Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Vis ; 19: 2596-605, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24379647

RESUMEN

PURPOSE: The aim of this study was to develop and characterize a new contact lens-associated fungal keratitis rat model and to assess the ability of non-invasive spectral-domain optical coherence tomography (SD-OCT) to detect pathological changes in vivo in fungal keratitis. METHODS: We used SD-OCT to image and measure the cornea of Sprague Dawley rats. Fusarium infection was initiated in the rat eye by fitting Fusarium solani-soaked contact lenses on the experimental eye, while the control animals received contact lenses soaked in sterile saline. The fungal infection was monitored with periodic slit-lamp examination and in vivo SD-OCT imaging of the rat eye, and confirmed by histology, counting of viable fungi in the infected rat cornea, and PCR with specific primers for Fusarium sp. RESULTS: We imaged and measured the rat cornea with SD-OCT. Custom-made contact lenses were developed based on the OCT measurements. Incubation of contact lenses in a F. solani suspension resulted in biofilm formation. We induced contact lens-associated Fusarium keratitis by fitting the rat eyes for 4 h with the Fusarium-contaminated contact lenses. The SD-OCT images of the cornea correlated well with the slit-lamp and histopathological results and clearly defined clinical signs of infection, namely, increased corneal thickening, loss of epithelial continuity, hyper-reflective areas representing infiltrates, and endothelial plaques characteristic of fungal infection. Moreover, in three cases, SD-OCT detected the infection without any clear findings on slit-lamp examination. Infection was confirmed with histological fungal staining, PCR, and microbiological culture positivity. CONCLUSIONS: We developed a highly reproducible rat contact lens model and successfully induced contact lens-associated Fusarium keratitis in this model. The clinical presentation of contact lens-associated Fusarium keratitis in the rat model is similar to the human condition. SD-OCT is a valuable tool that non-invasively revealed characteristic signs of the fungal infection and could provide sensitive, objective monitoring in fungal keratitis.


Asunto(s)
Lentes de Contacto/microbiología , Infecciones Fúngicas del Ojo/patología , Fusariosis/patología , Queratitis/patología , Animales , Soluciones para Lentes de Contacto , Modelos Animales de Enfermedad , Infecciones Fúngicas del Ojo/diagnóstico , Infecciones Fúngicas del Ojo/microbiología , Femenino , Fusariosis/diagnóstico , Fusariosis/microbiología , Fusarium/patogenicidad , Fusarium/fisiología , Humanos , Queratitis/diagnóstico , Queratitis/microbiología , Ratas , Ratas Sprague-Dawley , Tomografía de Coherencia Óptica
2.
Cancer Res ; 66(15): 7429-37, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16885338

RESUMEN

High-grade gliomas are devastating brain tumors associated with a mean survival of <50 weeks. Two of the most common genetic changes observed in these tumors are overexpression/mutation of the epidermal growth factor receptor (EGFR) vIII and loss of PTEN/MMAC1 expression. To determine whether somatically acquired EGFRvIII expression or Pten loss accelerates high-grade glioma development, we used a previously characterized RasB8 glioma-prone mouse strain, in which these specific genetic changes were focally introduced at 4 weeks of age. We show that both postnatal EGFRvIII expression and Pten inactivation in RasB8 mice potentiate high-grade glioma development. Moreover, we observe a concordant loss of Pten and EGFR overexpression in nearly all high-grade gliomas induced by either EGFRvIII introduction or Pten inactivation. This novel preclinical model of high-grade glioma will be useful in evaluating brain tumor therapies targeted to the pathways specifically dysregulated by EGFR expression or Pten loss.


Asunto(s)
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Glioma/genética , Fosfohidrolasa PTEN/deficiencia , Animales , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Cloruros , Medios de Contraste , Modelos Animales de Enfermedad , Receptores ErbB/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioma/enzimología , Glioma/patología , Imagen por Resonancia Magnética , Compuestos de Manganeso , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Fosfohidrolasa PTEN/genética
3.
Invest Ophthalmol Vis Sci ; 48(6): 2476-82, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17525173

RESUMEN

PURPOSE: The aim of this study was to quantify tumor cell proliferation and growth, analyze tumor blood vessel development, and determine the efficacy of antiangiogenic and angiostatic therapy in targeting mature vessels in retinal tumors of the LH(BETA)T(AG) mouse model for retinoblastoma. METHODS: LH(BETA)T(AG) mouse retinas were analyzed at 4, 8, 12, and 16 weeks of age. Tumor burden was analyzed by histology; cell proliferation, vessel density, angiogenesis, and vessel maturation were detected by immunofluorescence. To assess the efficacy of mature vessel targeting, 16-week-old mice were treated with single subconjunctival injections of the selective vascular-targeting drug combretastatin A4 prodrug (CA4P) or anecortave acetate, and eyes were analyzed 1 day and 1 week after injection to determine microvessel density and the number of angiogenic and mature vessels. RESULTS: Increased cell proliferation and angiogenesis were detected in the retinal inner nuclear layer (INL) before morphologic neoplastic changes were evident. As tumor size increased, angiogenesis diminished concomitantly with the appearance of mature vessels. Treatment with CA4P and anecortave acetate resulted in significant reductions in total vessel density. However, neither drug reduced the amount of alpha-smooth muscle actin (SMA)-positive, mature vessels. CONCLUSIONS: Results of this study provide new insight into the relationship between tumor growth and blood vessel development in the LH(BETA)T(AG) mouse and establish the framework for defining the selective action of two vessel-targeting drugs against new blood vessels compared with mature blood vessels. These findings suggest a high potential value in targeting the process of angiogenesis in the treatment of children with retinoblastoma.


Asunto(s)
Modelos Animales de Enfermedad , Neovascularización Patológica/patología , Neoplasias de la Retina/irrigación sanguínea , Retinoblastoma/irrigación sanguínea , Actinas/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Bibencilos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Endoglina , Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Pericitos/metabolismo , Pregnadienodioles/uso terapéutico , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/metabolismo , Retinoblastoma/patología , Estilbenos/uso terapéutico
4.
Cancer Res ; 63(24): 8573-7, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695164

RESUMEN

Whereas biallelic neurofibromatosis 1 (NF1) inactivation is observed in NF1-associated gliomas, astrocyte-restricted Nf1 conditional knockout mice do not develop gliomas. These observations suggest that NF1 glioma formation requires additional cellular or genetic conditions. To determine the effect of an Nf1 heterozygous brain environment on NF1 glioma formation, we generated Nf1+/- mice lacking Nf1 expression in astrocytes. In contrast to astrocyte-restricted Nf1 conditional knockout mice, Nf1+/- mice lacking Nf1 in astrocytes develop optic nerve gliomas. This mouse model demonstrates that Nf1+/- cells contribute to the pathogenesis of gliomas in NF1 and provides a tool for the preclinical evaluation of potential therapeutic interventions for these tumors.


Asunto(s)
Encéfalo/fisiología , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes de Neurofibromatosis 1/fisiología , Neurofibromatosis 1/genética , Glioma del Nervio Óptico/genética , Animales , Astrocitos/patología , Astrocitos/fisiología , Encéfalo/metabolismo , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/patología , Glioma del Nervio Óptico/patología
5.
Oncogene ; 21(25): 4050-9, 2002 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-12037687

RESUMEN

Tuberous sclerosis complex (TSC) is an autosomal dominant tumor predisposition syndrome characterized by benign proliferations (hamartomas). In the brain, individuals with TSC develop autism, mental retardation and seizures associated with focal cortical dysplasias, subependymal nodules, and subependymal giant cell astrocytomas (SEGAs). We hypothesize that dysregulated astrocyte function due to mutations in the tumor suppressor genes, TSC1 and TSC2, may contribute to the pathogenesis of these brain abnormalities. In this report, we demonstrate that mice heterozygous for a targeted defect in either the Tsc1 or Tsc2 genes(Tsc1+/- and Tsc2+/- mice) exhibit a 1.5-fold increase in the number of astrocytes in vivo. Whereas increased astrocyte numbers in vivo were suggestive of a proliferative advantage, Tsc2+/- primary astrocyte cultures did not show a cell-autonomous growth advantage, anchorage-independent growth, increased saturation density, or increased fluid-phase endocytosis compared to wild type astrocytes. Tsc2 null mouse embryonic fibroblasts (MEFs) however, did exhibit increased saturation density compared to Tsc2 wild type controls. In both Tsc2+/- astrocytes and Tsc2 null mouse embryonic fibroblasts, p27-Kip1 expression was decreased compared to wild type cells, and was reversed by tuberin re-expression in Tsc2-/- MEFs. In contrast, no change in endocytosis was observed upon tuberin re-expression in Tsc2-/- MEFs. Collectively, these results suggest Tsc heterozygosity may provide a non-cell-autonomous growth advantage for astrocytes that may involve p27-Kip1 expression.


Asunto(s)
Astrocitos/patología , Proteínas de Ciclo Celular/metabolismo , Proteínas/genética , Proteínas Represoras/genética , Esclerosis Tuberosa/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , División Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Endocitosis , Fibroblastos/metabolismo , Genes Supresores de Tumor , Heterocigoto , Pérdida de Heterocigocidad , Ratones , Ratones Noqueados , Proteínas/metabolismo , Proteínas Represoras/metabolismo , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa
6.
Invest Ophthalmol Vis Sci ; 51(6): 2860-4, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20107171

RESUMEN

PURPOSE: Gelatinases, matrix metalloproteinase (MMP)-2, and MMP-9 are known for their importance in angiogenesis and tumor biology. The purpose of this study was to test the hypothesis that anecortave acetate (AA) decreases transgenic retinoblastoma (RB) tumor burden by modulating gelatinase activity. METHODS: To assess the possible gelatinase modulation after AA treatment, a single subconjunctival injection of AA (300 microg) was delivered to the right eyes of 10-week-old LH(BETA)T(AG) mice. Eyes were evaluated for gelatinase expression and activity by gel and in situ zymography at 24 hours, 48 hours, and 1 week after treatment. RESULTS: Gel zymography of whole eye extracts and in situ zymography of retinal tumors showed strong gelatinase expression and activity within transgenic RB tumors. AA treatment in RB transgenic mice resulted in a significant decrease of gelatinase activity 1 week after AA treatment. Surprisingly, there was an initial transient upregulation of MMP-9 activity in whole eye extracts at 24 and 48 hours after AA treatment in both LH(BETA)T(AG) transgenic and wild-type mice. This increase was not observed in the tumors. CONCLUSIONS: As suggested by our data, inhibition of gelatinase activity appears to be a mechanism of action of AA. AA treatment results in a decrease in gelatinase activity that correlates with the significant decrease in tumor burden shown by the authors' previous studies. However, the significance of the initial, transient upregulation of gelatinase by AA injection is unknown, and further studies are warranted. Combining antiangiogenic agents with multiple mechanisms of action has the potential to enhance RB tumor control.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Modelos Animales de Enfermedad , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Pregnadienodioles/uso terapéutico , Neoplasias de la Retina/tratamiento farmacológico , Retinoblastoma/tratamiento farmacológico , Animales , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Transgénicos , Neoplasias de la Retina/enzimología , Neoplasias de la Retina/patología , Retinoblastoma/enzimología , Retinoblastoma/patología , Regulación hacia Arriba
7.
Ann Neurol ; 57(1): 119-27, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15622533

RESUMEN

Children affected with the inherited tumor predisposition syndrome, neurofibromatosis 1 (NF1), are prone to the development of low-grade astrocytic optic pathway tumors (optic pathway glioma [OPG]). Previously, we developed a model of NF1-associated astrocytoma (GFAPCre; Nf1(flox/mut) mice) in which mice develop optic nerve and chiasm glioma. To define the molecular pathogenesis of OPG, we used this mouse model to study the natural history of OPG formation using immunohistological and radiographic approaches. We observed that whereas astrocyte hyperplasia is present in the optic nerves associated with gross optic nerve thickening at 3 weeks of age, overt neoplastic changes were not seen until 2 months of age. Astrocyte proliferation was maximal between 3 weeks and 2 months of age, suggesting that the most rapid period of growth occurs early. Mouse OPG tumors were detected by magnetic resonance imaging at 2 months of age and exhibited contrast enhancement, as seen in human OPG. In addition, the mouse OPG tumors exhibited expression of proteins associated with astroglial progenitors, including nestin and brain lipid binding protein. Last, we observed neovascularization and microglial cell infiltration by 3 weeks of age before overt neoplastic transformation, suggesting that these cellular changes participate in the early stages of tumor formation.


Asunto(s)
Glioma/complicaciones , Neurofibromatosis 1/complicaciones , Neoplasias del Nervio Óptico/complicaciones , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Vasos Sanguíneos/fisiología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioma/metabolismo , Inmunohistoquímica/métodos , Indoles/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Antígeno Ki-67/metabolismo , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neoplasias del Nervio Óptico/metabolismo , Coloración y Etiquetado/métodos , Factor de von Willebrand/inmunología
8.
Development ; 132(24): 5577-88, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314489

RESUMEN

The gene responsible for neurofibromatosis type 1 (NF1) encodes a tumor suppressor that functions as a negative regulator of the Ras proto-oncogene. Individuals with germline mutations in NF1 are predisposed to the development of benign and malignant tumors of the peripheral and central nervous system (CNS). Children with this disease suffer a high incidence of optic gliomas, a benign but potentially debilitating tumor of the optic nerve; and an increased incidence of malignant astrocytoma, reactive astrogliosis and intellectual deficits. In the present study, we have sought insight into the molecular and cellular basis of NF1-associated CNS pathologies. We show that mice genetically engineered to lack NF1 in CNS exhibit a variety of defects in glial cells. Primary among these is a developmental defect resulting in global reactive astrogliosis in the adult brain and increased proliferation of glial progenitor cells leading to enlarged optic nerves. As a consequence, all of the mutant optic nerves develop hyperplastic lesions, some of which progress to optic pathway gliomas. These data point to hyperproliferative glial progenitors as the source of the optic tumors and provide a genetic model for NF1-associated astrogliosis and optic glioma.


Asunto(s)
Encéfalo/metabolismo , Neurofibromina 1/fisiología , Neuroglía/metabolismo , Glioma del Nervio Óptico/metabolismo , Células Madre/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Hiperplasia , Ratones , Ratones Noqueados , Mutación , Neurofibromina 1/genética , Neuroglía/patología , Nervio Óptico/patología , Glioma del Nervio Óptico/genética , Glioma del Nervio Óptico/patología
9.
Ann Neurol ; 52(3): 285-96, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12205640

RESUMEN

Persons affected with tuberous sclerosis complex (TSC) develop a wide range of neurological abnormalities including aberrant neuronal migration and seizures. In an effort to model TSC-associated central nervous system abnormalities in mice, we generated two independent lines of astrocyte-specific Tsc1 conditional knockout mice by using the Cre-LoxP system. Astrocyte-specific Tsc1-null mice exhibit electroencephalographically proven seizures after the first month of age and begin to die at 3 to 4 months. Tsc1-null mice show significant increases in astrocyte numbers throughout the brain by 3 weeks of age and abnormal neuronal organization in the hippocampus between 3 and 5 weeks. Moreover, cultured Tsc1-null astrocytes behave similar to wild-type astrocytes during log phase growth but demonstrate increased saturation density associated with reduced p27(Kip1) expression. Collectively, our results demonstrate that astrocyte-specific disruption of Tsc1 in mice provides a context-dependent growth advantage for astrocytes that results in abnormalities in neuronal organization and epilepsy.


Asunto(s)
Astrocitos/patología , Epilepsia/patología , Hipocampo/anomalías , Neuronas/patología , Proteínas/genética , Animales , Astrocitos/química , Proteínas de Ciclo Celular/genética , División Celular , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Electroencefalografía , Epilepsia/genética , Gliosis/patología , Hipocampo/patología , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas/análisis , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA