Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 292(35): 14381-14390, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684419

RESUMEN

To examine the biochemical influences that may contribute to the success of gene therapy for ocular disorders, the role of versican, a vitreous component, in adenoviral-mediated transgene expression was examined. Versican is a large chondroitin sulfate-containing, hyaluronic acid-binding proteoglycan present in the extracellular matrix and in ocular vitreous body. Y79 retinoblastoma cells and CD44-negative SK-N-DZ neuroblastoma cells transduced with adenoviral vectors in the presence of versican respond with an activation of transgene expression. Proteolysis of versican generates a hyaluronan-binding G1 domain. The addition of recombinant versican G1 to SK-N-DZ cells results in a similar activation of transgene expression, and treatment with dasatinib, an inhibitor of Src family kinases, also mimics the effects of versican. Enhancement is accompanied by an increase in signal transducer and activator of transcription 5 (STAT5) phosphorylation and is abrogated by treatment with C188-9, a STAT3/5 inhibitor, or with ruxolitinib, a Janus kinase 1/2 (JAK1/2) inhibitor. These data implicate versican G1 in enhancing adenoviral vector transgene expression in a hyaluronic acid-CD44 independent manner that is down-regulated by inhibitors of the JAK/STAT pathway and enhanced by inhibitors of the Src kinase pathway.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/terapia , Inhibidores de Proteínas Quinasas/farmacología , Versicanos/metabolismo , Adenoviridae/crecimiento & desarrollo , Adenoviridae/fisiología , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , ADN Recombinante/metabolismo , ADN Viral/metabolismo , Genes Reporteros/efectos de los fármacos , Vectores Genéticos , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/virología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción STAT/antagonistas & inhibidores , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Versicanos/química , Versicanos/genética , Replicación Viral/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
3.
J Biol Chem ; 287(39): 32697-707, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22865879

RESUMEN

The success of gene therapy in the ocular environment is partly due to the presence of hyaluronan in vitreous. Here we explore the mechanism of hyaluronan-mediated enhancement of adenoviral vector transgene expression. Introduction of hyaluronan receptor CD44 into CD44-negative cells followed by transduction in the presence of vitreous with an adenoviral vector containing an IL-12-coding transgene increases IL-12 secretion. We demonstrate that sequential CD44 proteolysis is responsible for hyaluronan-mediated enhancement. Metalloproteinase or γ-secretase inhibitors decrease adenoviral-mediated transgene expression. Deletion of these proteolytic sites in CD44 also inhibits transgene expression. Expression of CD44 with a mutation to prevent phosphorylation of serine 325 inhibits the response to vitreous. Expression of the CD44 intracellular domain enhances transgene expression in the absence of vitreous. CD44-mediated enhancement of gene expression was observed with vectors using different promoters and appears because of an increase in mRNA production, not because of an increase in vector transduction as determined by quantitative RT-PCR and quantitative PCR, respectively. These data fit a model where the interaction of hyaluronan in vitreous and CD44 modulates transgene expression by initiating CD44 proteolysis and release of the cytoplasmic domain, resulting in increased transgene transcription.


Asunto(s)
Adenoviridae , Expresión Génica , Vectores Genéticos , Receptores de Hialuranos/metabolismo , Proteolisis , Transgenes , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Células COS , Chlorocebus aethiops , Inhibidores Enzimáticos/farmacología , Humanos , Receptores de Hialuranos/genética , Ácido Hialurónico/genética , Ácido Hialurónico/metabolismo , Interleucina-12/biosíntesis , Interleucina-12/genética , Mutación , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Transcripción Genética
4.
Mol Ther ; 18(10): 1885-90, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20606647

RESUMEN

The ocular environment has been shown to induce tolerance to locally administered antigens. We therefore investigated whether there was a systemic immune response against adenoviral vectors injected into the vitreous of retinoblastoma patients enrolled in a phase 1 clinical trial of adenoviral-mediated thymidine kinase gene transfer. Sections of enucleated eyes were immunostained with antibodies against inflammatory cells. A trend toward increasing numbers of plasma cells, T cells, macrophages, and antigen-presenting cells was observed in the injected subjects' eyes, but systemically, there was no significant increase in the number of adenovirus-specific cytotoxic T lymphocytes (CTLs) or in adenovirus neutralizing antibodies. Therefore, in contrast to studies showing significant immunogenicity of Ad-RSVtk following injection into extraocular tumors, injection into the eye produces only a mild local inflammatory response without evidence of systemic cellular or humoral immune responses to adenovirus.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Retinoblastoma/inmunología , Retinoblastoma/terapia , Células Presentadoras de Antígenos/inmunología , Ojo/inmunología , Ojo/metabolismo , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Macrófagos/inmunología , Retinoblastoma/genética , Retinoblastoma/metabolismo , Linfocitos T/inmunología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
5.
Curr Urol Rep ; 12(2): 166-72, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21243456

RESUMEN

Since the first part of the 20th century, when physicians and surgeons were struggling to understand disorders of sex development (DSD) and the variety of associated anomalies they produced, there have been great strides in feminizing surgery for patients with DSD. Surgical techniques were devised and concepts about how best to manage these complicated patients were developed. In this article, the evolution of these concepts and refinements of technique for feminizing genitoplasty that have taken place over the past three quarters of a century are presented, and the surgical outcomes and complications of feminizing genitoplasty are critically reviewed.


Asunto(s)
Trastornos del Desarrollo Sexual/cirugía , Genitales/cirugía , Procedimientos Quirúrgicos Urogenitales/métodos , Femenino , Genitales/anomalías , Humanos , Masculino , Resultado del Tratamiento
6.
J Urol ; 184(1): 305-10, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20488468

RESUMEN

PURPOSE: The combination of trimethoprim/sulfamethoxazole is often used to treat uncomplicated urinary tract infections in children. The rationale for combining trimethoprim and sulfamethoxazole is that they may act synergistically to increase antibacterial activity. However, approximately 3% of patients show allergic reactions to sulfamethoxazole, of which some are serious (liver failure and Stevens-Johnson syndrome). We determined whether adding sulfamethoxazole is necessary to increase in vitro antibacterial activity for pediatric urinary tract infection compared to that of trimethoprim alone. MATERIALS AND METHODS: We prospectively identified 1,298 children with urinary tract infection (greater than 100,000 cfu/ml Escherichia coli) from a total of 4 American regions. In vitro susceptibility of bacterial isolates to sulfamethoxazole, trimethoprim and trimethoprim/sulfamethoxazole was determined using disk diffusion. Ampicillin susceptibility was tested at 2 sites. At 1 site all uropathogens from consecutive urinary isolates were evaluated. RESULTS: E. coli susceptibility to trimethoprim was 70%, comparable to the 70% of trimethoprim/sulfamethoxazole (p = 0.9) and higher than the 56.9% of sulfamethoxazole (p <0.05). This susceptibility pattern was without regional differences. At 2 sites susceptibility to trimethoprim was significantly higher than to ampicillin. At 1 site the susceptibility of other uropathogens to trimethoprim and trimethoprim/sulfamethoxazole was similar to that of E. coli. CONCLUSIONS: In children with urinary tract infection in vitro susceptibility to trimethoprim was comparable to that to trimethoprim/sulfamethoxazole and significantly higher than to sulfamethoxazole. This finding was similar at all sites. Adding sulfamethoxazole appears unnecessary and may represent a risk to patients. Trimethoprim can be used as an alternative to trimethoprim/sulfamethoxazole based on in vitro antibacterial susceptibility. Routine trimethoprim/sulfamethoxazole use for urinary tract infection should be carefully reevaluated.


Asunto(s)
Antiinfecciosos Urinarios/uso terapéutico , Sulfametoxazol/uso terapéutico , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Trimetoprim/uso terapéutico , Infecciones Urinarias/tratamiento farmacológico , Ampicilina/uso terapéutico , Análisis de Varianza , Distribución de Chi-Cuadrado , Niño , Preescolar , Combinación de Medicamentos , Quimioterapia Combinada , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Humanos , Lactante , Masculino , Pruebas de Sensibilidad Microbiana , Estudios Prospectivos , Resultado del Tratamiento , Estados Unidos , Infecciones Urinarias/microbiología
7.
PLoS One ; 15(7): e0236175, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32697798

RESUMEN

Adenoviruses cause upper respiratory infections, conjunctivitis, keratitis, and gastrointestinal illness. These can be fatal in immunocompromised individuals. Adenoviruses have also been engineered into viral vectors to deliver therapeutic genes or induce immunity as vaccine carriers. The success of ocular gene therapy is driven partly by the immunologic and biochemical influences of the intraocular environment. We have shown that versican and hyaluronan modulate adenoviral vector transgene expression through CD44 signaling. Herein we explored the role of these pathways on virus replication and viral protein expression of wild type adenovirus. We report that the addition of vitreous humor (which contains both versican and hyaluronan) increases viral hexon protein levels. Vitreous humor also increased wild type adenovirus DNA replication in vitro. Metalloproteinase and γ-secretase inhibitors, which inhibit CD44 proteolytic activation, blocked adenoviral replication in vitro. Similarly, protein kinase C and RhoA kinase inhibitors, both proteins associated with CD44 mediated pathways, also inhibited wild type adenoviral replication in vitro. Application of metalloproteinase and γ-secretase inhibitors to human conjunctival explants sharply decreased adenoviral vector gene expression. Our results demonstrate that pharmacologic delivery of these inhibitors is easily achievable. The inhibition of these enzymes should be explored as potential therapies of wild type adenoviral infections.


Asunto(s)
Infecciones por Adenoviridae/tratamiento farmacológico , Adenoviridae/efectos de los fármacos , Antivirales/farmacología , Vectores Genéticos/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Adenoviridae/fisiología , Infecciones por Adenoviridae/virología , Administración Oftálmica , Amidas/farmacología , Amidas/uso terapéutico , Antivirales/uso terapéutico , Conjuntiva/metabolismo , ADN Viral/genética , ADN Viral/aislamiento & purificación , Diaminas/farmacología , Diaminas/uso terapéutico , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/fisiología , Células HeLa , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Maleimidas/farmacología , Maleimidas/uso terapéutico , Metaloproteasas/antagonistas & inhibidores , Metaloproteasas/metabolismo , Permeabilidad , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteolisis/efectos de los fármacos , Piridinas/farmacología , Piridinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tiazoles/farmacología , Tiazoles/uso terapéutico , Versicanos/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Cuerpo Vítreo/metabolismo , Quinasas Asociadas a rho/metabolismo
9.
Mol Ther ; 15(3): 566-70, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17180120

RESUMEN

Adenovirus infection is a significant cause of ocular, respiratory, and gastrointestinal illness and can spread rapidly. Morbidity is considerable in immune-suppressed individuals and there is significant mortality. There are no effective therapies. During preclinical studies of adenoviral-mediated gene therapy for ocular disorders, we noticed a significant increase in transduction when the target cells were exposed to adenovirus in the presence of ocular vitreous. The vitreous is mainly comprised of water, collagen, and the large polysaccharide hyaluronan. In this paper, we report data that implicate hyaluronan in the adenoviral infectious process and show that interference with the interaction between hyaluronan and its cellular receptor CD44 can block adenovirus transduction in vitro and in vivo.


Asunto(s)
Adenoviridae/genética , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Transducción Genética , Animales , Línea Celular , Vectores Genéticos/genética , Humanos , Receptores de Hialuranos/genética , Ratones , Ratones Endogámicos C57BL , Mutación/genética
11.
J Clin Oncol ; 23(31): 7927-35, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16258092

RESUMEN

PURPOSE: To evaluate the feasibility and safety of adenovirus-mediated gene therapy as a treatment for tumor seeds in the vitreous of children with retinoblastoma. PATIENTS AND METHODS: An Institutional Biosafety Committee-, Institutional Review Board-, Recombinant DNA Advisory Committee-, and US Food and Drug Administration-approved phase I study used intrapatient dose escalation of adenoviral vector containing a herpes simplex thymidine kinase gene (AdV-TK) followed by systemic administration of ganciclovir to treat bilateral retinoblastoma with vitreous tumor seeding refractory to standard therapies. Vitreous tumor seeds were treated by intravitreous injection of AdV-TK adjacent to disease sites. Each injection was followed by ganciclovir delivered intravenously every 12 hours for 7 days. RESULTS: Eight patients with vitreous tumor seeds were enrolled. One patient who was treated with 10(8) viral particles (vp) had resolution of the tumor seeds around the injection site. The seven patients who were treated with doses > or = 10(10) vp had resolution of their vitreous tumor seeds documented by fundoscopy. Toxicity included mild inflammation at 10(10) vp and moderate inflammation, corneal edema, and increased intraocular pressure at 10(11) vp. One patient was free of active vitreous tumor seeds 38 months after therapy. There has been no evidence of extraocular spread of tumor along the needle tract in any patient. CONCLUSION: AdV-TK followed by ganciclovir can be administered safely to children with retinoblastoma. Suicide gene therapy may contribute to the treatment of children with retinoblastoma tumor seeds in the vitreous, a resistant complication of retinoblastoma.


Asunto(s)
Adenovirus Humanos/genética , Antivirales/uso terapéutico , Ganciclovir/uso terapéutico , Siembra Neoplásica , Neoplasias de la Retina/terapia , Retinoblastoma/terapia , Timidina Quinasa/uso terapéutico , Cuerpo Vítreo/patología , Niño , Terapia Combinada , Estudios de Factibilidad , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Humanos , Recurrencia Local de Neoplasia/enzimología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/terapia , Neoplasias de la Retina/enzimología , Neoplasias de la Retina/patología , Retinoblastoma/enzimología , Retinoblastoma/patología , Timidina Quinasa/genética , Cuerpo Vítreo/efectos de los fármacos
13.
Expert Rev Mol Med ; 5(1): 1-14, 2003 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-14987394

RESUMEN

Retinoblastoma (Rb) is the most common primary ocular malignancy of children and is caused by a mutation in the gene RB1. Approximately 40% of cases are associated with one or more constitutional mutations, and are therefore heritable, whereas the other 60% are sporadic. Rb is exclusively found in young children. In some cases, Rb tumours metastasise to extraocular organs including bone, lung and brain. Although there is no effective treatment for metastatic disease, non-metastatic cases can be cured by removal of the eye(enucleation). Newer treatment strategies emphasise salvaging the affected eye whenever possible. Animal models of Rb have been developed with xenograft and transgenic techniques. Each model has both strengths and weaknesses for exploring the mechanisms of disease development and progression and the efficacy of new treatment strategies.


Asunto(s)
Retinoblastoma/terapia , Animales , Niño , Modelos Animales de Enfermedad , Ojo/patología , Ojo/trasplante , Enucleación del Ojo , Humanos , Trasplante de Órganos/métodos , Retinoblastoma/diagnóstico , Retinoblastoma/epidemiología
14.
Invest Ophthalmol Vis Sci ; 45(6): 1680-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15161826

RESUMEN

PURPOSE: A number of ocular diseases are potentially amenable to gene therapy interventions if appropriate vectors for the targeted administration of therapeutic genes can be identified. In vitro and in vivo transduction efficiency of a Group C serotype 5 adenoviral vector containing the fiber domain derived from a Group B serotype 35 adenovirus and the gene encoding green fluorescent protein (AdV5/F35-GFP) was compared to an AdV5-GFP vector for transgene delivery to human retinoblastoma and to human and murine retinas. METHODS: The distribution of the adenoviral receptors CAR and CD46 on normal and malignant retinal tissues was determined using immunohistochemistry. Human retinoblastoma cells were incubated with either AdV5-GFP or AdV5/F35-GFP, and the expression of the reporter protein was compared using quantitative fluorescence and fluorescent-activated cell sorting. Mice were given a single subretinal injection of either viral vector, and eyes were enucleated at specified times after injection for histopathologic examination. Human cadaver eyes were similarly examined ex vivo. RESULTS: CAR was expressed in retina except in photoreceptor outer segments. CD46 was expressed in photoreceptor inner and outer segments. Both vectors efficiently transduced the human retinoblastoma cells in vitro. However, the amount of the transgene expressed using AdV5/F35-GFP was more than sixfold greater than that when AdV5-GFP was used. In vivo, AdV5/F35-GFP at doses as low as 10(5) infectious units (IU) transduced cells in all layers of the retina especially photoreceptors and occasional neuronal cells, and Müller cells as well as retinal pigment epithelial cells, whereas AdV5-GFP transduced only retinal pigment epithelial cells and occasional photoreceptors and Müller cells. CONCLUSIONS: AdV5/F35 chimeric vectors may be superior to AdV5 for gene therapy applications targeting the photoreceptor.


Asunto(s)
Adenovirus Humanos/genética , Antígenos CD/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Glicoproteínas de Membrana/metabolismo , Receptores Virales/metabolismo , Retina/metabolismo , Retinoblastoma/metabolismo , Animales , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Citometría de Flujo , Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Técnicas para Inmunoenzimas , Integrina alfaV/metabolismo , Proteínas Luminiscentes/genética , Proteína Cofactora de Membrana , Ratones , Ratones Endogámicos C57BL , Retinoblastoma/patología , Células Tumorales Cultivadas
15.
J Urol ; 182(4 Suppl): 1734; discussion 1735, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19692088
17.
PLoS One ; 8(6): e63519, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826078

RESUMEN

Verification that cell lines used for cancer research are derived from malignant cells in primary tumors is imperative to avoid invalidation of study results. Retinoblastoma is a childhood ocular tumor that develops from loss of functional retinoblastoma protein (pRb) as a result of genetic or epigenetic changes that affect both alleles of the RB1 gene. These patients contain unique identifiable genetic signatures specifically present in malignant cells. Primary cultures derived from retinoblastoma tumors can be established as non-adherent tumorspheres when grown in defined media or as attached monolayers when grown in serum-containing media. While the RB1 genotypes of tumorspheres match those of the primary tumor, adherent cultures have the germline RB1 genotype. Tumorspheres derived from pRb-negative tumors do not express pRb and express the neuroendocrine tumor markers synaptophysin and microtubule-associated protein 2 (MAP2). Adherent cells are synaptophysin-negative and express pRb, the epithelial cell marker cytokeratin that is expressed in the retinal pigmented epithelium and the vascular endothelial cell marker CD34. While tumorspheres are of malignant origin, our results cast doubt on the assumption that adherent tumor-derived cultures are always valid in vitro models of malignant cells and emphasize the need for validation of primary tumor cultures.


Asunto(s)
Adhesión Celular , Neoplasias del Ojo/patología , Retinoblastoma/patología , Diferenciación Celular , Neoplasias del Ojo/genética , Genes de Retinoblastoma , Genotipo , Humanos , Mutación , Retinoblastoma/genética , Células Tumorales Cultivadas
18.
Neuro Oncol ; 15(9): 1173-85, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23658322

RESUMEN

BACKGROUND: Seneca Valley virus (SVV-001) is a nonpathogenic oncolytic virus that can be systemically administered and can pass through the blood-brain barrier. We examined its therapeutic efficacy and the mechanism of tumor cell infection in pediatric malignant gliomas. METHODS: In vitro antitumor activities were examined in primary cultures, preformed neurospheres, and self-renewing glioma cells derived from 6 patient tumor orthotopic xenograft mouse models (1 anaplastic astrocytoma and 5 GBM). In vivo therapeutic efficacy was examined by systemic treatment of preformed xenografts in 3 permissive and 2 resistant models. The functional role of sialic acid in mediating SVV-001 infection was investigated using neuraminidase and lectins that cleave or competitively bind to linkage-specific sialic acids. RESULTS: SVV-001 at a multiplicity of infection of 0.5 to 25 replicated in and effectively killed primary cultures, preformed neurospheres, and self-renewing stemlike single glioma cells derived from 4 of the 6 glioma models in vitro. A single i.v. injection of SVV-001 (5 × 10(12) viral particles/kg) led to the infection of orthotopic xenografts without harming normal mouse brain cells, resulting in significantly prolonged survival in all 3 permissive and 1 resistant mouse models (P < .05). Treatment with neuraminidase and competitive binding using lectins specific for α2,3-linked and/or α2,6-linked sialic acid significantly suppressed SVV-001 infectivity (P < .01). CONCLUSION: SVV-001 possesses strong antitumor activity against pediatric malignant gliomas and utilizes α2,3-linked and α2,6-linked sialic acids as mediators of tumor cell infection. Our findings support the consideration of SVV-001 for clinical trials in children with malignant glioma.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Picornaviridae , Animales , Neoplasias Encefálicas/metabolismo , Modelos Animales de Enfermedad , Glioma/metabolismo , Inyecciones Intravenosas , Ratones , Ratones SCID , Ácido N-Acetilneuramínico/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Invest Ophthalmol Vis Sci ; 53(7): 3454-62, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22562503

RESUMEN

PURPOSE: Human retinoblastomas form during the proliferative phase of retina development and are caused by mutations that result in absent or functionally defective Rb protein. Similar tumors occur in mice only when multiple Rb gene family members are absent. We asked if retinal tumors can arise from an undifferentiated retinal cell. The tumor-initiating cells isolated from these tumors that formed in early embryonic murine retinas were characterized. METHODS: Transgenic mice were created using a Pax6 promoter to target expression of SV40 large T-antigen (T-Ag) in the undifferentiated murine embryonic retina. T-Ag, which sequesters all Rb family proteins and p53, is expressed in the retina and lens by murine embryonic day 10 (E10) and tumors are observed by E12.5. A cell line that is adherent in serum-containing media and forms neurospheres in supplemented serum-free media was developed from retinal tumors isolated on postnatal day 7. RESULTS: In all, 1.5% of attached cells form neurospheres when transferred to serum-free medium. All cultured cells express T-Ag, confirming that they derive from the original tumors; 0.5% of adherent cells express detectable levels of CD133. CD133+ FACS-sorted cells cultured in serum-free medium form 3-fold more neurospheres than do CD133- cells. Six of seven mice injected with CD133+ cells and one of seven mice injected with CD133- cells formed tumors during a 6-month period. Unlike primary adherent cells, primary and secondary tumors heterogeneously express markers of stem cells and differentiation similar to human retinoblastoma. CONCLUSIONS: CD133+ tumor-initiating cells can originate from proliferating undifferentiated precursor cells.


Asunto(s)
Antígenos CD/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Transformación Celular Neoplásica/patología , Glicoproteínas/metabolismo , Péptidos/metabolismo , Retina/embriología , Neoplasias de la Retina/embriología , Retinoblastoma/embriología , Antígeno AC133 , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Citometría de Flujo , Proteínas de Homeodominio/genética , Humanos , Inmunofenotipificación , Ratones , Ratones Transgénicos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/metabolismo , Retinoblastoma/patología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
20.
J Vis Exp ; (54)2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21847079

RESUMEN

Culturing retinoblastoma tumor cells in defined stem cell media gives rise to primary tumorspheres that can be grown and maintained for only a limited time. These cultured tumorspheres may exhibit markedly different cellular phenotypes when compared to the original tumors. Demonstration that cultured cells have the capability of forming new tumors is important to ensure that cultured cells model the biology of the original tumor. Here we present a protocol for propagating human retinoblastoma tumors in vivo using Rag2(-/-) immune deficient mice. Cultured human retinoblastoma tumorspheres of low passage or cells obtained from freshly harvested human retinoblastoma tumors injected directly into the vitreous cavity of murine eyes form tumors within 2-4 weeks. These tumors can be harvested and either further passaged into murine eyes in vivo or grown as tumorspheres in vitro. Propagation has been successfully carried out for at least three passages thus establishing a continuing source of human retinoblastoma tissue for further experimentation.


Asunto(s)
Trasplante de Neoplasias/métodos , Neoplasias de la Retina/inmunología , Neoplasias de la Retina/patología , Retinoblastoma/inmunología , Retinoblastoma/patología , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/inmunología , Humanos , Ratones , Trasplante de Neoplasias/inmunología , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA