Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Neurochem ; 122(2): 260-71, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22568433

RESUMEN

The male gender is determined by the sex-determining region on the Y chromosome (SRY) transcription factor. The unexpected action of SRY in the control of voluntary movement in male rodents suggests a role in the regulation of dopamine transmission and dopamine-related disorders with gender bias, such as Parkinson's disease. We investigated SRY expression in the human brain and function in vitro. SRY immunoreactivity was detected in the human male, but not female substantia nigra pars compacta, within a sub-population of tyrosine hydroxylase (TH) positive neurons. SRY protein also co-localized with TH positive neurons in the ventral tegmental area, and with GAD-positive neurons in the substantia nigra pars reticulata. Retinoic acid-induced differentiation of human precursor NT2 cells into dopaminergic cells increased expression of TH, NURR1, D2 R and SRY. In the human neuroblastoma cell line, M17, SRY knockdown resulted in a reduction in TH, DDC, DBH and MAO-A expression; enzymes which control dopamine synthesis and metabolism. Conversely, SRY over-expression increased TH, DDC, DBH, D2 R and MAO-A levels, accompanied by increased extracellular dopamine levels. A luciferase assay demonstrated that SRY activated a 4.6 kb 5' upstream regulatory region of the human TH promoter/nigral enhancer. Combined, these results suggest that SRY plays a role as a positive regulator of catecholamine synthesis and metabolism in the human male midbrain. This ancillary genetic mechanism might contribute to gender bias in fight-flight behaviours in men or their increased susceptibility to dopamine disorders, such as Parkinson's disease and schizophrenia.


Asunto(s)
Catecolaminas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Genes sry/genética , Mesencéfalo/metabolismo , Testículo/metabolismo , Adulto , Anciano , Western Blotting , Catecolaminas/biosíntesis , Células Cultivadas , Cromatografía Líquida de Alta Presión , Dopamina/metabolismo , Electroquímica , Femenino , Humanos , Inmunohistoquímica , Masculino , Mesencéfalo/citología , Persona de Mediana Edad , Plásmidos/genética , ARN/biosíntesis , ARN/aislamiento & purificación , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Sustancia Negra/citología , Sustancia Negra/metabolismo , Bancos de Tejidos , Transfección , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética
2.
Endocrinology ; 153(2): 901-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22128028

RESUMEN

Genome analysis of patients with disorders of sex development, and gain- and loss-of-function studies in mice indicate that gonadal development is regulated by opposing signals. In females, the Wnt/ß-catenin canonical pathway blocks testicular differentiation by repressing the expression of the Sertoli cell-specific gene Sox9 by an unknown mechanism. Using cell and embryonic gonad culture models, we show that activation of the Wnt/ß-catenin pathway inhibits the expression of Sox9 and Amh, whereas mRNA and protein levels of Sry and steroidogenic factor 1 (Sf1), two key transcriptional regulators of Sox9, are not altered. Ectopic activation of Wnt/ß-catenin signaling in male gonads led to a loss of Sf1 binding to the Tesco enhancer and absent Sox9 expression that we also observed in wild-type ovaries. Moreover, ectopic Wnt/ß-catenin signaling induced the expression of the female somatic cell markers, Bmp2 and Rspo1, as a likely consequence of Sox9 loss. Wnt/ß-catenin signaling in XY gonads did not, however, affect gene expression of the steroidogenic Leydig cell Sf1 target gene, Cyp11a1, or Sf1 binding to the Cyp11a1 promoter. Our data support a model in ovary development whereby activation of ß-catenin prevents Sf1 binding to the Sox9 enhancer, thereby inhibiting Sox9 expression and Sertoli cell differentiation.


Asunto(s)
Ovario/crecimiento & desarrollo , Factor de Transcripción SOX9/metabolismo , Factor Esteroidogénico 1/metabolismo , Proteínas Wnt/metabolismo , Animales , Hormona Antimülleriana/genética , Hormona Antimülleriana/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Ovario/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/genética , Procesos de Determinación del Sexo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Transducción de Señal/fisiología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
3.
Endocrinology ; 152(7): 2883-93, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21558314

RESUMEN

The transcription factor sex-determining region of the Y chromosome (SRY) plays a key role in human sex determination, because mutations in SRY cause disorders of sex development in XY individuals. During gonadal development, Sry in pre-Sertoli cells activates Sox9 gene transcription, committing the fate of the bipotential gonad to become a testis rather than an ovary. The high-mobility group domain of human SRY contains two independent nuclear localization signals, one bound by calmodulin (CaM) and the other by importin-ß. Although XY females carry SRY mutations in these nuclear localization signals that affect SRY nuclear import in transfected cells, it is not known whether these transport mechanisms are essential for gonadal development and sex determination. Here, we show that mouse Sry protein binds CaM and that a CaM antagonist reduces CaM binding, nuclear accumulation, and transcriptional activity of Sry in transfected cells. CaM antagonist treatment of cultured, sexually indifferent XY mouse fetal gonads led to reduced expression of the Sry target gene Sox9, defects in testicular cord formation, and ectopic expression of the ovarian markers Rspondin1 and forkhead box L2. These results indicate the importance of CaM for SRY nuclear import, transcriptional activity, testis differentiation, and sex determination.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Antígenos de Diferenciación/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Calmodulina/antagonistas & inhibidores , Proteína de la Región Y Determinante del Sexo/metabolismo , Espermatogénesis , Testículo/fisiología , Animales , Células COS , Calmodulina/metabolismo , Proteínas de Unión a Calmodulina/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Chlorocebus aethiops , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Técnicas de Cultivo de Órganos , Proteínas Recombinantes/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Procesos de Determinación del Sexo/efectos de los fármacos , Proteína de la Región Y Determinante del Sexo/genética , Cordón Espermático/efectos de los fármacos , Testículo/ultraestructura , Trombospondinas/metabolismo , Activación Transcripcional/efectos de los fármacos
4.
PLoS One ; 6(3): e17751, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21412441

RESUMEN

BACKGROUND: In human embryogenesis, loss of SRY (sex determining region on Y), SOX9 (SRY-related HMG box 9) or SF1 (steroidogenic factor 1) function causes disorders of sex development (DSD). A defining event of vertebrate sex determination is male-specific upregulation and maintenance of SOX9 expression in gonadal pre-Sertoli cells, which is preceded by transient SRY expression in mammals. In mice, Sox9 regulation is under the transcriptional control of SRY, SF1 and SOX9 via a conserved testis-specific enhancer of Sox9 (TES). Regulation of SOX9 in human sex determination is however poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: We show that a human embryonal carcinoma cell line (NT2/D1) can model events in presumptive Sertoli cells that initiate human sex determination. SRY associates with transcriptionally active chromatin in NT2/D1 cells and over-expression increases endogenous SOX9 expression. SRY and SF1 co-operate to activate the human SOX9 homologous TES (hTES), a process dependent on phosphorylated SF1. SOX9 also activates hTES, augmented by SF1, suggesting a mechanism for maintenance of SOX9 expression by auto-regulation. Analysis of mutant SRY, SF1 and SOX9 proteins encoded by thirteen separate 46,XY DSD gonadal dysgenesis individuals reveals a reduced ability to activate hTES. CONCLUSIONS/SIGNIFICANCE: We demonstrate how three human sex-determining factors are likely to function during gonadal development around SOX9 as a hub gene, with different genetic causes of 46,XY DSD due a common failure to upregulate SOX9 transcription.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY/genética , Mutación/genética , Factor de Transcripción SOX9/genética , Proteína de la Región Y Determinante del Sexo/genética , Factor Esteroidogénico 1/genética , Línea Celular , Elementos de Facilitación Genéticos/genética , Humanos , Masculino , Proteínas Mutantes/metabolismo , Especificidad de Órganos/genética , Factor de Transcripción SOX9/metabolismo , Testículo/metabolismo , Transactivadores/metabolismo , Regulación hacia Arriba/genética
5.
Int J Biochem Cell Biol ; 40(12): 2889-900, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18598779

RESUMEN

In most mammals, sex is determined by the presence or absence of the SRY gene. SRY encodes a DNA-binding HMG-box transcription factor which, during embryogenesis, is the initial trigger of testis differentiation from the bipotential gonad, yet its precise mode of function remains unclear. In ovarian development, R-spondin1 and Wnt4 act through the Wnt/beta-catenin-signaling pathway to regulate TCF-dependent expression of unknown target genes and repress testis development. Conversely, SRY may be necessary to prevent the development of ovaries by inhibiting the action of ovarian-determining genes. We hypothesize that SRY prevents Wnt/beta-catenin signaling, thereby inhibiting ovarian development. In HEK293T cells, SRY repressed beta-catenin-mediated TCF-dependent gene activation in the presence of a specific GSK3beta inhibitor or an activated beta-catenin mutant, suggesting that SRY inhibits Wnt signaling at the level of beta-catenin. Three SRY mutant proteins with nuclear localization defects, encoded by XY male-to-female patients, failed to inhibit beta-catenin; surprisingly four SRY sex reversed mutants with defective DNA-binding activity showed near wild-type SRY inhibitory activity. Moreover the potent transactivator SRY-VP16 fusion protein also showed wild-type SRY inhibitory activity. Thus SRY inhibition of beta-catenin involves neither DNA-binding nor transactivation functions of SRY. beta-Catenin and SRY interact in vitro and SRY expression triggered beta-catenin localization into specific nuclear bodies in NT2/D1 and Hela cells. We conclude that SRY inhibits beta-catenin-mediated Wnt signaling by a novel nuclear function of SRY that could be important in sex determination.


Asunto(s)
Genes sry , Transcripción Genética , beta Catenina/metabolismo , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Trastornos del Desarrollo Sexual , Femenino , Células HeLa , Humanos , Inmunohistoquímica , Riñón/citología , Masculino , Modelos Biológicos , Unión Proteica , Transducción de Señal , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética
6.
Trends Endocrinol Metab ; 19(6): 213-22, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18585925

RESUMEN

In the mammalian embryo, SRY and SOX9 are key Sertoli cell proteins that drive the development of the bipotential gonad into a testes rather than an ovary, leading ultimately to the male phenotype. Clinical SRY and SOX9 mutations causing disorders of sex development (DSD) highlight defective protein-protein interactions between SRY or SOX9, and carrier proteins required for nuclear import (importin-b and calmodulin) and nuclear export (CRM-1). The fine balance between import and export determines the levels of transcriptionally active SRY and SOX9 in the nucleus. Recently, post-translational modifications of SRY and SOX9 have been identified which affect nuclear transport. It is therefore timely that the consequences of sex-reversal mutation upon nuclear transport be reviewed. SRY and SOX9 mutations in DSD have uncovered regulatory sites for sumoylation, ubiquitination, acetylation and phosphorylation, many of which are essential for their transport and sex determining functions.


Asunto(s)
Factor de Transcripción SOX9/metabolismo , Proteína de la Región Y Determinante del Sexo/metabolismo , Animales , Femenino , Humanos , Masculino , Modelos Biológicos , Unión Proteica , Procesamiento Proteico-Postraduccional , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/fisiología , Procesos de Determinación del Sexo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/fisiología
7.
Dev Biol ; 314(1): 71-83, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18155190

RESUMEN

In mammals, sex is determined in the bipotential embryonic gonad by a balanced network of gene actions which when altered causes disorders of sexual development (DSD, formerly known as intersex). In the XY gonad, presumptive Sertoli cells begin to differentiate when SRY up-regulates SOX9, which in turn activates FGF9 and PGDS to maintain its own expression. This study identifies a new and essential component of FGF signaling in sex determination. Fgfr2 mutant XY mice on a mixed 129/C57BL6 genetic background had either normal testes, or developed ovotestes, with predominantly testicular tissue. However, backcrossing to C57BL6 mice resulted in a wide range of gonadal phenotypes, from hypoplastic testes to ovotestes with predominantly ovarian tissue, similar to Fgf9 knockout mice. Since typical male-specific FGF9-binding to the coelomic epithelium was abolished in Fgfr2 mutant XY gonads, these results suggest that FGFR2 acts as the receptor for FGF9. Pgds and SOX9 remained expressed within the testicular portions of Fgfr2 mutant ovotestes, suggesting that the Prostaglandin pathway acts independently of FGFR2 to maintain SOX9 expression. We could further demonstrate that double-heterozygous Fgfr2/Sox9 knockout mice developed ovotestes, demonstrating that both Fgfr2 and Sox9 can act as modifier intersex genes in the heterozygous state. In summary, we provide evidence that FGFR2 is important for male sex determination in mice, thereby rendering human FGFR2 a candidate gene for unsolved DSD cases such as 10q26 deletions.


Asunto(s)
Trastornos del Desarrollo Sexual , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Procesos de Determinación del Sexo , Animales , Femenino , Factor 9 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/fisiología , Gónadas/citología , Gónadas/embriología , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/fisiología , Masculino , Ratones , Ratones Noqueados , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factor de Transcripción SOX9 , Células de Sertoli/citología , Células de Sertoli/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
8.
Curr Biol ; 16(4): 415-20, 2006 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-16488877

RESUMEN

The central dogma of mammalian brain sexual differentiation has contended that sex steroids of gonadal origin organize the neural circuits of the developing brain. Recent evidence has begun to challenge this idea and has suggested that, independent of the masculinizing effects of gonadal secretions, XY and XX brain cells have different patterns of gene expression that influence their differentiation and function. We have previously shown that specific differences in gene expression exist between male and female developing brains and that these differences precede the influences of gonadal hormones. Here we demonstrate that the Y chromosome-linked, male-determining gene Sry is specifically expressed in the substantia nigra of the adult male rodent in tyrosine hydroxylase-expressing neurons. Furthermore, using antisense oligodeoxynucleotides, we show that Sry downregulation in the substantia nigra causes a statistically significant decrease in tyrosine hydroxylase expression with no overall effect on neuronal numbers and that this decrease leads to motor deficits in male rats. Our studies suggest that Sry directly affects the biochemical properties of the dopaminergic neurons of the nigrostriatal system and the specific motor behaviors they control. These results demonstrate a direct male-specific effect on the brain by a gene encoded only in the male genome, without any mediation by gonadal hormones.


Asunto(s)
Genes sry/fisiología , Caracteres Sexuales , Sustancia Negra/metabolismo , Animales , Dopamina/metabolismo , Regulación hacia Abajo , Femenino , Expresión Génica , Masculino , Ratones , Actividad Motora , Neostriado/metabolismo , Neuronas/metabolismo , Ratas , Tirosina 3-Monooxigenasa/metabolismo
9.
Mol Endocrinol ; 19(7): 1884-92, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15746192

RESUMEN

The sex-determining region of the Y chromosome (SRY) plays a key role in human sex determination, as mutations in SRY can cause XY sex reversal. Although some SRY missense mutations affect DNA binding and bending activities, it is unclear how others contribute to disease. The high mobility group domain of SRY has two nuclear localization signals (NLS). Sex-reversing mutations in the NLSs affect nuclear import in some patients, associated with defective importin-beta binding to the C-terminal NLS (c-NLS), whereas in others, importin-beta recognition is normal, suggesting the existence of an importin-beta-independent nuclear import pathway. The SRY N-terminal NLS (n-NLS) binds calmodulin (CaM) in vitro, and here we show that this protein interaction is reduced in vivo by calmidazolium, a CaM antagonist. In calmidazolium-treated cells, the dramatic reduction in nuclear entry of SRY and an SRY-c-NLS mutant was not observed for two SRY-n-NLS mutants. Fluorescence spectroscopy studies reveal an unusual conformation of SRY.CaM complexes formed by the two n-NLS mutants. Thus, CaM may be involved directly in SRY nuclear import during gonadal development, and disruption of SRY.CaM recognition could underlie XY sex reversal. Given that the CaM-binding region of SRY is well-conserved among high mobility group box proteins, CaM-dependent nuclear import may underlie additional disease states.


Asunto(s)
Calmodulina/metabolismo , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Trastornos del Desarrollo Sexual , Genes sry/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células COS , Calmodulina/análisis , Calmodulina/antagonistas & inhibidores , Núcleo Celular/química , Chlorocebus aethiops , Cromosomas Humanos X/genética , Cromosomas Humanos Y/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Femenino , Dominios HMG-Box/genética , Dominios HMG-Box/fisiología , Humanos , Imidazoles/farmacología , Masculino , Datos de Secuencia Molecular , Mutación , Señales de Localización Nuclear , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteína de la Región Y Determinante del Sexo , Factores de Transcripción/química , Factores de Transcripción/genética
10.
J Biol Chem ; 278(36): 33839-47, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12810722

RESUMEN

During mammalian sex determination, SOX9 is translocated into the nuclei of Sertoli cells within the developing XY gonad. The N-terminal nuclear localization signal (NLS) is contained within a SOX consensus calmodulin (CaM) binding region, thereby implicating CaM in nuclear import of SOX9. By fluorescence spectroscopy and glutaraldehyde cross-linking, we show that the SOX9 HMG domain and CaM interact in vitro. The formation of a SOX9.CaM binary complex is calcium-dependent and is accompanied by a conformational change in SOX9. A CaM antagonist, calmidazolium chloride (CDZ), was observed to block CaM recognition of SOX9 in vitro and inhibit both nuclear import and consequent transcriptional activity of SOX9 in treated cells. The significance of the SOX9-CaM interaction was highlighted by analysis of a missense SOX9 mutation, A158T, identified from a XY female with campomelic dysplasia/autosomal sex reversal (CD/SRA). This mutant binds importin beta normally despite defective nuclear import. Fluorescence and quenching studies indicate that in the unbound state, the A158T mutant shows a similar conformation to that of the WT SOX9, but in the presence of CaM, the mutant undergoes unusual conformational changes. Furthermore, SOX9-mediated transcriptional activation by cells expressing the A158T mutant is more sensitive to CDZ than cells expressing WT SOX9. These results suggest first that CaM is involved in the nuclear transport of SOX9 in a process likely to involve direct interaction and second, that CD/SRA can arise, at least in part, from a defect in CaM recognition, ultimately leading to reduced ability of SOX9 to activate transcription of cartilage and testes-forming genes.


Asunto(s)
Transporte Activo de Núcleo Celular , Calmodulina/metabolismo , Trastornos del Desarrollo Sexual , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Acrilamida/farmacología , Secuencia de Aminoácidos , Animales , Células COS , Calmodulina/química , Núcleo Celular/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Relación Dosis-Respuesta a Droga , Ácido Egtácico/farmacología , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Femenino , Glutaral/farmacología , Imidazoles/farmacología , Inmunohistoquímica , Masculino , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Mutación Missense , Señales de Localización Nuclear , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Factor de Transcripción SOX9 , Homología de Secuencia de Aminoácido , Células de Sertoli/metabolismo , Espectrometría de Fluorescencia , Factores de Tiempo , Transcripción Genética , Transfección , beta Carioferinas/química
11.
J Biol Chem ; 277(22): 19408-17, 2002 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11909852

RESUMEN

The transcription factor STAT1 plays a pivotal role in signal transduction of type I and II interferons (IFNs). STAT1 activation leads to changes in expression of key regulatory genes encoding caspases and cell cycle inhibitors. Deficient STAT1 expression in human cancer cells and virally mediated inhibition of STAT1 function have been associated with cellular resistance to IFNs and mycobacterial infection in humans. Thus, given the relative importance of STAT1, we isolated and characterized a human STAT1 intronic enhancer region displaying IFN-regulated activity. Functional analyses by transient expression identified a repressor region and type I and II IFN-inducible elements within the STAT1 enhancer sequence. A candidate IRF-E/GAS/IRF-E (IGI) sequence containing GAAANN nucleotide repeats was shown by gel shift assay to bind to IFN regulatory factor-1 (IRF-1), but not to IFN-stimulated gene factor-3 (ISGF-3) or STAT1-3. An additional larger IGI-binding complex containing IRF-1 was identified. Mutation of the GAAANN repeats within the IGI DNA element eliminated IRF-1 binding and the IFN-regulated activity of the STAT1 intronic enhancer region. Transfection of the IFN-resistant MM96 cell line to express increased levels of IRF-1 protein also elevated STAT1, STAT2, and p48/IRF-9 expression and enhanced cellular responsiveness to IFN-beta. Reciprocating regulation between IRF-1 and STAT1 genes and encoded proteins indicates that an intracellular amplifier circuit exists controlling cellular responsiveness to the IFNs.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Interferón Tipo I/farmacología , Interferón gamma/farmacología , Fosfoproteínas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Secuencias de Aminoácidos , Secuencia de Bases , Sitios de Unión , Northern Blotting , Western Blotting , Mapeo Cromosómico , Cromosomas Artificiales de Levadura , Cromosomas Humanos Par 2 , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Elementos de Facilitación Genéticos , Exones , Genes Reporteros , Vectores Genéticos , Humanos , Factor 1 Regulador del Interferón , Intrones , Luciferasas/metabolismo , Modelos Genéticos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Reacción en Cadena de la Polimerasa , Unión Proteica , Factor de Transcripción STAT1 , Factores de Tiempo , Transcripción Genética , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA