Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Ophthalmol ; 20(1): 12, 2020 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-31906897

RESUMEN

BACKGROUND: Tear film stability is the key event in ocular surface diseases. The purpose of this study is to evaluate spatial and temporal progression of the tear film breakup using an automatic non-invasive device. METHODS: Non-invasive tear breakup time (NITBUT) parameters, such as First NITBUT (F-NITBUT) and Average NITBUT (A-NITBUT), were evaluated in 132 glaucoma and 87 control eyes with the Keratograph 5 M device. Further analysis of this data was used to determine size, location and progression of tear film breakup with automatically identified breakup areas (BUA). The progression from First BUA (F-BUA) to total BUA (T-BUA) was expressed as Dry Area Growth Rate (DAGR). Differences between both groups were analysed using Student t-test for parametric data and Mann-Whitney U test for non-parametric data. Pearson's correlation coefficient was used to assess the relationship between parametric variables and Spearman in the case of non-parametric variables. RESULTS: F-NITBUT was 11.43 ± 7.83 s in the control group and 8.17 ± 5.73 in the glaucoma group (P = 0.010). A-NITBUT was 14.04 ± 7.21 and 11.82 ± 6.09 s in control and glaucoma groups, respectively (P = 0.028). F-BUA was higher in the glaucoma group than in the control group (2.73 and 2.28; P = 0.022) and was more frequently located at the centre of the cornea in the glaucoma group (P = 0.039). T-BUA was also higher in the glaucoma group than in the control group (13.24 and 9.76%; P = 0.012) and the DAGR was steeper in the glaucoma group than in the control group (34.38° and 27.15°; P = 0.009). CONCLUSIONS: Shorter NITBUT values and bigger, more central tear film breakup locations were observed in the glaucoma group than in the control group. The DAGR indicates that tear film rupture is bigger and increases faster in glaucomatous eyes than in normal eyes.


Asunto(s)
Técnicas de Diagnóstico Oftalmológico , Glaucoma/metabolismo , Lágrimas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Estudios Transversales , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
2.
Int Ophthalmol ; 40(3): 741-751, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31813109

RESUMEN

PURPOSE: To evaluate the quality of life of glaucoma patients using the Ocular Surface Disease Index (OSDI) questionnaire and their association with dry eye clinical signs. METHODS: The study included patients into three groups. The treated group diagnosed with bilateral open-angle glaucoma and treated with one or more topical medication at least 1 year. The operated group underwent glaucoma surgery without the need for topical medications. The control group entered subjects without ocular diseases or previous surgeries. Dry eye clinical signs were evaluated; noninvasive tear break-up time, Meibomian gland depletion (MGD), and conjunctival hyperemia were measured using the Keratograph 5 M. The total-OSDI (T-OSDI) score was divided into the visual field-OSDI and discomfort-OSDI scores. RESULTS: Two hundred and nine subjects participated in this cross-sectional study, 147 using glaucoma medications, 21 patients underwent glaucoma surgery and 41 were controls. The T-OSDI and subscores were higher in glaucoma patients compared with controls (p < 0.05); we found no differences between treated and surgically groups. Correlations were observed between the T-OSDI values and Schirmer test (p = 0.016), ocular surface staining (p < 0.001) and the MGD (p = 0.006). The subscores were associated with the ocular surface staining (VF p = 0.013 and D p = 0.003). In treated patients, the number of drops per day correlates with T-OSDI and subscores (p = 0.017 and p = 0.005). CONCLUSION: OSDI scores increased in the glaucoma patients compared to controls without significant changes between treated and surgical patients. OSDI scores were associated to dry eye signs and medication in glaucoma patients.


Asunto(s)
Antihipertensivos/administración & dosificación , Glaucoma/tratamiento farmacológico , Presión Intraocular/fisiología , Calidad de Vida , Campos Visuales/fisiología , Adulto , Estudios Transversales , Femenino , Estudios de Seguimiento , Glaucoma/fisiopatología , Humanos , Presión Intraocular/efectos de los fármacos , Masculino , Persona de Mediana Edad , Soluciones Oftálmicas , Estudios Prospectivos , Encuestas y Cuestionarios , Lágrimas/metabolismo
3.
Cell Tissue Res ; 359(3): 829-39, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25557989

RESUMEN

Colorectal cancer (CRC) cells often metastatize to the liver. Cancer-associated fibroblasts (CAFs) enhance metastasis by providing cytokines that create a favorable microenvironment and by inducing co-dissemination with tumor cells. However, the mechanisms of co-metastatization remain elusive. The aim of this study is to assess the role of TGFß1 in CRC cell-CAFs attachment and its impact on liver metastasis. CAFs were obtained after xenotransplantation of Mc38 cells into EGFP-C57BL/6 mice. Attachment experiments with CRC cells and CAFs (with or without TGFß1 and the inhibitory peptide P17) were carried out, as well as in vivo liver metastasis assays. TGFß1 induced adhesion of CRC cells to CAFs, whereas exposure to P17 abrogated this effect. Co-injection of Mc38 cells with CAFs intrasplenically increased liver metastasis, as compared to injection of tumor cells alone. Pretreatment of Mc38 cells with TGFß1 enhanced the metastatic burden, in comparison to untreated Mc38 + CAFs. TGFß1-pretreated Mc38 cells co-metastatized with CAFs to the liver in a highly efficient way. Importantly, the metastatic burden was significantly reduced (p < 0.001) when P17 was administered in mice. The number of PCNA+ and CD-31+ cells was also reduced by P17 in these animals, indicating a decrease in proliferation and angiogenesis upon TGFß1 signaling blockade. Through microarray analysis, we identified potential TGFß1-regulated genes that may mediate cancer cell-stroma interactions to increase metastasis. In conclusion, TGFß1 promotes co-travelling of CRC cells and CAFs to the liver to enhance metastasis. Our results strongly support the use of TGFß1 targeted drugs as a novel strategy to reduce liver metastasis in CRC patients.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/patología , Fibroblastos/patología , Neoplasias Hepáticas/secundario , Factor de Crecimiento Transformador beta1/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones Endogámicos C57BL
4.
Int J Cancer ; 135(11): 2516-27, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24473991

RESUMEN

New mouse models with specific drivers of genetic alterations are needed for preclinical studies. Herein, we created and characterized at the genetic level a new syngeneic model for lung cancer and metastasis in Balb-c mice. Tumor cell lines were obtained from a silica-mediated airway chronic inflammation that promotes tumorigenesis when combined with low doses of N-nitrosodimethylamine, a tobacco smoke carcinogen. Orthotopic transplantation of these cells induced lung adenocarcinomas, and their intracardiac injection led to prominent colonization of various organs (bone, lung, liver and brain). Driver gene alterations included a mutation in the codon 12 of KRAS (G-A transition), accompanied by a homozygous deletion of the WW domain-containing oxidoreductase (WWOX) gene. The mutant form of WWOX lacked exons 5-8 and displayed reduced protein expression level and activity. WWOX gene restoration decreased the in vitro and in vivo tumorigenicity, confirming the tumor suppressor function of this gene in this particular model. Interestingly, we found that cells displayed remarkable sphere formation ability with expression of specific lung cancer stem cell markers. Study of non-small-cell lung cancer patient cohorts demonstrated a deletion of WWOX in 30% of cases, with significant reduction in protein levels as compared to normal tissues. Overall, our new syngeneic mouse model provides a most valuable tool to study lung cancer metastasis in balb-c mice background and highlights the importance of WWOX deletion in lung carcinogenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/secundario , Modelos Animales de Enfermedad , Inflamación/patología , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/patología , Oxidorreductasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética , Proteínas ras/genética , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Animales , Apoptosis , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Proliferación Celular , Hibridación Genómica Comparativa , Transición Epitelial-Mesenquimal , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Inflamación/genética , Inflamación/mortalidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Estadificación de Neoplasias , Oxidorreductasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
5.
Exp Cell Res ; 319(3): 12-22, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23153552

RESUMEN

Colorectal cancer (CRC) frequently metastasizes to the liver, a phenomenon that involves the participation of transforming-growth-factor-ß(1) (TGFß(1)). Blockade of the protumorigenic effects elicited by TGFß(1) in advanced CRC could attenuate liver metastasis. We aimed in the present study to assess the antimetastatic effect of TGFß(1)-blocking peptides P17 and P144, and to study mechanisms responsible for this activity in a mouse model. Colon adenocarcinoma cells expressing luciferase were pretreated with TGFß(1) (Mc38-luc(TGFß1) cells), injected into the spleen of mice and monitored for tumor development. TGFß(1) increased primary tumor growth and liver metastasis, whereas systemic treatment of mice with either P17 or P144 significantly reduced tumor burden (p<0.01). In metastatic nodules, mitotic/apoptotic ratio, mesenchymal traits and angiogenesis (evaluated by CD-31, as well as circulating endothelial and progenitor cells) induced by TGFß(1) were consistently reduced following injection of peptides. In vitro experiments revealed a direct effect of TGFß(1) in Mc38 cells, which resulted in activation of Smad2, Smad3 and Smad1/5/8, and increased invasion and transendothelial migration, whereas blockade of TGFß(1)-signaling reverted these features. Because TGFß(1)-mediated epithelial-mesenchymal transition (EMT) has been suggested to induce a cancer stem cell (CSC) phenotype, we analyzed the ability of this cytokine to induce tumorsphere formation and the expression of CSC markers. In TGFß(1)-treated cells, tumorspheres were enriched in CD44 and SOX2, which were diminished in the presence of P17. Our data provide a preclinical rationale to evaluate P17 and P144 as potential therapeutic options for the treatment of metastatic CRC.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Hepáticas/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Fragmentos de Péptidos/uso terapéutico , Péptidos/uso terapéutico , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Adenocarcinoma/patología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Células Cultivadas , Neoplasias del Colon/patología , Transición Epitelial-Mesenquimal/fisiología , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Células Madre Neoplásicas/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , Péptidos/administración & dosificación , Péptidos/farmacología , Fenotipo , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
6.
Lab Invest ; 92(7): 952-66, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22546866

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common solid tumor and the third leading cause of cancer-related deaths. Currently available chemotherapeutic options are not curative due in part to tumor resistance to conventional therapies. We generated orthotopic HCC mouse models in immunodeficient NOD/SCID/IL2rγ null mice by injection of human alpha-feto protein (hAFP)- and/or luciferase-expressing HCC cell lines and primary cells from patients, where tumor growth and spread can be accurately monitored in a non-invasive way. In this model, low-dose metronomic administration of cyclophosphamide (LDM-CTX) caused complete regression of the tumor mass. A significant increase in survival (P<0.0001), reduced aberrant angiogenesis and hyperproliferation, and decrease in the number of circulating tumor cells were found in LDM-CTX-treated animals, in comparison with untreated mice. Co-administration of LDM-CTX with anti-VEGF therapy further improved the therapeutic efficacy. However, the presence of residual circulating hAFP levels suggested that some tumor cells were still present in livers of treated mice. Immunohistochemistry revealed that those cells had a hAFP+/CD13+/PCNA- phenotype, suggesting that they were dormant cancer stem cells (CSC). Indeed, discontinuation of therapy resulted in tumor regrowth. Moreover, in-vitro LDM-CTX treatment reduced hepatosphere formation in both number and size, and the resulting spheres were enriched in CD13+ cells indicating that these cells were particularly resistant to therapy. Co-treatment of the CD13-targeting drug, bestatin, with LDM-CTX leads to slower tumor growth and a decreased tumor volume. Therefore, combining a CD13 inhibitor, which targets the CSC-like population, with LDM-CTX chemotherapy may be used to eradicate minimal residual disease and improve the treatment of liver cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/patología , Células Madre Neoplásicas/patología , Administración Metronómica , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antineoplásicos Alquilantes/administración & dosificación , Antígenos CD13/antagonistas & inhibidores , Línea Celular Tumoral , Ciclofosfamida/administración & dosificación , Sinergismo Farmacológico , Humanos , Leucina/administración & dosificación , Leucina/análogos & derivados , Neoplasias Hepáticas Experimentales/fisiopatología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasia Residual/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/fisiología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer ; 10: 96, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21827695

RESUMEN

BACKGROUND: Mortality rates for advanced lung cancer have not declined for decades, even with the implementation of novel chemotherapeutic regimens or the use of tyrosine kinase inhibitors. Cancer Stem Cells (CSCs) are thought to be responsible for resistance to chemo/radiotherapy. Therefore, targeting CSCs with novel compounds may be an effective approach to reduce lung tumor growth and metastasis. We have isolated and characterized CSCs from non-small cell lung cancer (NSCLC) cell lines and measured their telomerase activity, telomere length, and sensitivity to the novel telomerase inhibitor MST312. RESULTS: The aldehyde dehydrogenase (ALDH) positive lung cancer cell fraction is enriched in markers of stemness and endowed with stem cell properties. ALDH+ CSCs display longer telomeres than the non-CSC population. Interestingly, MST312 has a strong antiproliferative effect on lung CSCs and induces p21, p27 and apoptosis in the whole tumor population. MST312 acts through activation of the ATM/pH2AX DNA damage pathway (short-term effect) and through decrease in telomere length (long-term effect). Administration of this telomerase inhibitor (40 mg/kg) in the H460 xenograft model results in significant tumor shrinkage (70% reduction, compared to controls). Combination therapy consisting of irradiation (10Gy) plus administration of MST312 did not improve the therapeutic efficacy of the telomerase inhibitor alone. Treatment with MST312 reduces significantly the number of ALDH+ CSCs and their telomeric length in vivo. CONCLUSIONS: We conclude that antitelomeric therapy using MST312 mainly targets lung CSCs and may represent a novel approach for effective treatment of lung cancer.


Asunto(s)
Benzamidas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Aldehído Deshidrogenasa/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzamidas/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Terapia Molecular Dirigida , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Telomerasa/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Methods Mol Biol ; 2282: 417-442, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928588

RESUMEN

The field of RNAi therapeutics has quickly adapted to the treatment of ocular diseases. Although the eye provides a unique system for the delivery of siRNAs, its complex structure and composition fostered the development of novel strategies for efficient gene silencing in the target compartment. Moreover, anterior and posterior segments differ in their multiple drug barriers and clearance mechanisms. This chapter summarizes the recent achievements in terms of routes of administration, chemical modifications, and delivery systems for siRNAs that specifically apply to eye disorders. Methods employed for siRNA detection/quantitation in ocular tissues are also described, together with safety concerns that need to be addressed to fulfill regulatory requirements of new drug approval. Even though RNAi therapies for ocular diseases have not yet translated into patient care, we document herein the rising number of candidate drugs currently under preclinical or clinical development.


Asunto(s)
Oftalmopatías/terapia , Ojo/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Animales , Disponibilidad Biológica , Oftalmopatías/genética , Oftalmopatías/metabolismo , Técnicas de Transferencia de Gen , Humanos , ARN Interferente Pequeño/metabolismo , Distribución Tisular
9.
J Exp Ther Oncol ; 8(3): 247-60, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20734923

RESUMEN

Evidence suggests that stem-like cells are responsible for initiation, maintenance and recurrence of solid tumors, including Glioblastoma Multiforme (GBM). GBM is an intractable, highly lethal tumor of the central nervous system. Although epidermal growth factor receptor (EGFR) is highly expressed in many GBMs, anti-EGFR therapies have been unsuccessful as treatment. Few studies have examined EGFR activation in GBM stem cells (GSCs) to determine if patient-specific GSCs are amenable to anti-EGFR therapy pre-clinically. We hypothesized that EGFR activation in GSCs varied between patients and was an important determinant of responsiveness to anti-EGFR therapy. Cell cycle and apoptosis analysis was performed on tumor-spheres by immuncytochemistry in the presence and absence of the AG1478. Second messenger pathways operative in these processes were elucidated by immunoblotting. EGFR activated AKT and inactivated GSK3beta in EGFR+/PTEN+ GSCs. AG1478 and erlotinib significantly decreased the total number of tumor-spheres that EGFR+/ PTEN+ GSCs generated and the rate of sphere formation. Inhibition of EGFR signaling by AG1478 increased GSC senescence and apoptosis, likely via inhibition of AKT and activation of GSK3beta. Sphere formation by EGFR-/ PTEN- GSCs was independent of EGF stimulation, but dependant on B27 growth supplement. Our data suggest that EGFR+/PTEN+ GSCs are susceptible to anti-EGFR therapy in vitro.


Asunto(s)
Receptores ErbB/fisiología , Glioblastoma/patología , Células Madre Neoplásicas/fisiología , Transducción de Señal/fisiología , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib , Glioblastoma/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Fosfohidrolasa PTEN/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/fisiología , Quinazolinas/farmacología , Tirfostinos/farmacología
10.
Neurosurg Focus ; 24(3-4): E28, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18341405

RESUMEN

OBJECT: Brain tumor stem cells (TSCs) hypothetically drive the malignant phenotype of glioblastoma multiforme (GBM), and evidence suggests that a better understanding of these TSCs will have profound implications for treating gliomas. When grown in vitro, putative TSCs grow as a solid sphere, making their subsequent characterization, particularly the cells within the center of the sphere, difficult. Therefore, the purpose of this study was to develop a new method to better understand the proteomic profile of the entire population of cells within a sphere. METHODS: Tumor specimens from patients with confirmed GBM and glioma models in mice were mechanically and enzymatically dissociated and grown in traditional stem cell medium to generate neurospheres. The neurospheres were then embedded in freezing medium, cryosectioned, and analyzed with immunofluorescence. RESULTS: By sectioning neurospheres as thinly as 5 mum, the authors overcame many of the problems associated with immunolabeling whole neurospheres, such as antibody penetration into the core of the sphere and intense background fluorescence that obscures the specificity of immunoreactivity. Moreover, the small quantity of material required and the speed with which this cryosectioning and immunolabeling technique can be performed make it an attractive tool for the rapid assessment of TSC character. CONCLUSIONS: This study is the first to show that cryosectioning of neurospheres derived from glioma models in mice and GBM in humans is a feasible method of better defining the stem cell profile of a glioma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/metabolismo , Animales , Glioma/metabolismo , Glioma/patología , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Células Tumorales Cultivadas
11.
Expert Opin Investig Drugs ; 27(4): 421-426, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29569947

RESUMEN

INTRODUCTION: Dry eye disease (DED) is characterized by an alteration of the tear film with ocular inflammation and neurosensory abnormalities. The main clinical signs of this condition are tear instability and ocular damage. Although DED has gained significant attention in the past few years, limited prescription treatment options are available for patients. Areas covered: The current manuscript summarizes the pre-clinical and clinical development of tivanisiran, a novel small interfering oligonucleotide of RNA (siRNA) used for the treatment of DED. Tivanisiran was designed to silence Transient Receptor Potential Vanilloid 1 (TRPV1); herein the chemistry and mechanism of action of this new compound is also described. Expert opinion: Drugs currently on the market mostly target the inflammatory component of the disease and show only partial efficacy. New compounds addressing other aspects of the disease would provide significant advantages and contribute to a more personalized treatment of the disease. Tivanisiran has been designed to reduce ocular discomfort and pain, and was shown to improve ocular hyperemia and tear quality in human and animal models. Consequently, if the results of the ongoing and future clinical trials meet their study endpoints, tivanisiran could be submitted to obtain approval for the treatment of DED.


Asunto(s)
Síndromes de Ojo Seco/terapia , ARN Interferente Pequeño/administración & dosificación , Canales Catiónicos TRPV/genética , Animales , Síndromes de Ojo Seco/genética , Silenciador del Gen , Humanos , Hiperemia/terapia , Oligonucleótidos/administración & dosificación , Oligonucleótidos/farmacología , ARN Interferente Pequeño/farmacología , Lágrimas/metabolismo
12.
Cancer Lett ; 414: 257-267, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29133238

RESUMEN

A major complication of colorectal cancer (CRC), one of the most frequent and deadly types of cancer, is disease progression via liver metastases. At this stage, very few treatment options are available for patients, and the disease remains incurable. Herein, we used a well-established mouse model of CRC liver metastasis (CLM) to identify new regulators of this process. Using serial transplantation of murine MC38 adenocarcinoma cells, we obtained liver metastatic variants that displayed extremely strong colonization abilities. Using these newly established cell lines, we performed gene expression arrays and microRNA (miR) profiling. Comparative and predictive analyses between the two arrays showed higher expression of c-met and concomitant reduction of miR-146a in the mestastatic variants. In CRC patients, expression levels of both c-met and miR-146a were similar between primary tumors and liver metastases. Interestingly, we identified c-met as a new target for miR-146a, as miR-146a was able to impede c-met translation. Of relevance, overexpression of miR-146a in metastatic clones showed reduced in vitro malignancy and abolished the development of primary tumor and liver metastases. Our results document a new mechanism for c-met regulation in CLM and highlight the crucial role of miR-146a in suppressing tumorigenesis.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-met/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Proteínas Proto-Oncogénicas c-met/metabolismo
13.
Biochem Pharmacol ; 71(3): 366-76, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16332358

RESUMEN

In vivo, the 21-aminosteroid U74389G prevents the decrease in cytochrome P450 (P450) activity produced by a turpentine-induced inflammatory reaction (TIIR). To investigate the underlying mechanism of action, four groups of rabbits were used, controls receiving or not U74389G, and rabbits with the inflammatory reaction receiving or not U74389G. Hepatocytes were isolated 48h later and incubated for 4 and 24h with the serum of the rabbits. In vivo, the TIIR diminished CYP1A1/2 and 3A6 expression, and enhanced hepatic malondialdehyde (MDA) and nitric oxide (NO*) concentrations (p<0.05). U74389G prevented the increase in MDA, as well as the decrease in CYP1A1/2 amounts and activity, but increased CYP3A6 expression by 40% (p<0.05). In vitro, compared with serum from control rabbits (S(CONT)), incubation of serum from rabbits with TIIR (S(TIIR)) for 4 and 24h with hepatocytes from rabbits with TIIR (H(TIIR)) reduced CYP1A2 and CYP3A6 activity (p<0.05) and increased the formation of NO* and MDA. In rabbits with TIIR pretreated with U74389G, the S(TIIR+U) failed to reduce CYP1A2 activity or to increase MDA, although increased NO* and further reduced CYP3A6 activity. On the other hand, in hepatocytes harvested from rabbits with TIIR pretreated with U74389G, S(TIIR) did not decrease CYP1A2 activity and did not enhance MDA, but still increased NO*. In vitro, the reduction of CYP1A2 and CYP3A6 activity by S(TIIR) is not associated to NF-kappaB activation. In conclusion, U74389G prevents CYP1A1/2 down-regulation and decrease in activity by a double mechanism: hindering the release of serum mediators and by averting intracellular events, effect possibly associated with its antioxidant activity. On the other hand, U74389G up-regulates CYP3A6 but inhibits its catalytic activity.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Inflamación/metabolismo , Pregnatrienos/farmacología , Sustancias Protectoras/farmacología , Animales , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A2/biosíntesis , Regulación hacia Abajo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Inflamación/inducido químicamente , Peroxidación de Lípido/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , Conejos , Suero/metabolismo , Trementina
14.
Invest Ophthalmol Vis Sci ; 57(14): 6447-6454, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27893109

RESUMEN

Purpose: To evaluate the efficacy and safety of SYL1001, a short interfering (si) RNA targeting the transient receptor potential cation channel subfamily V member 1 (TRPV1), for the treatment of dry eye disease (DED). Methods: This study combines a phase I and two phase II clinical trials to test different doses of SYL1001 in a total of 156 healthy subjects and patients with DED. After 10 days of treatment, the primary efficacy endpoints were the effect on (1) the scoring in the Visual Analogue Scale (VAS) and Ocular Surface Disease Index (OSDI) questionnaires, and (2) ocular tolerance evaluated by corneal fluorescein staining and conjunctival hyperemia. Secondary endpoints included the assessment of systemic and local tolerance. Results: Topical administration of SYL1001 1.125% once daily produced a significant decrease in VAS scores compared with placebo from day 4 until the end of treatment (change from baseline at day 10: -1.73 ± 0.32 vs. -0.91 ± 0.34; P = 0.013). For all treatments, OSDI scores were significantly reduced compared to their respective baseline values (P < 0.01), although no significant changes were detected between groups. Conjunctival hyperemia (quantified as normal or abnormal) significantly improved after instillation of SYL1001 1.125% compared with placebo (50% vs. 20%; P < 0.05). Excellent tolerability was reported, with no differences in the rates of occurrence of adverse events between groups. Conclusion: These trials achieved their primary endpoints of identifying the most effective dose of SYL1001 (1.125%). SYL1001 showed a large safety margin and may provide novel therapeutic opportunity for the relief of dry eye. (ClinicalTrials.gov numbers, NCT01438281, NCT01776658, and NCT02455999.).


Asunto(s)
Síndromes de Ojo Seco/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Canales Catiónicos TRPV/metabolismo , Lágrimas/metabolismo , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Síndromes de Ojo Seco/diagnóstico , Síndromes de Ojo Seco/metabolismo , Femenino , Humanos , Masculino , Soluciones Oftálmicas/administración & dosificación , Estudios Prospectivos , Canales Catiónicos TRPV/efectos de los fármacos , Lágrimas/efectos de los fármacos , Resultado del Tratamiento , Adulto Joven
15.
Front Biosci ; 10: 998-1009, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15769600

RESUMEN

The CCN genes encode secreted proteins, associated to the extracellular matrix. They are involved in diverse biological processes such as regulation of cell- adhesion, migration, proliferation, differentiation and survival. They play important roles in pregnancy, development, angiogenesis, wound repair and inflammation. Several lines of evidence support a role for CCN genes in fibrotic disorders and tumorigenesis. We will focus our attention in this review on two CCN proteins: CCN1 and CCN3, that appear to exert distinct and opposite effects. Recent data suggest that CCN1 acts as a tumor-promoting factor and a key regulator in cancer progression, while CCN3 exhibits suppressive capabilities. The possible opposite functions of CCN1 and CCN3 in tumorigenesis, and the relevance of the distinct expression profiles of these two genes observed in many cancers are discussed below.


Asunto(s)
Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neoplasias/metabolismo , Neovascularización Fisiológica/fisiología , Animales , Pruebas de Carcinogenicidad , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo , Proteína 61 Rica en Cisteína , Femenino , Humanos , Proteína Hiperexpresada del Nefroblastoma , Embarazo
16.
Cancer Lett ; 356(2 Pt B): 899-909, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25449776

RESUMEN

Id1 has been shown to play a critical role in tumorigenesis and angiogenesis. Moreover, recent reports have involved Id1 in the maintenance of cancer stem cell features in some tumor types. The Id1 gene generates two isoforms through alternative splicing: Id1a and Id1b. We have investigated the role of each isoform in cancer development. Using lentiviral systems we modified the endogenous expression of each of these isoforms in cancer cells and analyzed their biological effect both in vitro and in vivo. Overexpression of Id1b in murine CT26 and 3LL cells caused a G0/G1 cell cycle arrest and reduced proliferation, clonogenicity and phospho-ERK1/2 levels, while increasing p27 levels. High levels of Id1a had an opposite effect and the proportion of cells in the S phase increased significantly. In vivo models confirmed the inhibitory role of Id1b in primary tumor growth and metastasis. Through microarray analysis we found that the cancer stem cell (CSC) markers ALDH1A1 and Notch-1 were up-regulated specifically in Id1b-overexpressing cells. By using qPCR we also found overexpression of Sca-1, Tert, Sox-2 and Oct-4 in these cells. Increased levels of Id1b promoted self-renewal and CSC-like properties, as shown by their high capacity for developing secondary tumorspheres and retaining the PKH26 dye. The acquisition of CSC phenotype was confirmed in human PC-3 cells that overexpressed Id1b. Our results show that Id1b maintains cells in a quiescent state and promotes self-renewal and CSC-like features. On the contrary, Id1a promotes cell proliferation.


Asunto(s)
Empalme Alternativo , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Neoplasias del Colon/patología , Proteína 1 Inhibidora de la Diferenciación/genética , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Animales , Apoptosis , Western Blotting , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Ciclo Celular , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Humanos , Técnicas para Inmunoenzimas , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Isoformas de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncotarget ; 6(29): 27288-303, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26318423

RESUMEN

The spread of lung cancer cells to distant sites represents a common event associated with poor prognosis. A fraction of tumor cells named cancer stem cells (CSCs) have the ability to overcome therapeutic stress and remain quiescent. However, whether these CSCs have also the capacity to initiate and sustain metastasis remains unclear. Here, we used tumor sphere cultures (TSC) isolated from mouse and human lung cancer models to enrich for CSCs, and assessed their metastatic potential as compared to non-CSCs. As expected, TSC overexpressed a variety of stem cell markers and displayed chemoresistance. The CSC phenotype of TSC was confirmed by their higher growth ability in soft agar and tumorigenic potential in vivo, despite their reduced in vitro cell growth kinetics. Surprisingly, the appearance of spontaneous lung metastases was strongly delayed in mice injected with TSC as compared to non-TSC cells. Similarly, this finding was confirmed in several other models of metastasis, an effect associated with a retarded colonization activity. Interestingly, such delay correlated with a quiescent phenotype whose underlined mechanisms included an increase in p27 protein and lower phospho-ERK1/2 levels. Thus, these data suggest that cells enriched for CSC properties display an impaired metastatic activity, a finding with potential clinical implications.


Asunto(s)
Metástasis de la Neoplasia , Células Madre Neoplásicas/citología , Esferoides Celulares/citología , Agar/química , Animales , Antineoplásicos/uso terapéutico , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Osteólisis , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Br J Pharmacol ; 137(6): 881-91, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12411420

RESUMEN

1. Acute moderate hypoxia modifies the catalytic activity and expression of certain isoenzymes of hepatic cytochrome P450 (P450). The aim of this study was to document whether hypoxia affects hepatic P450 directly or through the release of serum mediators. 2. Rabbits were subjected to a FiO(2) of 8% for 48 h, sacrificed, and serum and hepatocytes were isolated; hepatocytes from control and rabbits with hypoxia were incubated with serum from control and hypoxic rabbits for 4 and 24 h, and total P450 content, CYP1A1, 1A2 and 3A6 activities and expressions were assessed. Sera were fractionated by size exclusion chromatography and fractions tested for their ability to modify activity and amount of P450, and serum mediators were identified through neutralization experiments. 3. Total serum and fractions with proteins of 15-23 and 65-94 kDa of M(r) reduced P450 content and expression of CYP1A1, 1A2 and 3A6, as well as CYP1A1, 1A2 and 3A6 mRNA. Total serum and the fraction with 32-44 kDa proteins increased CYP3A6 activity and protein and mRNA. The serum mediators implicated in the decrease in activity and expression of CYP1A1, 1A2 and 3A6 were interferon-gamma (IFN-gamma), interleukin-1beta (IL-1beta) and IL-2. Erythropoietin (Epo) was partly responsible for the increase in P450 content and CYP3A6 expression. 4. In conclusion, acute moderate hypoxia diminishes the activity and expression of CYP1A1, 1A2 and CYP1A1, 1A2 mRNA, and increases CYP3A6 protein, activity and CYP3A6 mRNA. Several mechanisms contribute to these changes in P450, among them the release of cytokines acting as serum mediators.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Proteínas Sanguíneas/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Hipoxia/fisiopatología , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/farmacología , Northern Blotting , Western Blotting , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Eritropoyetina/sangre , Eritropoyetina/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Interferón gamma/sangre , Interferón gamma/farmacología , Interleucina-1/sangre , Interleucina-1/farmacología , Interleucina-2/sangre , Interleucina-2/farmacología , Hígado/enzimología , Masculino , Peso Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Regulación hacia Arriba/efectos de los fármacos
19.
Biochem Pharmacol ; 68(3): 573-82, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15242823

RESUMEN

Incubation of serum from rabbits with a turpentine-induced inflammatory reaction and from humans with an upper respiratory viral infection with hepatocytes from rabbits with a turpentine-induced inflammatory reaction for 4h reduces total cytochrome P450 content and activity of cytochrome P450 isoforms CYP1A1/1A2 and 3A6 without affecting the expression of these proteins. To document the signal transduction pathways implicated in the decrease in CYP1A1/1A2 and 3A6 activity, hepatocytes from rabbits with a turpentine-induced inflammatory reaction were incubated with serum from rabbits with a turpentine-induced inflammatory reaction, serum from individuals with a viral infection and interleukin-6 for 4h in presence of inhibitors of protein kinases. The sera-induced decrease in CYP1A1/1A2 and 3A6 activity was partially prevented by the inhibition of Janus-associated protein tyrosine kinase, double-stranded RNA-dependent protein kinase, protein kinase C, and p42/44 mitogen-activated protein kinase. The serum from rabbits with a turpentine-induced inflammatory reaction increased the phosphorylation of Erk1/2, effect prevented by PD98059 but not by bis-indolylmaleimide, a specific inhibitor of protein kinase C. The results demonstrated that the decrease in total cytochrome P450 content and in CYP1A1/1A2 and 3A6 activity by sera and interleukin-6 involves the activation of protein tyrosine kinases, p42/44 mitogen-activated protein kinase and protein kinase C. Indirect evidence supported that nitric oxide is implicated in the decrease in activity of these enzymes.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Plasma/enzimología , Transducción de Señal/fisiología , Animales , Proteínas Portadoras/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/enzimología , Interleucina-6/farmacología , Masculino , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Plasma/efectos de los fármacos , Conejos , Trementina
20.
Cell Commun Signal ; 1(1): 1, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-14606958

RESUMEN

The CCN family of genes consists presently of six members in human (CCN1-6) also known as Cyr61 (Cystein rich 61), CTGF (Connective Tissue Growth Factor), NOV (Nephroblastoma Overexpressed gene), WISP-1, 2 and 3 (Wnt-1 Induced Secreted Proteins). Results obtained over the past decade have indicated that CCN proteins are matricellular proteins, which are involved in the regulation of various cellular functions, such as proliferation, differentiation, survival, adhesion and migration. The CCN proteins have recently emerged as regulatory factors involved in both internal and external cell signaling. CCN3 was reported to physically interact with fibulin-1C, integrins, Notch and S100A4. Considering that, the conformation and biological activity of these proteins are dependent upon calcium binding, we hypothesized that CCN3 might be involved in signaling pathways mediated by calcium ions.In this article, we review the data showing that CCN3 regulates the levels of intracellular calcium and discuss potential models that may account for the biological effects of CCN3.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA