Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 30(10): 1331-1350.e11, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37802038

RESUMO

Mesial temporal lobe epilepsy (MTLE) is the most common focal epilepsy. One-third of patients have drug-refractory seizures and are left with suboptimal therapeutic options such as brain tissue-destructive surgery. Here, we report the development and characterization of a cell therapy alternative for drug-resistant MTLE, which is derived from a human embryonic stem cell line and comprises cryopreserved, post-mitotic, medial ganglionic eminence (MGE) pallial-type GABAergic interneurons. Single-dose intrahippocampal delivery of the interneurons in a mouse model of chronic MTLE resulted in consistent mesiotemporal seizure suppression, with most animals becoming seizure-free and surviving longer. The grafted interneurons dispersed locally, functionally integrated, persisted long term, and significantly reduced dentate granule cell dispersion, a pathological hallmark of MTLE. These disease-modifying effects were dose-dependent, with a broad therapeutic range. No adverse effects were observed. These findings support an ongoing phase 1/2 clinical trial (NCT05135091) for drug-resistant MTLE.


Assuntos
Epilepsia do Lobo Temporal , Hipocampo , Camundongos , Animais , Humanos , Hipocampo/patologia , Epilepsia do Lobo Temporal/patologia , Epilepsia do Lobo Temporal/cirurgia , Convulsões/patologia , Convulsões/cirurgia , Interneurônios/fisiologia , Encéfalo/patologia
2.
Cereb Cortex ; 28(6): 1946-1958, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28449024

RESUMO

The neocortex of primates, including humans, contains more abundant and diverse inhibitory neurons compared with rodents, but the molecular foundations of these observations are unknown. Through integrative gene coexpression analysis, we determined a consensus transcriptional profile of GABAergic neurons in mid-gestation human neocortex. By comparing this profile to genes expressed in GABAergic neurons purified from neonatal mouse neocortex, we identified conserved and distinct aspects of gene expression in these cells between the species. We show here that the calcium-binding protein secretagogin (SCGN) is robustly expressed by neocortical GABAergic neurons derived from caudal ganglionic eminences (CGE) and lateral ganglionic eminences during human but not mouse brain development. Through electrophysiological and morphometric analyses, we examined the effects of SCGN expression on GABAergic neuron function and form. Forced expression of SCGN in CGE-derived mouse GABAergic neurons significantly increased total neurite length and arbor complexity following transplantation into mouse neocortex, revealing a molecular pathway that contributes to morphological differences in these cells between rodents and primates.


Assuntos
Neurônios GABAérgicos/metabolismo , Neocórtex/embriologia , Neurogênese/fisiologia , Secretagoginas/metabolismo , Animais , Humanos , Interneurônios/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/metabolismo , Transcriptoma
3.
Cell Stem Cell ; 19(4): 544-557, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27666009

RESUMO

Neuropathic pain and bladder dysfunction represent significant quality-of-life issues for many spinal cord injury patients. Loss of GABAergic tone in the injured spinal cord may contribute to the emergence of these symptoms. Previous studies have shown that transplantation of rodent inhibitory interneuron precursors from the medial ganglionic eminence (MGE) enhances GABAergic signaling in the brain and spinal cord. Here we look at whether transplanted MGE-like cells derived from human embryonic stem cells (hESC-MGEs) can mitigate the pathological effects of spinal cord injury. We find that 6 months after transplantation into injured mouse spinal cords, hESC-MGEs differentiate into GABAergic neuron subtypes and receive synaptic inputs, suggesting functional integration into host spinal cord. Moreover, the transplanted animals show improved bladder function and mitigation of pain-related symptoms. Our results therefore suggest that this approach may be a valuable strategy for ameliorating the adverse effects of spinal cord injury.


Assuntos
Interneurônios/transplante , Neuralgia/etiologia , Neuralgia/terapia , Traumatismos da Medula Espinal/complicações , Transplante de Células-Tronco , Bexiga Urinária/fisiopatologia , Animais , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Sobrevivência Celular , Feminino , Células-Tronco Embrionárias Humanas/citologia , Humanos , Camundongos , Neuralgia/patologia , Bexiga Urinária/patologia
4.
Cell ; 163(1): 55-67, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406371

RESUMO

Radial glia, the neural stem cells of the neocortex, are located in two niches: the ventricular zone and outer subventricular zone. Although outer subventricular zone radial glia may generate the majority of human cortical neurons, their molecular features remain elusive. By analyzing gene expression across single cells, we find that outer radial glia preferentially express genes related to extracellular matrix formation, migration, and stemness, including TNC, PTPRZ1, FAM107A, HOPX, and LIFR. Using dynamic imaging, immunostaining, and clonal analysis, we relate these molecular features to distinctive behaviors of outer radial glia, demonstrate the necessity of STAT3 signaling for their cell cycle progression, and establish their extensive proliferative potential. These results suggest that outer radial glia directly support the subventricular niche through local production of growth factors, potentiation of growth factor signals by extracellular matrix proteins, and activation of self-renewal pathways, thereby enabling the developmental and evolutionary expansion of the human neocortex.


Assuntos
Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Animais , Ciclo Celular , Humanos , Macaca , Camundongos , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neuroglia/citologia , Neuroglia/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Análise de Célula Única , Nicho de Células-Tronco
5.
Science ; 344(6180): 1240622, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24723614

RESUMO

Many neurologic and psychiatric disorders are marked by imbalances between neural excitation and inhibition. In the cerebral cortex, inhibition is mediated largely by GABAergic (γ-aminobutyric acid-secreting) interneurons, a cell type that originates in the embryonic ventral telencephalon and populates the cortex through long-distance tangential migration. Remarkably, when transplanted from embryos or in vitro culture preparations, immature interneurons disperse and integrate into host brain circuits, both in the cerebral cortex and in other regions of the central nervous system. These features make interneuron transplantation a powerful tool for the study of neurodevelopmental processes such as cell specification, cell death, and cortical plasticity. Moreover, interneuron transplantation provides a novel strategy for modifying neural circuits in rodent models of epilepsy, Parkinson's disease, mood disorders, and chronic pain.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Desenvolvimento Embrionário , Interneurônios/fisiologia , Interneurônios/transplante , Transtornos Mentais/terapia , Doenças do Sistema Nervoso/terapia , Animais , Contagem de Células , Separação Celular , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Humanos , Camundongos
6.
Cell Stem Cell ; 12(5): 573-86, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23642366

RESUMO

Directed differentiation from human pluripotent stem cells (hPSCs) has seen significant progress in recent years. However, most differentiated populations exhibit immature properties of an early embryonic stage, raising concerns about their ability to model and treat disease. Here, we report the directed differentiation of hPSCs into medial ganglionic eminence (MGE)-like progenitors and their maturation into forebrain type interneurons. We find that early-stage progenitors progress via a radial glial-like stem cell enriched in the human fetal brain. Both in vitro and posttransplantation into the rodent cortex, the MGE-like cells develop into GABAergic interneuron subtypes with mature physiological properties along a prolonged intrinsic timeline of up to 7 months, mimicking endogenous human neural development. MGE-derived cortical interneuron deficiencies are implicated in a broad range of neurodevelopmental and degenerative disorders, highlighting the importance of these results for modeling human neural development and disease.


Assuntos
Diferenciação Celular , Interneurônios/citologia , Neurogênese , Prosencéfalo/citologia , Potenciais de Ação , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Divisão Celular/genética , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interneurônios/metabolismo , Eminência Mediana/citologia , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neuroglia/citologia , Neuroglia/metabolismo , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Sinapses/metabolismo , Telencéfalo/citologia , Fator Nuclear 1 de Tireoide , Fatores de Tempo , Fatores de Transcrição/metabolismo
7.
PLoS One ; 8(5): e61956, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658702

RESUMO

The medial ganglionic eminence (MGE) is an embryonic forebrain structure that generates the majority of cortical interneurons. MGE transplantation into specific regions of the postnatal central nervous system modifies circuit function and improves deficits in mouse models of epilepsy, Parkinson's disease, pain, and phencyclidine-induced cognitive deficits. Herein, we describe approaches to generate MGE-like progenitor cells from mouse embryonic stem (ES) cells. Using a modified embryoid body method, we provided gene expression evidence that mouse ES-derived Lhx6(+) cells closely resemble immature interneurons generated from authentic MGE-derived Lhx6(+) cells. We hypothesized that enhancers that are active in the mouse MGE would be useful tools in detecting when ES cells differentiate into MGE cells. Here we demonstrate the utility of enhancer elements [422 (DlxI12b), Lhx6, 692, 1056, and 1538] as tools to mark MGE-like cells in ES cell differentiation experiments. We found that enhancers DlxI12b, 692, and 1538 are active in Lhx6-GFP(+) cells, while enhancer 1056 is active in Olig2(+) cells. These data demonstrate unique techniques to follow and purify MGE-like derivatives from ES cells, including GABAergic cortical interneurons and oligodendrocytes, for use in stem cell-based therapeutic assays and treatments.


Assuntos
Corpos Embrioides/fisiologia , Elementos Facilitadores Genéticos , Células-Tronco Neurais/fisiologia , Prosencéfalo/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Separação Celular , Células Cultivadas , Corpos Embrioides/transplante , Feminino , Citometria de Fluxo , Neurônios GABAérgicos/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Coloração e Rotulagem , Transcriptoma , Transdução Genética , Proteína Vermelha Fluorescente
9.
BMC Dev Biol ; 10: 2, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20064216

RESUMO

BACKGROUND: Female reproductive potential, or the ability to propagate life, is limited in mammals with the majority of oocytes lost before birth. In mice, surviving perinatal oocytes are enclosed in ovarian follicles for subsequent oocyte development and function in the adult. Before birth, fetal germ cells of both sexes develop in clusters, or germline cysts, in the undifferentiated gonad. Upon sex determination of the fetal gonad, germ cell cysts become organized into testicular or ovarian cord-like structures and begin to interact with gonadal somatic cells. Although germline cysts and testicular cords are required for spermatogenesis, the role of cyst and ovarian cord formation in mammalian oocyte development and female fertility has not been determined. RESULTS: Here, we examine whether intact fetal ovarian germ and somatic cell cord structures are required for oocyte development using mouse gonad re-aggregation and transplantation to disrupt gonadal organization. We observed that germ cells from disrupted female gonad prior to embryonic day e13.5 completed prophase I of meiosis but did not survive following transplantation. Furthermore, re-aggregated ovaries from e13.5 to e15.5 developed with a reduced number of oocytes. Oocyte loss occurred before follicle formation and was associated with an absence of ovarian cord structure and ovary disorganization. However, disrupted ovaries from e16.5 or later were resistant to the re-aggregation impairment and supported robust oocyte survival and development in follicles. CONCLUSIONS: Thus, we demonstrate a critical window of oocyte development from e13.5 to e16.5 in the intact fetal mouse ovary, corresponding to the establishment of ovarian cord structure, which promotes oocyte interaction with neighboring ovarian somatic granulosa cells before birth and imparts oocytes with competence to survive and develop in follicles. Because germline cyst and ovarian cord structures are conserved in the human fetal ovary, the identification of genetic components and molecular mechanisms of pre-follicle stage germ and somatic cell structures may be important for understanding human female infertility. In addition, this work provides a foundation for development of a robust fetal ovarian niche and transplantation based system to direct stem cell-derived oocyte differentiation as a potential therapeutic strategy for the treatment of infertility.


Assuntos
Oócitos/citologia , Ovário/citologia , Animais , Feminino , Feto , Células da Granulosa/metabolismo , Meiose , Camundongos , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Ovário/fisiologia
10.
Hum Mol Genet ; 18(22): 4376-89, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19696121

RESUMO

Ten to 15% of couples are infertile, with the most common causes being linked to the production of few or no oocytes or sperm. Yet, our understanding of human germ cell development is poor, at least in part due to the inaccessibility of early stages to genetic and developmental studies. Embryonic stem cells (ESCs) provide an in vitro system to study oocyte development and potentially treat female infertility. However, most studies of ESC differentiation to oocytes have not documented fundamental properties of endogenous development, making it difficult to determine the physiologic relevance of differentiated germ cells. Here, we sought to establish fundamental parameters of oocyte development during ESC differentiation to explore suitability for basic developmental genetic applications using the mouse as a model prior to translating to the human system. We demonstrate a timeline of definitive germ cell differentiation from ESCs in vitro that initially parallels endogenous oocyte development in vivo by single-cell expression profiling and analysis of functional milestones including responsiveness to defined maturation media, shared genetic requirement of Dazl, and entry into meiosis. However, ESC-derived oocyte maturation ultimately fails in vitro. To overcome this obstacle, we transplant ESC-derived oocytes into an ovarian niche to direct their functional maturation and, thereby, present rigorous evidence of oocyte physiologic relevance and a potential therapeutic strategy for infertility.


Assuntos
Células-Tronco Embrionárias/transplante , Infertilidade Feminina/terapia , Oócitos/crescimento & desenvolvimento , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Infertilidade Feminina/fisiopatologia , Meiose , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/citologia , Oócitos/metabolismo , Oócitos/transplante , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
11.
Endocr Rev ; 30(3): 264-83, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19366753

RESUMO

Female reproductive potential is limited in the majority of species due to oocyte depletion. Because functional human oocytes are restricted in number and accessibility, a robust system to differentiate oocytes from stem cells would enable a thorough investigation of the genetic, epigenetic, and environmental factors affecting human oocyte development. Also, the differentiation of functional oocytes from stem cells may permit the success of human somatic cell nuclear transfer for reprogramming studies and for the production of patient-specific embryonic stem cells (ESCs). Thus, ESC-derived oocytes could ultimately help to restore fertility in women. Here, we review endogenous and ESC-derived oocyte development, and we discuss the potential and challenges for differentiating functional oocytes from ESCs.


Assuntos
Células-Tronco Embrionárias/fisiologia , Oócitos/fisiologia , Oogênese/fisiologia , Animais , Técnicas de Cultura de Células , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Oócitos/citologia , Oogênese/genética
12.
Genesis ; 47(2): 74-84, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19133679

RESUMO

In this study, we characterized the promoter activity of a 1.7 kb sequence in the 5' flanking region of the mouse Deleted in Azoospermia-Like (Dazl) gene. We found the 1.7 kb sequence sufficient to drive robust germ cell-specific expression of green fluorescent protein (GFP) in adult mouse testis and lower levels of expression in adult ovary and in fetal and newborn gonads of both sexes. This expression pattern was confirmed in two independently-derived transgenic mouse lines. In adult testis, Dazl-GFP exhibited a developmentally-regulated, stage-specific expression pattern during spermatogenesis. GFP was highly expressed in spermatocyte stages, with strongest expression in pachytene spermatocytes. Weaker expression was observed in round and elongating spermatids, as well as spermatogonial cells. In the fetal gonad, GFP transcript was detected by e12.5 in both sexes; however, GFP fluorescence was only detected during later embryonic stages. In addition, we produced mouse embryonic stem cell (ESC) lines harboring the Dazl-GFP reporter and used this reporter to isolate putative germ cell populations derived from mouse ESCs following embryoid body differentiation and fluorescence activated cell sorting.


Assuntos
Genes Reporter , Proteínas de Fluorescência Verde/genética , Ovário/metabolismo , Proteínas de Ligação a RNA/genética , Testículo/metabolismo , Regiões 5' não Traduzidas , Animais , Animais Recém-Nascidos , Sequência de Bases , Diferenciação Celular , Separação Celular , Primers do DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Transgênicos , Oogênese/genética , Ovário/citologia , Ovário/embriologia , Ovário/crescimento & desenvolvimento , Gravidez , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Espermatogênese/genética , Testículo/citologia , Testículo/embriologia , Testículo/crescimento & desenvolvimento
13.
Stem Cells ; 27(1): 138-49, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18927477

RESUMO

Several reports have documented the derivation of pluripotent cells (multipotent germline stem cells) from spermatogonial stem cells obtained from the adult mouse testis. These spermatogonia-derived stem cells express embryonic stem cell markers and differentiate to the three primary germ layers, as well as the germline. Data indicate that derivation may involve reprogramming of endogenous spermatogonia in culture. Here, we report the derivation of human multipotent germline stem cells (hMGSCs) from a testis biopsy. The cells express distinct markers of pluripotency, form embryoid bodies that contain derivatives of all three germ layers, maintain a normal XY karyotype, are hypomethylated at the H19 locus, and express high levels of telomerase. Teratoma assays indicate the presence of human cells 8 weeks post-transplantation but limited teratoma formation. Thus, these data suggest the potential to derive pluripotent cells from human testis biopsies but indicate a need for novel strategies to optimize hMGSC culture conditions and reprogramming.


Assuntos
Células-Tronco Pluripotentes/citologia , Espermatogônias/citologia , Biomarcadores/metabolismo , Biópsia , Diferenciação Celular , Linhagem Celular , Separação Celular , Forma Celular , Metilação de DNA , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Masculino , Repetições Minissatélites/genética , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas/genética , Análise de Sequência de DNA , Cariotipagem Espectral , Espermatogônias/metabolismo , Sulfitos , Telomerase/metabolismo , Testículo/citologia , Testículo/metabolismo
14.
CSH Protoc ; 2008: pdb.prot5045, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21356914

RESUMO

INTRODUCTIONThe potential for human embryonic stem cells (hESCs) to differentiate into all three embryonic germ layers, and the germline, can be used as a tool to understand the mechanisms of cell lineage-specific differentiation and development in vitro and in vivo. Therefore, it is important to develop technologies for the manipulation of hESCs and their effective utilization in research. One such manipulation is genetic modification. Reliable methods for genetic modification of hESCs with ubiquitous and tissue-specific reporters will assist in the identification and isolation of specific cell types following hESC differentiation. This protocol describes a method for hESC transduction with a ubiquitous eGFP reporter construct using self-inactivating lentivirus, followed by single-cell isolation and expansion of homogenous genetically modified hESCs using fluorescence-activated cell sorting (FACS).

15.
Stem Cells Dev ; 16(1): 109-17, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17348809

RESUMO

Genetic modification of human embryonic stem (hES) cells is essential for studies of gene function and differentiation. The expression of transgenes may direct tissue-specific differentiation and aid in the identification of various differentiated cell types. Stable genomic integration of transgenes is optimal because hES cell differentiation can span several days to weeks and include numerous cell divisions, and establishing homogeneous modified cell lines will facilitate research studies. Herein we provide a method for producing and expanding hES cell lines from single cells that have been isolated by fluorescence-activated cell sorting (FACS) following genetic modification by lentivirus vectors. Using this method, we have established enhanced green fluorescent protein (eGFP)-expressing hES cell lines that are pluripotent, contain a diploid chromosomal content, and stably express eGFP following more than 2 months of routine culture and in vivo differentiation.


Assuntos
Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Transdução Genética , Diferenciação Celular , Células Clonais , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transgenes
16.
Cancer Cell ; 6(6): 611-23, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15607965

RESUMO

ONYX-015 is an adenovirus that lacks the E1B-55K gene product for p53 degradation. Thus, ONYX-015 was conceived as an oncolytic virus that would selectively replicate in p53-defective tumor cells. Here we show that loss of E1B-55K leads to the induction, but not the activation, of p53 in ONYX-015-infected primary cells. We use a novel adenovirus mutant, ONYX-053, to demonstrate that loss of E1B-55K-mediated late viral RNA export, rather than p53 degradation, restricts ONYX-015 replication in primary cells. In contrast, we show that tumor cells that support ONYX-015 replication provide the RNA export function of E1B-55K. These data reveal that tumor cells have altered mechanisms for RNA export and resolve the controversial role of p53 in governing ONYX-015 oncolytic selectivity.


Assuntos
Adenoviridae/genética , Neoplasias/virologia , RNA Viral/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Replicação Viral/genética , Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Apoptose , Western Blotting , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Efeito Citopatogênico Viral/genética , DNA Viral/biossíntese , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Células Epiteliais/virologia , Expressão Gênica/genética , Células HCT116 , Humanos , Hibridização in Situ Fluorescente , Modelos Biológicos , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Reação em Cadeia da Polimerase , Biossíntese de Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-mdm2 , Transporte de RNA , Proteína Supressora de Tumor p14ARF/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Ensaio de Placa Viral , Proteínas Virais/metabolismo , Vacinas Virais , Proteína X Associada a bcl-2
17.
J Biomol Screen ; 9(8): 704-11, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15634797

RESUMO

With the sequence of the human genome at hand, target discovery strategies are needed that can rapidly identify novel gene products involved in human disease pathways. In this article, the authors describe a cell-based, high-throughput assay that can identify gene products capable of modulating the vascular endothelial growth factor (VEGF) and tumor necrosis factor alpha (TNFalpha) signaling pathways in human endothelial cells. The assay uses real-time PCR technology to measure downstream reporter mRNA transcripts induced upon cytokine stimulation in a 96-well plate format and has been adapted for use with recombinant adenoviruses. The authors specifically demonstrate modulation of cytokine-driven reporter transcripts using drug inhibitors and through adenoviral-mediated expression of known signaling intermediates of the respective pathways. In addition, they have used an arrayed library of 350 recombinant adenoviruses to screen for novel modulators of the VEGF and TNFalpha pathways. The high-throughput screening capacity and sensitivity of this system make it a useful tool for new drug target identification.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Endotélio Vascular/efeitos dos fármacos , Genômica/métodos , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adenoviridae/genética , Bioensaio , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Biblioteca Gênica , Técnicas de Transferência de Genes , Genes Reporter , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Reação em Cadeia da Polimerase , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tromboplastina/genética , Tromboplastina/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...