Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Biol Chem ; 291(19): 10201-9, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-26966181

RESUMEN

Tumor suppressor pRb represses Skp2, a substrate-recruiting subunit of the SCF(Skp2) ubiquitin ligase. Rb1(+/-) mice incur "two-hit" pituitary tumorigenesis; Skp2(-/-);Rb1(+/-) mice do not. Rb1(-/-) embryos die on embryonic day (E) 14.5-15.5. Here, we report that Skp2(-/-);Rb1(-/-) embryos died on E11.5, establishing an organismal level synthetic lethal relationship between Rb1 and Skp2 On E10.5, Rb1(-/-) placentas showed similarly active proliferation and similarly inactive apoptosis as WT placenta, whereas Rb1(-/-) embryos showed ectopic proliferation without increased apoptosis in the brain. Combining Skp2(-/-) did not reduce proliferation or increase apoptosis in the placentas but induced extensive apoptosis in the brain. We conditionally deleted Rb1 in neuronal lineage with Nes-Cre and reproduced the brain apoptosis in E13.5 Nes-Cre;Rb1(lox/lox);Skp2(-/-) embryos, demonstrating their synthetic lethal relationship at a cell autonomous level. Nes-Cre-mediated Rb1 deletion increased expression of proliferative E2F target genes in the brains of Skp2(+/+) embryos; the increases rose higher with activation of expression of apoptotic E2F target genes in Skp2(-/-) embryos. The brain apoptosis was independent of p53 but coincident with proliferation. The highly activated expression of proliferative and apoptotic E2F target genes subsided with gradually reduced roles of Skp2 in preventing p27 protein accumulation in the brain in late gestation, allowing the embryos to reach full term with normally sized brains. These findings establish that Rb1 and Skp2 deletions are synthetic lethal and suggest how this lethal relationship might be circumvented, which could help design better therapies for pRb-deficient cancer.


Asunto(s)
Apoptosis , Pérdida del Embrión , Embrión de Mamíferos , Desarrollo Embrionario/genética , Proteína de Retinoblastoma , Proteínas Quinasas Asociadas a Fase-S , Animales , Apoptosis/genética , Encéfalo/embriología , Encéfalo/patología , Factores de Transcripción E2F/genética , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
EMBO J ; 32(6): 844-57, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23403926

RESUMEN

pRb is frequently inactivated in tumours by mutations or phosphorylation. Here, we investigated whether pRb plays a role in obesity. The Arcuate nucleus (ARC) in hypothalamus contains antagonizing POMC and AGRP/NPY neurons for negative and positive energy balance, respectively. Various aspects of ARC neurons are affected in high-fat diet (HFD)-induced obesity mouse model. Using this model, we show that HFD, as well as pharmacological activation of AMPK, induces pRb phosphorylation and E2F target gene de-repression in ARC neurons. Some affected neurons express POMC; and deleting Rb1 in POMC neurons induces E2F target gene de-repression, cell-cycle re-entry, apoptosis, and a hyperphagia-obesity-diabetes syndrome. These defects can be corrected by combined deletion of E2f1. In contrast, deleting Rb1 in the antagonizing AGRP/NPY neurons shows no effects. Thus, pRb-E2F1 is an obesity suppression mechanism in ARC POMC neurons and HFD-AMPK inhibits this mechanism by phosphorylating pRb in this location.


Asunto(s)
Dieta Alta en Grasa , Grasas de la Dieta/farmacología , Hipotálamo , Obesidad/genética , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/fisiología , Adenilato Quinasa/metabolismo , Adenilato Quinasa/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo/genética , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/fisiología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Peso Corporal Ideal/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Obesidad/metabolismo , Obesidad/patología , Fosforilación/efectos de los fármacos , Proopiomelanocortina/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo
3.
J Biol Chem ; 290(9): 5797-809, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25583987

RESUMEN

p27Kip1 (p27) is an inhibitor of cyclin-dependent kinases. Inhibiting p27 protein degradation is an actively developing cancer therapy strategy. One focus has been to identify small molecule inhibitors to block recruitment of Thr-187-phosphorylated p27 (p27T187p) to SCF(Skp2/Cks1) ubiquitin ligase. Since phosphorylation of Thr-187 is required for this recruitment, p27T187A knockin (KI) mice were generated to determine the effects of systemically blocking interaction between p27 and Skp2/Cks1 on tumor susceptibility and other proliferation related mouse physiology. Rb1(+/-) mice develop pituitary tumors with full penetrance and the tumors are invariably Rb1(-/-), modeling tumorigenesis by two-hit loss of RB1 in humans. Immunization induced humoral immunity depends on rapid B cell proliferation and clonal selection in germinal centers (GCs) and declines with age in mice and humans. Here, we show that p27T187A KI prevented pituitary tumorigenesis in Rb1(+/-) mice and corrected decline in humoral immunity in older mice following immunization with sheep red blood cells (SRBC). These findings reveal physiological contexts that depend on p27 ubiquitination by SCF(Skp2-Cks1) ubiquitin ligase and therefore help forecast clinical potentials of Skp2/Cks1-p27T187p interaction inhibitors. We further show that GC B cells and T cells use different mechanisms to regulate their p27 protein levels, and propose a T helper cell exhaustion model resembling that of stem cell exhaustion to understand decline in T cell-dependent humoral immunity in older age.


Asunto(s)
Sustitución de Aminoácidos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inmunidad Humoral/genética , Hipófisis/metabolismo , Neoplasias Hipofisarias/genética , Proteína de Retinoblastoma/genética , Factores de Edad , Alanina/genética , Alanina/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Eritrocitos/inmunología , Citometría de Flujo , Técnicas de Sustitución del Gen , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Inmunidad Humoral/inmunología , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Hipófisis/patología , Neoplasias Hipofisarias/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ovinos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Treonina/genética , Treonina/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 34(9): 1917-23, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25035345

RESUMEN

OBJECTIVE: Interindividual variation in pathways affecting cellular cholesterol metabolism can influence levels of plasma cholesterol, a well-established risk factor for cardiovascular disease. Inherent variation among immortalized lymphoblastoid cell lines from different donors can be leveraged to discover novel genes that modulate cellular cholesterol metabolism. The objective of this study was to identify novel genes that regulate cholesterol metabolism by testing for evidence of correlated gene expression with cellular levels of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) mRNA, a marker for cellular cholesterol homeostasis, in a large panel of lymphoblastoid cell lines. APPROACH AND RESULTS: Expression array profiling was performed on 480 lymphoblastoid cell lines established from participants of the Cholesterol and Pharmacogenetics (CAP) statin clinical trial, and transcripts were tested for evidence of correlated expression with HMGCR as a marker of intracellular cholesterol homeostasis. Of these, transmembrane protein 55b (TMEM55B) showed the strongest correlation (r=0.29; P=4.0E-08) of all genes not previously implicated in cholesterol metabolism and was found to be sterol regulated. TMEM55B knockdown in human hepatoma cell lines promoted the decay rate of the low-density lipoprotein receptor, reduced cell surface low-density lipoprotein receptor protein, impaired low-density lipoprotein uptake, and reduced intracellular cholesterol. CONCLUSIONS: Here, we report identification of TMEM55B as a novel regulator of cellular cholesterol metabolism through the combination of gene expression profiling and functional studies. The findings highlight the value of an integrated genomic approach for identifying genes that influence cholesterol homeostasis.


Asunto(s)
Colesterol/metabolismo , Linfocitos/metabolismo , Receptores de LDL/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Perfilación de la Expresión Génica , Células Hep G2 , Hepatocitos/metabolismo , Homeostasis , Humanos , Hidroximetilglutaril-CoA Reductasas/biosíntesis , Hidroximetilglutaril-CoA Reductasas/genética , Líquido Intracelular/metabolismo , Metabolismo de los Lípidos/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
5.
PLoS Genet ; 8(11): e1003058, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166513

RESUMEN

Although statin drugs are generally efficacious for lowering plasma LDL-cholesterol levels, there is considerable variability in response. To identify candidate genes that may contribute to this variation, we used an unbiased genome-wide filter approach that was applied to 10,149 genes expressed in immortalized lymphoblastoid cell lines (LCLs) derived from 480 participants of the Cholesterol and Pharmacogenomics (CAP) clinical trial of simvastatin. The criteria for identification of candidates included genes whose statin-induced changes in expression were correlated with change in expression of HMGCR, a key regulator of cellular cholesterol metabolism and the target of statin inhibition. This analysis yielded 45 genes, from which RHOA was selected for follow-up because it has been found to participate in mediating the pleiotropic but not the lipid-lowering effects of statin treatment. RHOA knock-down in hepatoma cell lines reduced HMGCR, LDLR, and SREBF2 mRNA expression and increased intracellular cholesterol ester content as well as apolipoprotein B (APOB) concentrations in the conditioned media. Furthermore, inter-individual variation in statin-induced RHOA mRNA expression measured in vitro in CAP LCLs was correlated with the changes in plasma total cholesterol, LDL-cholesterol, and APOB induced by simvastatin treatment (40 mg/d for 6 wk) of the individuals from whom these cell lines were derived. Moreover, the minor allele of rs11716445, a SNP located in a novel cryptic RHOA exon, dramatically increased inclusion of the exon in RHOA transcripts during splicing and was associated with a smaller LDL-cholesterol reduction in response to statin treatment in 1,886 participants from the CAP and Pravastatin Inflamation and CRP Evaluation (PRINCE; pravastatin 40 mg/d) statin clinical trials. Thus, an unbiased filter approach based on transcriptome-wide profiling identified RHOA as a gene contributing to variation in LDL-cholesterol response to statin, illustrating the power of this approach for identifying candidate genes involved in drug response phenotypes.


Asunto(s)
Biomarcadores Farmacológicos/metabolismo , Colesterol , Simvastatina/administración & dosificación , Proteína de Unión al GTP rhoA , Alelos , Línea Celular Transformada , Colesterol/genética , Colesterol/metabolismo , Ensayos Clínicos como Asunto , Expresión Génica/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/genética , Polimorfismo de Nucleótido Simple , Pravastatina/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
6.
Mol Cancer Ther ; 6(2): 684-91, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17308064

RESUMEN

Skp2 fulfills the definition of an oncoprotein with its frequent overexpression in cancer cells and oncogenic activity in various laboratory assays and therefore is a potential cancer therapy target. The best-known function of Skp2 is that of an F-box protein of the SCF(Skp2)-Roc1 E3 ubiquitin ligase targeting the cyclin-dependent kinase inhibitor p27(Kip1). Knockdown of Skp2 generally leads to accumulation of p27 but its effects on cancer cells are less certain. Another function of Skp2 is its stable interaction with cyclin A, which directly protects cyclin A from inhibition by p27 in in vitro kinase assays. Here, we report that an 18-residue blocking peptide of Skp2-cyclin A interaction can indirectly inhibit cyclin A/Cdk2 kinase activity dependent on the presence of p27 in in vitro kinase assays. Transmembrane delivery of this blocking peptide can induce cell death in a panel of four cancer cell lines in which Skp2 knockdown only have mild inhibitory effects. This Skp2-cyclin A interaction blocking peptide can synergize with a previously identified E2F1-derived LDL peptide, which blocks its access to cyclin A, in killing cancer cells. IC(50) of the Skp2-cyclin A blocking peptide correlated with abundance of Skp2, its intended target, in cancer cells. These results suggest that Skp2-cyclin A interaction plays an important role in cancer cell survival and is an attractive target for cancer drug discovery.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ciclina A/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Western Blotting , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Ciclina A/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Receptores de LDL/metabolismo , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Fase-S/genética , Células Tumorales Cultivadas/efectos de los fármacos
7.
Nat Commun ; 5: 3463, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24632684

RESUMEN

One mechanism of tumour suppression by pRb is repressing E2F1. Hence, E2f1 deletion diminishes tumorigenesis following Rb1 loss. However, E2F1 promotes both proliferation and apoptosis. It therefore remains unclear how de-repressed E2F1 promotes tumorigenesis. Another mechanism of pRb function is repressing Skp2 to elevate p27 to arrest proliferation. However, Skp2 deletion induced apoptosis, not proliferation arrest, in Rb1-deficient pituitary tumorigenesis. Here we show that Rb1 deletion induces higher expression of E2F1 target genes in the absence of Skp2. E2F1 binds less cyclin A but more target promoters when Rb1 is deleted with Skp2 knockout or p27T187A knockin, suggesting that stabilized p27 prevents cyclin A from binding and inhibiting E2F1. In Rb1-deficient pituitary tumorigenesis, Skp2 deletion or p27T187A mutation converts E2F1's role from proliferative to apoptotic. These findings delineate a pRb-Skp2-p27-cyclin A-E2F1 pathway that determines whether E2F1 is proliferative or apoptotic in Rb1-deficient tumorigenesis.


Asunto(s)
Apoptosis , Regulación hacia Abajo , Factor de Transcripción E2F1/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Animales , Proliferación Celular , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Noqueados , Neoplasias/fisiopatología , Hipófisis/metabolismo , Proteína de Retinoblastoma/genética , Proteínas Quinasas Asociadas a Fase-S/genética
8.
Cancer Cell ; 24(5): 645-59, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24229711

RESUMEN

pRb and p53 are two major tumor suppressors. Here, we found that p53 activates expression of Pirh2 and KPC1, two of the three ubiquitin ligases for p27. Loss of p53 in the absence of Skp2, the third ubiquitin ligase for p27, shrinks the cellular pool of p27 ubiquitin ligases to accumulate p27 protein. In the absence of pRb and p53, p27 was unable to inhibit DNA synthesis in spite of its abundance, but could inhibit division of cells that maintain DNA replication with rereplication. This mechanism blocked pRb/p53 doubly deficient pituitary and prostate tumorigenesis lastingly coexistent with bromodeoxyuridine-labeling neoplastic lesions, revealing an unconventional cancer cell vulnerability when pRb and p53 are inactivated.


Asunto(s)
Carcinogénesis/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/genética , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Carcinogénesis/metabolismo , Línea Celular Tumoral , Senescencia Celular , Replicación del ADN , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
Cell Cycle ; 9(11): 2118-23, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20505340

RESUMEN

A major goal of tumor suppressor research is to neutralize the tumorigenic effects of their loss. Since loss of pRb does not induce tumorigenesis in many types of cells, natural mechanisms may neutralize the tumorigenic effects of pRb loss in these cells. For susceptible cells, neutralizing the tumorigenic effects of pRb loss could logically be achieved by correcting the deregulated activities of pRb targets to render pRb-deficient cells less abnormal. This line of research has unexpectedly revealed that knocking out the pRb target Skp2 did not render Rb1 deficient cells less abnormal but, rather, induced apoptosis in them, thereby completely blocking tumorigenesis in Rb1+/- mice and after targeted deletion of Rb1 in pituitary intermediate lobe (IL). Skp2 is a substrate-recruiting component of the SCFSkp2 E3 biquitin ligase; one of its substrates is Thr187-phosphorylated p27Kip1. A p27T187A knockin (KI) mutation phenocopied Skp2 knockout (KO) in inducing apoptosis following Rb1 loss. Thus, Skp2 KO or p27T187A KI are synthetic lethal with pRb inactivation. Since homozygous p27T187A KI mutations show no adverse effects in mice, inhibiting p27T187 phosphorylation or p27T187p ubiquitination could be a highly therapeutic and minimally toxic intervention strategy for pRb deficiency-induced tumorigenesis.


Asunto(s)
Mutación , Neoplasias/genética , Proteína de Retinoblastoma/genética , Animales , Apoptosis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción E2F/metabolismo , Genes Letales , Ratones , Neoplasias/patología , Mutación Puntual , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
10.
Nat Genet ; 42(1): 83-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19966802

RESUMEN

Heterozygosity of the retinoblastoma gene Rb1 elicits tumorigenesis in susceptible tissues following spontaneous loss of the remaining functional allele. Inactivation of previously studied retinoblastoma protein (pRb) targets partially inhibited tumorigenesis in Rb1(+/-) mice. Here we report that inactivation of pRb target Skp2 (refs. 7,8) completely prevents spontaneous tumorigenesis in Rb1(+/-) mice. Targeted Rb1 deletion in melanotrophs ablates the entire pituitary intermediate lobe when Skp2 is inactivated. Skp2 inactivation does not inhibit aberrant proliferation of Rb1-deleted melanotrophs but induces their apoptotic death. Eliminating p27 phosphorylation on T187 in p27T187A knock-in mice reproduces the effects of Skp2 knockout, identifying p27 ubiquitination by SCF(Skp2) ubiquitin ligase as the underlying mechanism for Skp2's essential tumorigenic role in this setting. RB1-deficient human retinoblastoma cells also undergo apoptosis after Skp2 knockdown; and ectopic expression of p27, especially the p27T187A mutant, induces apoptosis. These results reveal that Skp2 becomes an essential survival gene when susceptible cells incur Rb1 deficiency.


Asunto(s)
Proliferación Celular , Neoplasias Experimentales/genética , Proteína de Retinoblastoma/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Animales , Apoptosis , Western Blotting , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Mutación , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Hipófisis/metabolismo , Hipófisis/patología , Interferencia de ARN , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Análisis de Supervivencia , Factores de Tiempo
11.
Mol Biol Evol ; 23(3): 633-43, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16319090

RESUMEN

Male sexual behavior in the fruit fly Drosophila melanogaster is regulated by fruitless (fru), a sex-determination gene specifying the synthesis of BTB-Zn finger proteins that likely function as male-specific transcriptional regulators. Expression of fru in the nervous system specifies male sexual behavior and the muscle of Lawrence (MOL), an abdominal muscle that develops in males but not in females. We have isolated the fru ortholog from the malaria mosquito Anopheles gambiae and show the gene's conserved genomic structure. We demonstrate that male-specific mosquito fru protein isoforms arise by conserved mechanisms of sex-specifically activated and alternative exon splicing. A male-determining function of mosquito fru is revealed by ectopic expression of the male mosquito isoform FRUMC in fruit flies; this results in MOL development in both fru-mutant males and fru+ females who otherwise develop no MOL. In parallel, we provide evidence of a unique feature of muscle differentiation within the fifth abdominal segment of male mosquitoes that strongly resembles the fruit fly MOL. Given these conserved features within the context of 250 Myr of evolutionary divergence between Drosophila and Anopheles, we hypothesize that fru is the prototypic gene of male sexual behavior among dipteran insects.


Asunto(s)
Anopheles/genética , Evolución Biológica , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Abdomen/anatomía & histología , Secuencia de Aminoácidos , Animales , Anopheles/fisiología , Drosophila melanogaster/fisiología , Femenino , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/anatomía & histología , Músculo Esquelético/metabolismo , Isoformas de Proteínas/genética , Empalme del ARN , Elementos de Respuesta , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA