Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Invest Ophthalmol Vis Sci ; 55(4): 2130-9, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24595385

RESUMEN

PURPOSE: Uveal melanoma (UM) tumors require large doses of radiation therapy (RT) to achieve tumor ablation, which frequently results in damage to adjacent normal tissues, leading to vision-threatening complications. Approximately 50% of UM patients present with activating somatic mutations in the gene encoding for G protein αq-subunit (GNAQ), which lead to constitutive activation of downstream pathways, including protein kinase C (PKC). In this study, we investigated the impact of small-molecule PKC inhibitors bisindolylmaleimide I (BIM) and sotrastaurin (AEB071), combined with ionizing radiation (IR), on survival in melanoma cell lines. METHODS: Cellular radiosensitivity was determined by using a combination of proliferation, viability, and clonogenic assays. Cell-cycle effects were measured by flow cytometry. Transcriptomic and proteomic profiling were performed by quantitative real-time PCR, reverse-phase protein array analysis, and immunofluorescence. RESULTS: We found that the PKC inhibitors combined with IR significantly decreased the viability, proliferation, and clonogenic potential of GNAQ(mt), but not GNAQ(wt)/BRAF(mt) cells, compared with IR alone. Combined treatment increased the antiproliferative and proapoptotic effects of IR in GNAQ(mt) cells through delayed DNA-damage resolution and enhanced induction of proteins involved in cell-cycle arrest, cell-growth arrest, and apoptosis. CONCLUSIONS: Our preclinical results suggest that combined modality treatment may allow for reductions in the total RT dose and/or fraction size, which may lead to better functional organ preservation in the treatment of primary GNAQ(mt) UM. These findings suggest future clinical trials combining PKC inhibitors with RT in GNAQ(mt) UM warrant consideration.


Asunto(s)
ADN de Neoplasias/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Melanoma/enzimología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Úvea/enzimología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Terapia Combinada , Citometría de Flujo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/efectos de la radiación , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Humanos , Melanoma/genética , Melanoma/terapia , Radiación Ionizante , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/terapia
2.
Invest Ophthalmol Vis Sci ; 52(10): 7248-55, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21828154

RESUMEN

PURPOSE: Inhibitors of B-Raf and MEK kinases hold promise for the management of cutaneous melanomas harboring BRAF mutations. BRAF mutations are rare in uveal melanomas (UMs), but somatic mutations in the G protein α subunits Gαq and Gα11 (encoded by GNAQ and GNA11, respectively) occur in a mutually exclusive pattern in ∼80% of UMs. The impact of B-Raf and MEK inhibitors on Gα-mutant UMs remains unknown. METHODS: The impact of the B-Raf inhibitor PLX4720, the MEK inhibitor AZD6244, and the Akt inhibitor MK2206 on UM cell lines was assessed with the use of cell viability, proliferation, and apoptosis assays and immunoblot analysis. RESULTS: BRAF-mutant UM cells were sensitive to both PLX4720 and AZD6244, undergoing cell cycle arrest but not apoptosis. UM cells with a Gα-protein mutation (GNAQ or GNA11) were mildly sensitive to AZD6244 but completely resistant to PLX4720. In fact, PLX4720 paradoxically increased ERK phosphorylation in Gα-mutant UM cells. The combination of AZD6244 with PLX4720 had synergistic anticancer activity in BRAF-mutant cells but not in Gα-mutant cells. The Akt inhibitor MK2206 sensitized BRAF-mutant cells to both PLX4720 and AZD6244 and sensitized Gα-mutant cells to AZD6244 but did not overcome the resistance of the Gα-mutant cells to PLX4720. CONCLUSIONS: The response of UM cells to inhibition of B-Raf, MEK, and Akt depends on their genotype. Future use of such targeted therapies in clinical trials of UM patients will require careful design and patient selection based on genotype to provide personalized and effective therapy.


Asunto(s)
Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Neoplasias de la Úvea/genética , Apoptosis/efectos de los fármacos , Bencimidazoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Genotipo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Immunoblotting , Etiquetado Corte-Fin in Situ , Indoles/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Melanoma/patología , Medicina de Precisión , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sulfonamidas/farmacología , Neoplasias de la Úvea/patología
3.
Mol Vis ; 15: 1418-28, 2009 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-19641635

RESUMEN

PURPOSE: The pathophysiology of diabetic retinopathy involves leukocyte adhesion to retinal vasculature, early blood-retinal barrier breakdown, capillary nonperfusion, and endothelial cell death. We investigated the involvement of tumor necrosis factor alpha (TNF-alpha) in diabetes-related histopathological changes in two relevant rodent models. METHODS: In short-term studies, Long-Evans rats with streptozotocin-induced diabetes were treated with or without the TNF-alpha inhibitor, etanercept. For long-term studies, tumor necrosis factor receptor I (TNF-RI)-deficient mice and TNF-RII-deficient mice, as well as C57/Bl6 wild-type mice, were fed 30% galactose for up to 20 months. The retinal histopathological alterations of hypergalactosemia were analyzed in trypsin digest preparations. Endothelial cell injury and apoptosis in rat retinas were evaluated by propidium iodide, TUNEL, CytoDeath staining, and DNA fragmentation ELISA. Caspase 3 and 8 activity was evaluated by immunoblotting and quantitative enzymatic activity assay. RESULTS: Etanercept suppressed caspase activation, retinal cell injury, and apoptosis in short-term diabetic rats. Pericyte and endothelial cell loss were also reduced in long-term hypergalactosemic mice. Long-term studies demonstrated that pericyte loss and endothelial cell loss were reduced in comparison to wild-type diabetic controls. CONCLUSIONS: Our study identifies an important role for TNF-alpha in the pathogenesis of signature diabetic retinopathy pathologies and demonstrates that etanercept can inhibit retinal cell death and long-term complication of diabetes. Taken together, our results suggest that etanercept could prove beneficial in preventing both early and late vascular diabetic complications.


Asunto(s)
Apoptosis , Retinopatía Diabética/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Capilares/efectos de los fármacos , Capilares/patología , Caspasa 3/metabolismo , Inhibidores de Caspasas , Fragmentación del ADN/efectos de los fármacos , Retinopatía Diabética/enzimología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Activación Enzimática/efectos de los fármacos , Etanercept , Conducta Alimentaria/efectos de los fármacos , Galactosa/administración & dosificación , Galactosa/farmacología , Galactosemias/patología , Inmunoglobulina G/farmacología , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratas , Ratas Long-Evans , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Retina/efectos de los fármacos , Retina/enzimología , Retina/patología , Factores de Tiempo
4.
Invest Ophthalmol Vis Sci ; 50(10): 4898-904, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19553613

RESUMEN

PURPOSE: The pathophysiology of diabetic retinopathy is mediated by leukocyte adhesion to the vascular endothelium of the diabetic retina, which results in endothelial injury, blood-retina barrier breakdown, and capillary nonperfusion. Leukocyte adhesion is triggered by the interaction of vascular endothelium adhesion molecules, such as ICAM-1, with leukocyte integrins, such as CD18. Inhibition of ICAM-1/CD18 signaling suppresses but does not completely abolish the cardinal manifestations of diabetic retinopathy, suggesting a role for additional adhesion molecules. Integrin alpha 4 (CD49d), in complex with integrin beta1, forms very late antigen-4 (VLA-4), which interacts with vascular cell adhesion molecule-1. The authors have now studied the role of integrin alpha 4/CD49d in the pathogenesis of diabetic retinopathy. METHODS: Diabetes mellitus was induced in Long Evans rats with streptozotocin, and an anti-alpha 4 integrin/CD49d neutralizing antibody was injected 5 and 10 days later. Two weeks after streptozotocin administration, vascular leakage was quantified with the Evans Blue technique. Leukostasis was measured with a static adhesion assay ex vivo and the FITC-lectin perfusion method in vivo. Retinal VEGF and TNF-alpha levels and NF-kappaB activity were measured by ELISA. RESULTS: Blockade of alpha 4 integrin/CD49d attenuated the diabetes-induced upregulation of NF-kappaB activation, VEGF, and TNF-alpha protein levels and reduced significantly diabetes-induced leukocyte adhesion and vascular leakage. CONCLUSIONS: These data identify alpha 4 integrin/CD49d as a mediator of leukocyte adhesion and the resultant early signature abnormalities of diabetic retinopathy. Inhibition of this signaling pathway may hold promise for clinical activity in patients with diabetes.


Asunto(s)
Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Integrina alfa4/fisiología , Animales , Anticuerpos Bloqueadores , Barrera Hematorretinal , Permeabilidad Capilar , Adhesión Celular , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Leucocitos/fisiología , Leucostasis/etiología , Leucostasis/metabolismo , Masculino , FN-kappa B/metabolismo , Ratas , Ratas Long-Evans , Vasos Retinianos/citología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Invest Ophthalmol Vis Sci ; 50(9): 4072-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19387079

RESUMEN

PURPOSE: To characterize the molecular sequelae induced in retinoblastoma (Rb) cells by histone deacetylase inhibitors (HDACIs). Hydroxamic acid-based HDACIs such as vorinostat (suberoylanilide hydroxamic acid) induce the differentiation and apoptosis of transformed cells. Vorinostat has demonstrated significant anticancer activity against hematologic and solid tumors at doses well tolerated by patients and has been approved for the treatment of patients with cutaneous T-cell lymphoma. METHODS: The authors evaluated the effects of the HDACIs vorinostat and m-carboxycinnamic acid bis-hydroxamide on the Rb cell lines Y79 and WERI-Rb1 with the use of the MTT assay, BrdU incorporation assay, flow cytometry, immunoblotting, gene-expression profiling, quantitative RT-PCR, and NF-kappaB DNA-binding assay. RESULTS: Both HDACIs were effective against both Rb cell lines, inducing growth arrest and apoptosis in vitro. Vorinostat increased p53 expression and activated caspases -8, -9 and -3, whereas caspase inhibition abrogated vorinostat-induced apoptosis. Vorinostat downregulated baseline NF-kappaB activity and potentiated the activity of the DNA-damaging chemotherapeutic doxorubicin. Gene expression profiling and qRT-PCR demonstrated that vorinostat modulated the mRNA levels of genes important for signal transduction, cell cycle, cellular metabolism, stress response, apoptosis, extracellular matrix synthesis, and cell differentiation. Notably, several transcripts involved in the ephrin and Notch signaling pathways were upregulated. CONCLUSIONS: HDACIs, such as vorinostat, induce caspase-dependent apoptosis in Rb cells, downregulate baseline NF-kappaB activity, and potentiate the effectiveness of conventional chemotherapy. The finding that vorinostat augments the effectiveness of doxorubicin provides a rationale for future clinical studies looking at the use of vorinostat in combination with conventional chemotherapy in Rb.


Asunto(s)
Apoptosis , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Neoplasias de la Retina/patología , Retinoblastoma/patología , Acetilación , Bromodesoxiuridina/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cinamatos/farmacología , Doxorrubicina/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Immunoblotting , FN-kappa B/metabolismo , Proteínas de Neoplasias/genética , Neoplasias de la Retina/enzimología , Retinoblastoma/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo , Vorinostat
6.
Invest Ophthalmol Vis Sci ; 48(10): 4706-19, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17898295

RESUMEN

PURPOSE: To evaluate the potential of proteasome inhibitors, a novel class of antitumor agents, for the treatment of retinoblastoma. The proteasome inhibitor bortezomib (PS-341, Velcade; Millennium Pharmaceuticals, Cambridge, MA), approved by the US Food and Drug Administration for the treatment of multiple myeloma, is being studied for the treatment of several other malignancies. Among other effects, it inactivates the transcription factor nuclear factor-kappaB (NF-kappaB) by blocking the degradation of its inhibitor, IkappaB. NF-kappaB, which is constitutively active in human retinoblastoma cells and promotes their survival, represents a therapeutic target for patients with this malignancy. METHODS: The authors evaluated the effect of bortezomib on the retinoblastoma cell lines Y79 and WERI-Rb1 in vitro using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry with propidium iodide, gene expression profiling, RT-PCR, and immunoblotting. RESULTS: Bortezomib induced caspase-dependent apoptosis in both retinoblastoma cell lines at clinically achievable concentrations. Bortezomib upregulated heat-shock proteins, other stress-response proteins, proapoptotic molecules, cell-cycle regulators, transcription factors, cytokines, and several proteasome subunits and solute carrier proteins, whereas it downregulated antiapoptotic and adhesion molecules. Bortezomib also induced cleavage of caspases, Bid and poly(ADP-ribose) polymerase (PARP), and sensitized retinoblastoma cells to doxorubicin. CONCLUSIONS: Bortezomib induces a stress response and triggers caspase-dependent apoptosis in human retinoblastoma cells at clinically achievable concentrations. This study provides insight into the molecular mechanism(s) of the antitumor activity of bortezomib and a basis for future preclinical studies leading to clinical trials of bortezomib, alone or in combination with conventional chemotherapy, to improve patient outcomes in retinoblastoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Neoplasias de la Retina/patología , Retinoblastoma/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Bortezomib , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Formazáns , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Indicadores y Reactivos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Retina/genética , Neoplasias de la Retina/metabolismo , Retinoblastoma/genética , Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio , Células Tumorales Cultivadas , Regulación hacia Arriba
7.
J Clin Endocrinol Metab ; 92(12): 4845-52, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17848408

RESUMEN

CONTEXT: The Bcl-2 family of proteins regulates apoptosis in various models and may represent a promising therapeutic target in human malignancies. OBJECTIVE/METHODS: We evaluated the sensitivity of thyroid carcinoma cell lines (two papillary, one follicular, two anaplastic, three medullary) in vitro to BH3I-1 and BH3I-2', two cell-permeable inhibitors of the Bcl-2 homology (BH)-3 domain-mediated interaction between proapoptotic and antiapoptotic Bcl-2 family members. The thyroid carcinoma cell line FRO was stably transfected with cDNA for Bcl-2 or constitutively active Akt and evaluated for sensitivity to BH3-domain inhibition. RESULTS: BH3-domain inhibition disrupted the mitochondrial membrane potential in thyroid carcinoma cells, induced caspase-dependent apoptosis, and potently sensitized them to sublethal concentrations of doxorubicin and the proteasome inhibitor bortezomib (Velcade). Overexpression of constitutively active Akt suppressed BH3I-1-induced cell death. Bcl-2-overexpressing FRO cells were more resistant to conventional chemotherapeutic agents (such as doxorubicin) but significantly more sensitive to BH3I-1 than control cells and were found to overexpress caspase-9, caspase-8, Bmf, Bok, and Bik transcripts and express less A1, BRaf, and FLIP transcripts. CONCLUSIONS: Bcl-2 expression protects thyroid carcinomas against chemotherapy-induced apoptosis. Nevertheless, overexpression of Bcl-2 may result in "oncogene addiction" of the cancer cell, which can be exploited by using BH3-domain inhibitors alone or in combination with other agents, including conventional chemotherapeutics (such as doxorubicin) or novel targeted therapies (such as the proteasome inhibitor bortezomib), for the treatment of aggressive thyroid cancer, including the medullary and anaplastic types.


Asunto(s)
Carcinoma Medular/genética , Carcinoma/genética , Genes bcl-2/genética , Neoplasias de la Tiroides/genética , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Western Blotting , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Colorimetría , Doxorrubicina/farmacología , Humanos , Potenciales de la Membrana/fisiología , Proteína Oncogénica v-akt/genética , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Sales de Tetrazolio , Tiazoles , Transcripción Genética
8.
FASEB J ; 21(9): 2113-23, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17400913

RESUMEN

Heat shock protein (Hsp) 90 inhibitors, such as 17-allylamino-17-demethoxy-geldanamycin (17-AAG), constitute promising novel therapeutic agents. We investigated the anti-inflammatory activity of 17-AAG in endotoxin-induced uveitis (EIU) in rats. After the induction of EIU with a footpad injection of lipopolysaccharide (LPS), female Lewis rats received a single intraperitoneal. (i.p.) injection of 17-AAG or vehicle. Twenty-four hours later, the retinas were extracted and assayed for leukocyte adhesion; blood-retinal barrier breakdown; VEGF, TNF-alpha, IL-1beta, and CD14 protein levels; NF-kappaB and HIF-1alpha activity; hsp90 and 70 levels and expression and phosphorylation of the tight junction proteins ZO-1 and occludin. 17-AAG treatment significantly suppressed the LPS-induced increase in retinal leukocyte adhesion; vascular leakage; NF-kappaB, HIF-1alpha, p38, and PI-3K activity; and VEGF, TNF-alpha, and IL-1beta levels. 17-AAG also suppressed phosphorylation of ZO-1 and occludin by inhibiting their association with p38 and PI-3K. Although 17-AAG treatment did not reduce the LPS-induced increase in total CD14 levels in leukocytes, it significantly decreased membrane CD14 levels. These data suggest that Hsp90 inhibition suppresses several cardinal manifestations of endotoxin-induced uveitis in the rat. 17-AAG has demonstrated a favorable safety profile in clinical trials in cancer patients and represents a promising therapeutic agent for the treatment of inflammatory eye diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Benzoquinonas/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/uso terapéutico , Uveítis Anterior/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/farmacología , Toxinas Bacterianas/toxicidad , Barrera Hematorretinal/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Membrana Celular/química , Evaluación Preclínica de Medicamentos , Endotoxinas/toxicidad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-1beta/sangre , Leucocitos/química , Leucostasis/etiología , Leucostasis/prevención & control , Receptores de Lipopolisacáridos/sangre , Masculino , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Ocludina , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Long-Evans , Vasculitis Retiniana/inducido químicamente , Vasculitis Retiniana/prevención & control , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Uveítis Anterior/inducido químicamente , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/sangre , Proteína de la Zonula Occludens-1 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Mol Cancer Ther ; 6(3): 1070-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17363500

RESUMEN

B-Raf is an important mediator of cell proliferation and survival signals transduced via the Ras-Raf-MEK-ERK cascade. BRAF mutations have been detected in several tumors, including papillary thyroid carcinoma, but the precise role of B-Raf as a therapeutic target for thyroid carcinoma is still under investigation. We analyzed a panel of 93 specimens and 14 thyroid carcinoma cell lines for the presence of BRAF mutations and activation of the mitogen-activated protein/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. We also compared the effect of a B-Raf small inhibitory RNA construct and the B-Raf kinase inhibitor AAL881 on both B-Raf wild-type and mutant thyroid carcinoma cell lines. We found a high prevalence of the T1799A (V600E) mutation in papillary and anaplastic carcinoma specimens and cell lines. There was no difference in patient age, B-Raf expression, Ki67 immunostaining, or clinical stage at presentation between wild-type and BRAF(V600E) specimens. Immunodetection of phosphorylated and total forms of MEK and ERK revealed no difference in their phosphorylation between wild-type and BRAF(V600E) patient specimens or cell lines. Furthermore, a small inhibitory RNA construct targeting the expression of both wild-type B-Raf and B-Raf(V600E) induced a comparable reduction of viability in both wild-type and BRAF(V600E) mutant cancer cells. Interestingly, AAL881 inhibited MEK and ERK phosphorylation and induced apoptosis preferentially in BRAF(V600E)-harboring cells than wild-type ones, possibly because of better inhibitory activity against B-Raf(V600E). We conclude that B-Raf is important for the pathophysiology of thyroid carcinomas irrespective of mutational status. Small molecule inhibitors that selectively target B-Raf(V600E) may provide clinical benefit for patients with thyroid cancer.


Asunto(s)
Adenocarcinoma Folicular/genética , Carcinoma Papilar/genética , Isoquinolinas/farmacología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/genética , Adenocarcinoma Folicular/patología , Adenocarcinoma Folicular/prevención & control , Adulto , Anciano , Apoptosis/efectos de los fármacos , Carcinoma/genética , Carcinoma/patología , Carcinoma/prevención & control , Carcinoma Papilar/patología , Carcinoma Papilar/prevención & control , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/prevención & control , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/prevención & control , Células Tumorales Cultivadas/efectos de los fármacos
10.
Graefes Arch Clin Exp Ophthalmol ; 245(2): 258-66, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16741710

RESUMEN

BACKGROUND: The benzoquinoid antibiotic 17-allylaminogeldanamycin (17-AAG) inhibits the Ras/Raf/MEK and PI3-Kinase signaling pathways and down-regulates vascular endothelial factor expression. Here we use a mouse model of oxygen-induced retinopathy to investigate the effect of 17-AAG on retinal neovascularization and vascular recovery. MATERIAL AND METHODS: C57BL/6 mice were exposed to 75% oxygen from postnatal day 7 (P7) to P12 and recovered in room air thereafter. Beginning with P12 mice were treated for 5 days by daily IP injection of 17-AAG (12.5 mg/kg body weight) micro dispersed in an emulsion of 4% Lipoid EPC, 5% sucrose, and 0.9% NaCl or Wortmannin (100 microg/kg body weight). On P17, the retinal vascular and avascular area, neovascular blood vessel tufts, and main vessel tortuosity were quantified after perfusion of the mice with FITC-Concanavalin A. The mRNA levels of VEGF, angiopoietin 1 and 2 were quantified by real-time RT-PCR. RESULTS: After 17-AAG treatment, a reduction of the vascular area was measured from 37.8 +/- 5.2% to 30.8 +/- 5.7% (P = 0.005), and an increase of the avascular area from 10.8 +/- 5.6% to 20.3 +/- 6.6% (P = 0.001). No alteration of the vascular pattern, the number of blood vessel tufts and the main vessel tortuosity was achieved by treatment with the PI-3 kinase inhibitor Wortmannin. After treatment with 17-AAG, the numbers of tufts (127.9 33.2) were different from the controls (173.7 +/- 55.2, P = 0.035), but not the main vessel tortuosity. No significant change in VEGF and angiopoietin 1 mRNA expression could be achieved with either of the treatments. Wortmannin treatment also did not change the angiopoietin 2 mRNA level, whereas the level was reduced in 17-AAG treated mice retina from 436-fold (+/- 64) to 200-fold (+/-55) (P = 0.035). CONCLUSION: An IP injection of 17-AAG is able to reduce angioproliferative retinopathy in a mouse model for oxygen-induced retinopathy. Our data indicate that the mechanism does not involve a direct or indirect reduction of the VEGF mRNA level, but acts downstream of the VEGF pathway. Thus, 17-AAG probably does not work by PI-3 kinase inhibition but via the Ras/Raf/MEK pathway. These data underline the potential utility of tyrosine kinase inhibitors in hypoxia induced neovascularization.


Asunto(s)
Benzoquinonas/uso terapéutico , Modelos Animales de Enfermedad , Lactamas Macrocíclicas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Neovascularización Retiniana/tratamiento farmacológico , Retinopatía de la Prematuridad/tratamiento farmacológico , Angiopoyetina 1/genética , Angiopoyetina 2/genética , Animales , Femenino , Angiografía con Fluoresceína , Humanos , Recién Nacido , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , ARN Mensajero/metabolismo , Neovascularización Retiniana/diagnóstico , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Retinopatía de la Prematuridad/diagnóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética
11.
J Clin Endocrinol Metab ; 91(10): 4013-21, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16849420

RESUMEN

CONTEXT: The ubiquitin-proteasome pathway is a major pathway for degradation of intracellular proteins. Proteasome inhibitors constitute a novel class of antitumor agents with preclinical and clinical evidence of activity against hematological malignancies and solid tumors. The proteasome inhibitor bortezomib (PS-341, Velcade) has been approved by the Food and Drug Administration for the treatment of multiple myeloma and is being studied intensely in several other malignancies. Its mechanism of action is complex but appears to include the inhibition of inhibitory-kappaB degradation, which leads to inactivation of the transcriptional factor nuclear factor-kappaB (NF-kappaB). NF-kappaB has been implicated in the pathophysiology of the most aggressive forms of thyroid carcinoma, i.e. medullary and anaplastic. OBJECTIVE AND METHODS: We evaluated the effect of bortezomib on a panel of thyroid carcinoma cell lines, originating from papillary, follicular, anaplastic, and medullary carcinomas. RESULTS: Bortezomib induced apoptosis in medullary and anaplastic cell lines with IC(50) values well within the range of clinically achievable concentrations and much lower than respective IC(50) values for other solid malignancies. Bortezomib inhibited NF-kappaB activity; increased p53, p21, and jun expression; and induced caspase-dependent apoptosis. Sensitivity of thyroid carcinoma cells to bortezomib was partially decreased by overexpression of Bcl-2 or treatment with IGF-I, whereas the combination of bortezomib with chemotherapy (doxorubicin) was synergistic. CONCLUSIONS: These data provide both insights into the molecular mechanisms of antitumor activity of proteasome inhibitors and the rationale for future clinical trials of bortezomib, alone or in combination with conventional chemotherapy, to improve patient outcome in medullary and anaplastic thyroid carcinomas.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Carcinoma Medular/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Inhibidores de Proteasoma , Pirazinas/farmacología , Neoplasias de la Tiroides/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Bortezomib , Caspasas/fisiología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/análisis , Doxorrubicina/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Neoplasias de la Tiroides/patología , Proteína p53 Supresora de Tumor/análisis
12.
J Clin Endocrinol Metab ; 91(9): 3662-6, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16822827

RESUMEN

CONTEXT: The epidermal growth factor receptor (EGFR), a transmembrane tyrosine kinase (TK) receptor that mediates proliferation and survival signaling, is expressed in a wide variety of normal and neoplastic tissues. EGFR inhibitors have produced objective responses in patients with non-small-cell lung carcinomas harboring activating EGFR TK domain somatic mutations. OBJECTIVE AND METHODS: Because the EGFR pathway has been reported to be important for the pathophysiology of thyroid carcinoma, we investigated the expression and mutational status of EGFR in 14 thyroid carcinoma cell lines as well as its functional role by evaluating their in vitro sensitivity to AEE788, a new dual-family EGFR/ErbB2 and vascular endothelial growth factor receptor TK inhibitor. We also evaluated the mutational status, mRNA and protein expression, as well as phosphorylation status of EGFR in a panel of thyroid carcinoma specimens. RESULTS: EGFR expression and phosphorylation in the thyroid carcinoma cell lines and tissue specimens were present but not stronger than in noncancerous thyroid tissue. EGFR TK domain mutations were detected in two of 62 histological specimens (3.2%) but not in cell lines. All thyroid carcinoma cell lines were significantly less sensitive (IC(50) at least 25-fold higher) in vitro to AEE788 than a primary culture of EGFR-mutant lung carcinoma cells. CONCLUSIONS: Thyroid carcinoma cells overall are poorly responsive to clinically relevant concentrations of AEE788 in vitro. The presence of EGFR-activating TK domain mutations may identify a small minority of thyroid cancer patients that may benefit from EGFR inhibitors, but additional preclinical evidence of efficacy is needed.


Asunto(s)
Carcinoma/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Purinas/farmacología , Neoplasias de la Tiroides/tratamiento farmacológico , Adolescente , Adulto , Anciano , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mutación , FN-kappa B/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Estructura Terciaria de Proteína , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo
13.
Clin Cancer Res ; 12(12): 3705-12, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778096

RESUMEN

PURPOSE: The death receptor Fas is present in thyroid carcinomas, yet fails to trigger apoptosis. Interestingly, Fas has been reported to be actually overexpressed in papillary thyroid carcinomas, suggesting that it may confer a survival advantage. EXPERIMENTAL DESIGN: We investigated the expression and activation status of Fas pathway mediators in thyroid carcinoma cell lines and tumor specimens. RESULTS: All cell lines tested express Fas-associated death domain, procaspase-8, procaspase-9, and procaspase-3; resistance to Fas-mediated apoptosis could not be attributed to lack of any of these apoptosis mediators. Moreover, Fas death domain mutations were not found in our study. The proteasome inhibitors MG132 and PS-341 (bortezomib, Velcade), which lead to accumulation of the nuclear factor kappaB (NF-kappaB) inhibitor IkappaB, did not sensitize SW579 cells to Fas-mediated apoptosis, suggesting that resistance to Fas-mediated apoptosis is not due to proteasome or NF-kappaB activity. Cross-linking of Fas in vitro induced recruitment of Fas-associated death domain-like interleukin-1beta-converting enzyme inhibitory protein (FLIP) instead of procaspase-8. Inhibition of FLIP expression with a FLIP antisense oligonucleotide resulted in significant sensitization to Fas-mediated apoptosis. Fas cross-linking promoted BrdUrd incorporation; activated the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase, NF-kappaB, and activator protein-1 pathways in thyroid carcinoma cells in vitro; and protected cells from tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. We also found that good prognosis papillary thyroid carcinoma specimens exhibited higher immunoreactivity for cleaved (activated) caspase-8 than poor prognosis tumors. CONCLUSIONS: In thyroid carcinomas, the proteolytic cleavage and activation of caspase-8 depends on the balance between expression levels for procaspase-8 and FLIP and correlates with favorable clinical prognosis. Fas may actually stimulate proliferation and confer a survival advantage to thyroid cancer cells.


Asunto(s)
Transducción de Señal/fisiología , Neoplasias de la Tiroides/patología , Receptor fas/fisiología , Apoptosis , Secuencia de Bases , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , División Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mutación , Oligonucleótidos Antisentido , Receptores del Factor de Necrosis Tumoral/fisiología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/fisiopatología , Transfección
14.
FASEB J ; 20(10): 1689-91, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16807368

RESUMEN

Choroidal neovascularization (CNV) is responsible for the severe visual loss in age-related macular degeneration. CNV formation is considered to be due to an imbalance between pro- and antiangiogenic factors that lead to neovascular growth from the choriocapillaris into the subretinal space. To define whether FasL overexpression in retinal pigment epithelial cells (RPE) can inhibit choroidal neovascularization through Fas-FasL-mediated apoptosis, we examined the role of this pathway in a mouse model of laser-induced choroidal neovascularization. FasL was expressed in the retinal pigment epithelium of transgenic mice. Polymerase chain reaction (PCR), immunoblot, and immunohistochemistry confirmed that the transgene FasL was specifically expressed in RPE. The established laser model was used to induce choroidal neovascularization (CNV) in wild-type (WT) and transgenic mice. CNV formation was compared with respect to fluorescein angiographic leakage (at days 0 and 14 after laser injury) and histological appearance. The lesions were assessed on RPE-choroidal flatmounts after CD31-labeling and with confocal microscopy after perfusion with rhodamine-labeled concanavalin A (Con A). Apoptosis was quantified by TUNEL positivity and caspase activation. FasL mRNA and protein were highly expressed in the RPE of the transgenic mice before and after laser photocoagulation. In contrast, FasL was only weakly expressed in the RPE layer of WT C57BL/6J mice. While ruptures of Bruch's membrane and CNV formation were observed histologically two weeks after laser photocoagulation in transgenic as well as control eyes, the shape and size of CNV lesions were reduced in the transgenic mice. The area of leakage was decreased by 70% in FasL transgenic mice compared with WT mice (P<0.005). The number of TUNEL-positive cells was greater in FasL-overexpressing mice and correlated with the expression of activated caspases. Th expression of other antiangiogenic factors such as PEDF remained unchanged. The specific overexpression of FasL in RPE layer reduced CNV formation in our laser model. Our results strongly point to the FasL-Fas pathway as a potential therapeutic target in controlling pathological choroidal neovascularization.


Asunto(s)
Coroides/irrigación sanguínea , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/uso terapéutico , Neovascularización Patológica/prevención & control , Epitelio Pigmentado Ocular/citología , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/uso terapéutico , Animales , Apoptosis , Lámina Basal de la Coroides/patología , Capilares , Proteína Ligando Fas , Rayos Láser/efectos adversos , Degeneración Macular , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Epitelio Pigmentado Ocular/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Factores de Necrosis Tumoral/análisis
15.
Blood ; 107(3): 1092-100, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16234364

RESUMEN

We show that multiple myeloma (MM), the second most commonly diagnosed hematologic malignancy, is responsive to hsp90 inhibitors in vitro and in a clinically relevant orthotopic in vivo model, even though this disease does not depend on HER2/neu, bcr/abl, androgen or estrogen receptors, or other hsp90 chaperoning clients which are hallmarks of tumor types traditionally viewed as attractive clinical settings for use of hsp90 inhibitors, such as the geldanamycin analog 17-AAG. This class of agents simultaneously suppresses in MM cells the expression and/or function of multiple levels of insulin-like growth factor receptor (IGF-1R) and interleukin-6 receptor (IL-6R) signaling (eg, IKK/NF-kappaB, PI-3K/Akt, and Raf/MAPK) and downstream effectors (eg, proteasome, telomerase, and HIF-1alpha activities). These pleiotropic proapoptotic effects allow hsp90 inhibitors to abrogate bone marrow stromal cell-derived protection on MM tumor cells, and sensitize them to other anticancer agents, including cytotoxic chemotherapy and the proteasome inhibitor bortezomib. These results indicate that hsp90 can be targeted therapeutically in neoplasias that may not express or depend on molecules previously considered to be the main hsp90 client proteins. This suggests a more general role for hsp90 in chaperoning tumor- or tissue-type-specific constellations of client proteins with critical involvement in proliferative and antiapoptotic cellular responses, and paves the way for more extensive future therapeutic applications of hsp90 inhibition in diverse neoplasias, including MM.


Asunto(s)
Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Mieloma Múltiple/metabolismo , Rifabutina/análogos & derivados , Transducción de Señal/efectos de los fármacos , Andrógenos/metabolismo , Apoptosis/efectos de los fármacos , Benzoquinonas , Ácidos Borónicos/farmacología , Bortezomib , Sinergismo Farmacológico , Estrógenos/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinasa I-kappa B/metabolismo , Lactamas Macrocíclicas , Mieloma Múltiple/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/biosíntesis , Receptores de Interleucina-6/biosíntesis , Rifabutina/farmacología , Células Tumorales Cultivadas , Quinasas raf/metabolismo
16.
Clin Cancer Res ; 11(10): 3958-65, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15897598

RESUMEN

Histone deacetylases (HDAC) and histone acetyltransferases exert opposing enzymatic activities that modulate the degree of acetylation of histones and other intracellular molecular targets, thereby regulating gene expression, cellular differentiation, and survival. HDAC inhibition results in accumulation of acetylated histones and induces differentiation and/or apoptosis in transformed cells. In this study, we characterized the effect of two HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and m-carboxycinnamic acid bis-hydroxamide, on thyroid carcinoma cell lines, including lines originating from anaplastic and medullary carcinomas. In these models, both SAHA and m-carboxycinnamic acid bis-hydroxamide induced growth arrest and caspase-mediated apoptosis and increased p21 protein levels, retinoblastoma hypophosphorylation, BH3-interacting domain death agonist cleavage, Bax up-regulation, down-regulation of Bcl-2, A1, and Bcl-x(L) expression, and cleavage of poly(ADP-ribose) polymerase and caspase-8, -9, -3, -7, and -2. Transfection of Bcl-2 cDNA partially suppressed SAHA-induced cell death. SAHA down-regulated the expression of the apoptosis inhibitors FLIP and cIAP-2 and sensitized tumor cells to cytotoxic chemotherapy and death receptor activation. Our studies provide insight into the tumor type-specific mechanisms of antitumor effects of HDAC inhibitors and a framework for future clinical applications of HDAC inhibitors in patients with thyroid cancer, including histologic subtypes (e.g., anaplastic and medullary thyroid carcinomas) for which limited, if any, therapeutic options are available.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma/patología , Cinamatos/farmacología , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Neoplasias de la Tiroides/patología , Apoptosis , Muerte Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células Tumorales Cultivadas , Vorinostat
17.
Invest Ophthalmol Vis Sci ; 46(1): 358-66, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15623796

RESUMEN

PURPOSE: Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL)/Apo2L are members of the TNFalpha family that can trigger apoptosis in susceptible cells via respective death receptors (DRs). FasL cross-links its receptor Fas, resulting in recruitment and proteolytic activation of caspase-8, which initiates the downstream apoptotic cascade. TRAIL signals through its receptors DR4 and DR5, which can activate caspase-8 as well. This study was undertaken to investigate the functional status of the FasL and TRAIL apoptotic pathways in retinoblastoma (Rb) cells. METHODS: The human Rb cell lines Y79 and WERI-Rb1 were evaluated for their response to the Fas cross-linking antibody CH11 and recombinant TRAIL, as well as for cell surface presence and mutational status of Fas, DR4, and DR5 by flow cytometry and genomic DNA sequencing, respectively. The expression of caspase-8 and its inhibitor FLIP, as well as their recruitment to the DR signaling complex were studied by immunoblot analysis. RESULTS: Rb cells express Fas, DR4, and DR5 on their surfaces, yet were resistant to DR-mediated apoptosis. This was not due to DR mutations or secretion of the soluble decoy Fas, antiapoptotic NF-kappaB activity, or FLIP overexpression, but to the absence of caspase-8 expression. The demethylating agent 5-aza-2'-deoxycytidine restored caspase-8 expression and sensitivity to DR-mediated apoptosis. CONCLUSIONS: Rb cells are resistant to DR-mediated apoptosis because of a deficiency in caspase-8 expression secondary to epigenetic gene silencing by overmethylation. The data help delineate the apoptotic pathways in Rb cells and suggest that the combination of demethylating agents with DR-activating modalities, such as TRAIL receptor monoclonal antibodies, may benefit patients with retinoblastoma.


Asunto(s)
Apoptosis , Caspasas/genética , Silenciador del Gen/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/patología , Azacitidina/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Caspasa 8 , Inhibidores de Caspasas , Supervivencia Celular , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Citometría de Flujo , Humanos , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular/farmacología , Glicoproteínas de Membrana/metabolismo , FN-kappa B/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Neoplasias de la Retina/enzimología , Retinoblastoma/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Receptor fas/metabolismo
18.
FASEB J ; 18(12): 1450-2, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15231732

RESUMEN

Diabetic retinopathy is a leading cause of adult vision loss and blindness. Much of the retinal damage that characterizes the disease results from retinal vascular leakage and nonperfusion. Diabetic retinal vascular leakage, capillary nonperfusion, and endothelial cell damage are temporary and spatially associated with retinal leukocyte stasis in early experimental diabetes. Retinal leukostasis increases within days of developing diabetes and correlates with the increased expression of retinal intercellular adhesion molecule-1 (ICAM-1) and CD18. Mice deficient in the genes encoding for the leukocyte adhesion molecules CD18 and ICAM-1 were studied in two models of diabetic retinopathy with respect to the long-term development of retinal vascular lesions. CD18-/- and ICAM-1-/- mice demonstrate significantly fewer adherent leukocytes in the retinal vasculature at 11 and 15 months after induction of diabetes with STZ. This condition is associated with fewer damaged endothelial cells and lesser vascular leakage. Galactosemia of up to 24 months causes pericyte and endothelial cell loss and formation of acellular capillaries. These changes are significantly reduced in CD18- and ICAM-1-deficient mice. Basement membrane thickening of the retinal vessels is increased in long-term galactosemic animals independent of the genetic strain. Here we show that chronic, low-grade subclinical inflammation is responsible for many of the signature vascular lesions of diabetic retinopathy. These data highlight the central and causal role of adherent leukocytes in the pathogenesis of diabetic retinopathy. They also underscore the potential utility of anti-inflammatory treatment in diabetic retinopathy.


Asunto(s)
Retinopatía Diabética/etiología , Retinopatía Diabética/patología , Inflamación/complicaciones , Inflamación/patología , Animales , Antígenos CD18/genética , Antígenos CD18/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Galactosa/sangre , Galactosa/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/metabolismo , Retina/patología , Factores de Tiempo
19.
Am J Pathol ; 165(2): 457-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277220

RESUMEN

Diabetic retinopathy is a leading cause of blindness in the Western world. Aberrant intercellular adhesion molecule-1 expression and leukocyte adhesion have been implicated in its pathogenesis, raising the possibility of an underlying chronic inflammatory mechanism. In the current study, the role of insulin-like growth factor (IGF)-I in these processes was investigated. We found that systemic inhibition of IGF-I signaling with a receptor-neutralizing antibody, or with inhibitors of PI-3 kinase (PI-3K), c-Jun kinase (JNK), or Akt, suppressed retinal Akt, JNK, HIF-1alpha, nuclear factor (NF)-kappaB, and AP-1 activity, vascular endothelial growth factor (VEGF) expression, as well as intercellular adhesion molecule-1 levels, leukostasis, and blood-retinal barrier breakdown, in a relevant animal model. Intravitreous administration of IGF-I increased retinal Akt, JNK, HIF-1alpha, NF-kappaB, and AP-1 activity, and VEGF levels. IGF-I stimulated VEGF promoter activity in vitro, mainly via HIF-1alpha, and secondarily via NF-kappaB and AP-1. In conclusion, IGF-I participates in the pathophysiology of diabetic retinopathy by inducing retinal VEGF expression via PI-3K/Akt, HIF-1alpha, NF-kappaB, and secondarily, JNK/AP-1 activation. Taken together, these in vitro and in vivo signaling studies thus identify potential targets for pharmacological intervention to preserve vision in patients with diabetes.


Asunto(s)
Retinopatía Diabética/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Epitelio Pigmentado Ocular/metabolismo , Animales , Barrera Hematorretinal/efectos de los fármacos , Barrera Hematorretinal/fisiología , Retinopatía Diabética/patología , Ensayo de Cambio de Movilidad Electroforética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Molécula 1 de Adhesión Intercelular/metabolismo , MAP Quinasa Quinasa 4 , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Long-Evans , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Cancer Cell ; 5(3): 221-30, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15050914

RESUMEN

Insulin-like growth factors and their receptor (IGF-1R) have been implicated in cancer pathophysiology. We demonstrate that IGF-1R is universally expressed in various hematologic (multiple myeloma, lymphoma, leukemia) and solid tumor (breast, prostate, lung, colon, thyroid, renal, adrenal cancer, retinoblastoma, and sarcoma) cells. Specific IGF-1R inhibition with neutralizing antibody, antagonistic peptide, or the selective kinase inhibitor NVP-ADW742 has in vitro activity against diverse tumor cell types (particularly multiple myeloma), even those resistant to conventional therapies, and triggers pleiotropic antiproliferative/proapoptotic molecular sequelae, delineated by global transcriptional and proteomic profiling. NVP-ADW742 monotherapy or its combination with cytotoxic chemotherapy had significant antitumor activity in an orthotopic xenograft MM model, providing in vivo proof of principle for therapeutic use of selective IGF-1R inhibitors in cancer.


Asunto(s)
Neoplasias Hematológicas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Antineoplásicos , Médula Ósea/metabolismo , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Perfilación de la Expresión Génica , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Mieloma Múltiple , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Trasplante Heterólogo/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA