene expression and FKHR-induced apoptosis. Analysis of gene expression changes induced by an activated version of Akt confirms that FKHR is a major target through which Akt regulates transcription in endothelial cells. We use RNA interference to demonstrate that FKHR is required for the expression of genes (including Ang-2) that have important vascular functions. Our data suggest a novel, tissue-specific role for the Akt/FKHR pathway in the vasculature and suggest a mechanistic basis for the previously described actions of Ang-1 as a regulator of endothelial cell survival and blood vessel stability.


Asunto(s)
Angiopoyetina 1/farmacología , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Proteínas Serina-Treonina Quinasas , Factores de Transcripción/fisiología , Angiopoyetina 1/fisiología , Animales , Apoptosis/efectos de los fármacos , Secuencia de Bases , Bovinos , Células Cultivadas , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Endotelio Vascular/citología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/farmacología , Transducción de Señal , Factores de Transcripción/genética
11.
Nat Struct Biol ; 10(1): 38-44, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12469114

RESUMEN

Angiopoietins are a recently discovered family of angiogenic factors that interact with the endothelial receptor tyrosine kinase Tie2, either as agonists (angiopoietin-1) or as context-dependent agonists/antagonists (angiopoietin-2). Here we show that angiopoietin-1 has a modular structure unlike any previously characterized growth factor. This modular structure consists of a receptor-binding domain, a dimerization motif and a superclustering motif that forms variable-sized multimers. Genetic engineering of precise multimers of the receptor-binding domain of angiopoietin-1, using surrogate multimerization motifs, reveals that tetramers are the minimal size required for activating endothelial Tie2 receptors. In contrast, engineered dimers can antagonize endothelial Tie2 receptors. Surprisingly, angiopoietin-2 has a modular structure and multimerization state similar to that of angiopoietin-1, and its antagonist activity seems to be a subtle property encoded in its receptor-binding domain.


Asunto(s)
Angiopoyetinas/química , Angiopoyetinas/metabolismo , Receptor TIE-2/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Angiopoyetina 1/química , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/química , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Angiopoyetinas/genética , Animales , Células CHO , Cricetinae , Cricetulus , Dimerización , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Modelos Moleculares , Fosforilación , Unión Proteica , Ingeniería de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
12.
Proc Natl Acad Sci U S A ; 99(17): 11393-8, 2002 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12177445

RESUMEN

Vascular endothelial growth factor (VEGF) plays a critical role during normal embryonic angiogenesis and also in the pathological angiogenesis that occurs in a number of diseases, including cancer. Initial attempts to block VEGF by using a humanized monoclonal antibody are beginning to show promise in human cancer patients, underscoring the importance of optimizing VEGF blockade. Previous studies have found that one of the most effective ways to block the VEGF-signaling pathway is to prevent VEGF from binding to its normal receptors by administering decoy-soluble receptors. The highest-affinity VEGF blocker described to date is a soluble decoy receptor created by fusing the first three Ig domains of VEGF receptor 1 to an Ig constant region; however, this fusion protein has very poor in vivo pharmacokinetic properties. By determining the requirements to maintain high affinity while extending in vivo half life, we were able to engineer a very potent high-affinity VEGF blocker that has markedly enhanced pharmacokinetic properties. This VEGF-Trap effectively suppresses tumor growth and vascularization in vivo, resulting in stunted and almost completely avascular tumors. VEGF-Trap-mediated blockade may be superior to that achieved by other agents, such as monoclonal antibodies targeted against the VEGF receptor.


Asunto(s)
Antineoplásicos/farmacología , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Factores de Crecimiento Endotelial/inmunología , Endotelio Vascular/fisiología , Linfocinas/antagonistas & inhibidores , Linfocinas/inmunología , Melanoma Experimental/tratamiento farmacológico , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Células 3T3 , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/tratamiento farmacológico , División Celular , Diseño de Fármacos , Factores de Crecimiento Endotelial/farmacología , Matriz Extracelular/fisiología , Humanos , Regiones Constantes de Inmunoglobulina/genética , Inmunoglobulina G/genética , Linfocinas/farmacología , Melanoma Experimental/irrigación sanguínea , Ratones , Ratones Endogámicos BALB C , Fosforilación , Ingeniería de Proteínas , Rabdomiosarcoma/irrigación sanguínea , Rabdomiosarcoma/tratamiento farmacológico , Venas Umbilicales , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Am J Pathol ; 160(5): 1683-93, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12000720

RESUMEN

Diabetic retinopathy remains a leading cause of irreversible blindness. A critical early pathology in the disease is the adhesion of leukocytes to the retinal vasculature, a process that occurs, in part, via intercellular adhesion molecule-1. Once leukocyte adhesion occurs, endothelial cell injury ensues, as does blood-retinal barrier breakdown. Here we show that angiopoietin-1 can prevent and reverse these diabetic retinal vascular changes in both new and established diabetes. Angiopoietin-1, when given intravitreally to newly diabetic rats, normalized retinal vascular endothelial growth factor (VEGF) and intercellular adhesion molecule-1 mRNA and protein levels, leading to reductions in leukocyte adhesion, endothelial cell injury, and blood-retinal barrier breakdown. When an adenovirus coding for angiopoietin-1 was given systemically to mice with established diabetes, it similarly inhibited leukocyte adhesion and endothelial cell injury and blood-retinal barrier breakdown. These changes coincided with reductions in retinal eNOS, nitric oxide, Akt (protein kinase B), and MAP kinase activity, known mediators of VEGF bioactivity and leukocyte adhesion. When endogenous VEGF bioactivity was inhibited with a soluble Flt-1/Fc chimera, retinal Akt kinase activity was significantly reduced in vivo. Taken together, these data document new vascular and anti-inflammatory bioactivities for angiopoietin-1 and identify it as the first naturally occurring protein that directly protects the retinal vasculature in diabetes.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Glicoproteínas de Membrana/uso terapéutico , Proteínas Serina-Treonina Quinasas , Angiopoyetina 1 , Animales , Barrera Hematorretinal/efectos de los fármacos , Bovinos , Adhesión Celular/efectos de los fármacos , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Activación Enzimática/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/citología , Leucocitos/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Masculino , Glicoproteínas de Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...