Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 26(16): 3056-3068, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28535287

RESUMEN

Myotonic dystrophy Type 1 (DM1) is a rare genetic disease caused by the expansion of CTG trinucleotide repeats ((CTG)exp) in the 3' untranslated region of the DMPK gene. The repeat transcripts sequester the RNA binding protein Muscleblind-like protein 1 (MBNL1) and hamper its normal function in pre-mRNA splicing. Overexpressing exogenous MBNL1 in the DM1 mouse model has been shown to rescue the splicing defects and reverse myotonia. Although a viable therapeutic strategy, pharmacological modulators of MBNL1 expression have not been identified. Here, we engineered a ZsGreen tag into the endogenous MBNL1 locus in HeLa cells and established a flow cytometry-based screening system to identify compounds that increase MBNL1 level. The initial screen of small molecule compound libraries identified more than thirty hits that increased MBNL1 expression greater than double the baseline levels. Further characterization of two hits revealed that the small molecule HDAC inhibitors, ISOX and vorinostat, increased MBNL1 expression in DM1 patient-derived fibroblasts and partially rescued the splicing defect caused by (CUG)exp repeats in these cells. These findings demonstrate the feasibility of this flow-based cytometry screen to identify both small molecule compounds and druggable targets for MBNL1 upregulation.


Asunto(s)
Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/metabolismo , Proteína Quinasa de Distrofia Miotónica/genética , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Regiones no Traducidas 3' , Empalme Alternativo , Exones , Citometría de Flujo/métodos , Células HeLa , Humanos , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido , Repeticiones de Trinucleótidos
2.
Hum Mol Genet ; 23(7): 1869-78, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24234649

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by a lack of the dystrophin protein and has no effective treatment at present. Zebrafish provide a powerful in vivo tool for high-throughput therapeutic drug screening for the improvement of muscle phenotypes caused by dystrophin deficiency. Using the dystrophin-deficient zebrafish, sapje, we have screened a total of 2640 compounds with known modes of action from three drug libraries to identify modulators of the disease progression. Six compounds that target heme oxygenase signaling were found to rescue the abnormal muscle phenotype in sapje and sapje-like, while upregulating the inducible heme oxygenase 1 (Hmox1) at the protein level. Direct Hmox1 overexpression by injection of zebrafish Hmox1 mRNA into fertilized eggs was found to be sufficient for a dystrophin-independent restoration of normal muscle via an upregulation of cGMP levels. In addition, treatment of mdx(5cv) mice with the PDE5 inhibitor, sildenafil, which was one of the six drugs impacting the Hmox1 pathway in zebrafish, significantly increased the expression of Hmox1 protein, thus making Hmox1 a novel target for the improvement of dystrophic symptoms. These results demonstrate the translational relevance of our zebrafish model to mammalian models and support the use of zebrafish to screen for new drugs to treat human DMD. The discovery of a small molecule and a specific therapeutic pathway that might mitigate DMD disease progression could lead to significant clinical implications.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Distrofina/genética , Hemo-Oxigenasa 1/biosíntesis , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , GMP Cíclico/biosíntesis , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Distrofina/deficiencia , Hemo-Oxigenasa 1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , ARN Mensajero/genética , Transducción de Señal/genética , Citrato de Sildenafil , Sulfonas/farmacología , Regulación hacia Arriba , Pez Cebra/genética
3.
Nat Genet ; 37(3): 225-32, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15711547

RESUMEN

We combined large-scale mRNA expression analysis and gene mapping to identify genes and loci that control hematopoietic stem cell (HSC) function. We measured mRNA expression levels in purified HSCs isolated from a panel of densely genotyped recombinant inbred mouse strains. We mapped quantitative trait loci (QTLs) associated with variation in expression of thousands of transcripts. By comparing the physical transcript position with the location of the controlling QTL, we identified polymorphic cis-acting stem cell genes. We also identified multiple trans-acting control loci that modify expression of large numbers of genes. These groups of coregulated transcripts identify pathways that specify variation in stem cells. We illustrate this concept with the identification of candidate genes involved with HSC turnover. We compared expression QTLs in HSCs and brain from the same mice and identified both shared and tissue-specific QTLs. Our data are accessible through WebQTL, a web-based interface that allows custom genetic linkage analysis and identification of coregulated transcripts.


Asunto(s)
Genoma Humano , Células Madre Hematopoyéticas/citología , Proteínas Portadoras/genética , Humanos , Datos de Secuencia Molecular , Sitios de Carácter Cuantitativo , ARN Mensajero/genética
5.
Dev Cell ; 8(2): 153-66, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15691758

RESUMEN

Precise control of hematopoietic stem cell (HSC) proliferation and differentiation is needed to maintain a lifetime supply of blood cells. Using genome-wide ENU mutagenesis and phenotypic screening, we have identified a mouse line that harbors a point mutation in the transactivation (TA) domain of the transcription factor c-Myb (M303V), which reduces c-Myb-dependent TA by disrupting its interaction with the transcriptional coactivator p300. The biological consequences of the c-Myb(M303V/M303V) mutation include thrombocytosis, megakaryocytosis, anemia, lymphopenia, and the absence of eosinophils. Detailed analysis of hematopoiesis in c-Myb(M303V/M303V) mice reveals distinct blocks in T cell, B cell, and red blood cell development, as well as a remarkable 10-fold increase in the number of HSCs. Cell cycle analyses show that twice as many HSCs from c-Myb(M303V/M303V) animals are actively cycling. Thus c-Myb, through interaction with p300, controls the proliferation and differentiation of hematopoietic stem and progenitor cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas c-myb/fisiología , Transactivadores/fisiología , Animales , Linfocitos B/citología , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , ADN/genética , Proteína p300 Asociada a E1A , Femenino , Genes myb , Hematopoyesis/genética , Hematopoyesis/fisiología , Técnicas In Vitro , Masculino , Megacariocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Biológicos , Proteínas Nucleares/genética , Fenotipo , Mutación Puntual , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myb/química , Proteínas Proto-Oncogénicas c-myb/genética , Linfocitos T/citología , Trombocitosis/genética , Transactivadores/genética , Activación Transcripcional
6.
Mamm Genome ; 21(5-6): 247-57, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512339

RESUMEN

The Tail Suspension Test (TST), which measures behavioral despair, is widely used as an animal model of human depressive disorders and antidepressant efficacy. In order to identify novel genes involved in the regulation of TST performance, we crossed an inbred strain exhibiting low immobility in the TST (RIIIS/J) with two high-immobility strains (C57BL/6J and NZB/BlNJ) to create two distinct F2 hybrid populations. All F2 offspring (n = 655) were genotyped at high density with a panel of SNP markers. Whole-genome interval mapping of the F2 populations identified statistically significant quantitative trait loci (QTLs) on mouse chromosomes (MMU) 4, 6, and X. Microarray analysis of hippocampal gene expression in the three parental strains was used to identify potential candidate genes within the MMUX QTLs identified in the NZB/BlNJ x RIIIS/J cross. Expression of Gabra3, which encodes the GABA(A) receptor alpha3 subunit, was robust in the hippocampus of B6 and RIIIS mice but absent from NZB hippocampal tissue. To verify the role of Gabra3 in regulating TST behavior in vivo, mice were treated with SB-205384, a positive modulator of the alpha3 subunit. SB-205384 significantly reduced TST immobility in B6 mice without affecting general activity, but it had no effect on behavior in NZB mice. This work suggests that GABRA3 regulates a behavioral endophenotype of depression and establishes this gene as a viable new target for the study and treatment of human depression.


Asunto(s)
Conducta Animal , Sitios de Carácter Cuantitativo , Receptores de GABA-A/genética , Aminopiridinas , Animales , Cruzamientos Genéticos , Depresión/genética , Genotipo , Suspensión Trasera , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NZB , Tiofenos
7.
Drug Metab Dispos ; 37(6): 1269-76, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19324942

RESUMEN

Expression quantitative trait loci (eQTL) mapping can be used to identify the genetic variations that underlie inherited differences in gene transcription. We performed eQTL mapping by combining whole genome transcriptional data from the hypothalami of 33 strains of inbred mice with a detailed haplotype map of those same strains, revealing 10,655 trans associations and 31 cis eQTLs. One of the cis associations was found to be driven by strain-specific variation in the expression of Glutathione S-transferase, mu 5 (Gstm5). Gstm5 is one of seven members of the glutathione S-transferase, Mu family of genes. The glutathione S-transferases are phase II metabolic enzymes and are key regulators of drug and toxin clearance. In mouse, all seven family members are tightly clustered on mouse chromosome 3. Investigation of the Gstm5 cis association in multiple tissues types revealed that an 84-kilobase region on MMU3 acts as a haplotype-specific locus control region for the glutathione S-transferase, Mu cluster. In the strains that share the minor haplotype, drastic reductions in mRNA levels in multiple members of the Gst Mu family were observed. The strain-specific differences in Gst Mu transcription characterized here accurately model the human population, in which extreme variations in expression of GST Mu family members have been observed. Furthermore, the reduction in Gst Mu levels has important relevance for pharmacology and toxicology studies conducted in these strains. For instance, the reduced levels of Gst Mu in general and Gstm5 in particular have implications in models of dopamine metabolism, Parkinson's disease, and chemical neurotoxicity.


Asunto(s)
Perfilación de la Expresión Génica , Variación Genética , Glutatión Transferasa/genética , Sitios de Carácter Cuantitativo , Animales , Mapeo Cromosómico , Genoma , Genotipo , Glutatión Transferasa/metabolismo , Humanos , Células Híbridas , Masculino , Ratones , Ratones Endogámicos , Reacción en Cadena de la Polimerasa
8.
Neuropsychopharmacology ; 33(6): 1312-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17609676

RESUMEN

Despite widespread use of antidepressants, the factors underlying the behavioral response to antidepressants are unknown. It has been shown that antidepressant treatment promotes the proliferation and survival of neurons in the adult hippocampus via enhanced serotonergic signaling, but it is unclear whether hippocampal neurogenesis is responsible for the behavioral response to antidepressants. Furthermore, a large subpopulation of patients fails to respond to antidepressant treatment due to presumed underlying genetic factors. In the present study, we have used the phenotypic and genotypic variability of inbred mouse strains to show that there is a genetic component to both the behavioral and neuronal effects of chronic fluoxetine treatment, and that this antidepressant induces an increase in hippocampal cell proliferation only in the strains that also show a positive behavioral response to treatment. Furthermore, the behavioral and neuronal responses are associated with an upregulation of genes known to promote neuronal proliferation and survival. These results suggest that inherent genetic predisposition to increased serotonin-induced neurogenesis may be a determinant of antidepressant efficacy.


Asunto(s)
Conducta Animal/efectos de los fármacos , Fluoxetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Análisis de Varianza , Animales , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fluoxetina/análogos & derivados , Fluoxetina/sangre , Hipocampo/citología , Hipocampo/efectos de los fármacos , Pérdida de Tono Postural/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Inhibidores Selectivos de la Recaptación de Serotonina/sangre , Factores de Tiempo
9.
Addict Biol ; 13(1): 88-94, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18269382

RESUMEN

OPRL1 encodes the nociceptin receptor, which has been shown to be involved in alcohol dependence in previous studies. In the present study, we investigated the association between genetic polymorphisms of OPRL1 and alcohol dependence in a Scandinavian population. We genotyped 15 single nucleotide polymorphisms (SNPs) spanning the OPRL1 locus and found that SNP rs6010718 was significantly associated with both Type I and Type II alcoholics (P < 0.05). Linkage disequilibrium and haplotype analysis identified two haplotype blocks in this region. Furthermore, two haplotypes composed of five tag SNPs showed significant association with alcohol dependence. These findings suggest that genetic variants of the OPRL1 gene play a role in alcohol dependence in the Scandinavian population, warranting further investigation at the OPRL1 locus.


Asunto(s)
Alcoholismo/genética , Genotipo , Polimorfismo de Nucleótido Simple/genética , Receptores Opioides/genética , Adulto , Alcoholismo/rehabilitación , Alelos , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Haplotipos/genética , Humanos , Desequilibrio de Ligamiento , Masculino , Factores de Riesgo , Suecia , Receptor de Nociceptina
10.
PLoS Biol ; 2(12): e393, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15534693

RESUMEN

Rapid expansion of available data, both phenotypic and genotypic, for multiple strains of mice has enabled the development of new methods to interrogate the mouse genome for functional genetic perturbations. In silico mapping provides an expedient way to associate the natural diversity of phenotypic traits with ancestrally inherited polymorphisms for the purpose of dissecting genetic traits. In mouse, the current single nucleotide polymorphism (SNP) data have lacked the density across the genome and coverage of enough strains to properly achieve this goal. To remedy this, 470,407 allele calls were produced for 10,990 evenly spaced SNP loci across 48 inbred mouse strains. Use of the SNP set with statistical models that considered unique patterns within blocks of three SNPs as an inferred haplotype could successfully map known single gene traits and a cloned quantitative trait gene. Application of this method to high-density lipoprotein and gallstone phenotypes reproduced previously characterized quantitative trait loci (QTL). The inferred haplotype data also facilitates the refinement of QTL regions such that candidate genes can be more easily identified and characterized as shown for adenylate cyclase 7.


Asunto(s)
Mapeo Cromosómico , Biología Computacional/métodos , Polimorfismo de Nucleótido Simple , Adenilil Ciclasas/genética , Alelos , Animales , Cruzamientos Genéticos , Cálculos Biliares/metabolismo , Genoma , Haplotipos , Desequilibrio de Ligamiento , Lipoproteínas HDL/metabolismo , Modelos Logísticos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos , Modelos Genéticos , Modelos Estadísticos , Fenotipo , Filogenia , Sitios de Carácter Cuantitativo , Especificidad de la Especie
11.
J Bone Miner Res ; 21(1): 105-12, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16355279

RESUMEN

UNLABELLED: BMD is highly heritable; however, little is known about the genes. To identify loci controlling BMD, we conducted a QTL analysis in a (B6 x 129) F2 population of mice. We report on additional QTLs and also narrow one QTL by combining the data from multiple crosses and through haplotype analysis. INTRODUCTION: Previous studies have identified quantitative trait loci (QTL) that determine BMD in mice; however, identification of genes underlying QTLs is impeded by the large size of QTL regions. MATERIALS AND METHODS: To identify loci controlling BMD, we performed a QTL analysis of 291 (B6 x 129) F2 females. Total body and vertebral areal BMD (aBMD) were determined by peripheral DXA when mice were 20 weeks old and had consumed a high-fat diet for 14 weeks. RESULTS AND CONCLUSIONS: Two QTLs were common for both total body and vertebral aBMD: Bmd20 on chromosome (Chr) 6 (total aBMD; peak cM 26, logarithm of odds [LOD] 3.8, and vertebral aBMD; cM 32, LOD 3.6) and Bmd22 on Chr 1 (total aBMD; cM 104, LOD 2.5, and vertebral aBMD; cM 98, LOD 2.6). A QTL on Chr 10 (Bmd21, cM 68, LOD 3.0) affected total body aBMD and a QTL on Chr 7 (Bmd9, cM 44, LOD 2.7) affected vertebral aBMD. A pairwise genome-wide search did not reveal significant gene-gene interactions. Collectively, the QTLs accounted for 21.6% of total aBMD and 17.3% of vertebral aBMD of the F(2) population variances. Bmd9 was previously identified in a cross between C57BL/6J and C3H/HeJ mice, and we narrowed this QTL from 34 to 22 cM by combining the data from these crosses. By examining the Bmd9 region for conservation of ancestral alleles among the low allele strains (129S1/SvImJ and C3H/HeJ) that differed from the high allele strain (C57BL/6J), we further narrowed the region to approximately 9.9 cM, where the low allele strains share a common haplotype. Identifying the genes for these QTLs will enhance our understanding of skeletal biology.


Asunto(s)
Densidad Ósea/genética , Cruzamientos Genéticos , Sitios de Carácter Cuantitativo/genética , Animales , Haplotipos , Ratones , Ratones Endogámicos
12.
BMC Bioinformatics ; 7: 61, 2006 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-16466585

RESUMEN

BACKGROUND: Finding the genetic causes of quantitative traits is a complex and difficult task. Classical methods for mapping quantitative trail loci (QTL) in miceuse an F2 cross between two strains with substantially different phenotype and an interval mapping method to compute confidence intervals at each position in the genome. This process requires significant resources for breeding and genotyping, and the data generated are usually only applicable to one phenotype of interest. Recently, we reported the application of a haplotype association mapping method which utilizes dense genotyping data across a diverse panel of inbred mouse strains and a marker association algorithm that is independent of any specific phenotype. As the availability of genotyping data grows in size and density, analysis of these haplotype association mapping methods should be of increasing value to the statistical genetics community. RESULTS: We describe a detailed comparative analysis of variations on our marker association method. In particular, we describe the use of inferred haplotypes from adjacent SNPs, parametric and nonparametric statistics, and control of multiple testing error. These results show that nonparametric methods are slightly better in the test cases we study, although the choice of test statistic may often be dependent on the specific phenotype and haplotype structure being studied. The use of multi-SNP windows to infer local haplotype structure is critical to the use of a diverse panel of inbred strains for QTL mapping. Finally, because the marginal effect of any single gene in a complex disease is often relatively small, these methods require the use of sensitive methods for controlling family-wise error. We also report our initial application of this method to phenotypes cataloged in the Mouse Phenome Database. CONCLUSION: The use of inbred strains of mice for QTL mapping has many advantages over traditional methods. However, there are also limitations in comparison to the traditional linkage analysis from F2 and RI lines. Application of these methods requires careful consideration of algorithmic choices based on both theoretical and practical factors. Our findings suggest general guidelines, though a complete evaluation of these methods can only be performed as more genetic data in complex diseases becomes available.


Asunto(s)
Algoritmos , Mapeo Cromosómico/métodos , Análisis Mutacional de ADN/métodos , Haplotipos/genética , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Análisis de Secuencia de ADN/métodos , Animales , Variación Genética/genética , Ratones , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Alineación de Secuencia/métodos
14.
Mol Cancer ; 4: 28, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-16083501

RESUMEN

BACKGROUND: Introduction of cDNA or genomic clones of the tumor suppressor in lung cancer 1 (TSLC1) gene into the non-small cell lung cancer line, A549, reverses tumorigenic growth properties of these cells. These results and the observation that TSLC1 is down-regulated in a number of tumors suggest that TSLC1 functions as a critical switch mediating repression of tumorigenesis. RESULTS: To investigate this mechanism, we compared growth properties of A549 with the TSLC1-containing derivative. We found a G1/S phase transition delay in 12.2. Subtractive hybridization, quantitative PCR, and TranSignal Protein/DNA arrays were used to identify genes whose expression changed when TSLC1 was up-regulated. Members of common G1/S phase regulatory pathways such as TP53, MYC, RB1 and HRAS were not differentially expressed, indicating that TSLC1 may function through an alternative pathway(s). A number of genes involved in cell proliferation and tumorigenesis were differentially expressed, notably genes in the Ras-induced senescence pathway. We examined expression of several of these key genes in human tumors and normal lung tissue, and found similar changes in expression, validating the physiological relevance of the A549 and 12.2 cell lines. CONCLUSION: Gene expression and cell cycle differences provide insights into potential downstream pathways of TSLC1 that mediate the suppression of tumor properties in A549 cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Regulación Neoplásica de la Expresión Génica , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Inmunoglobulinas/genética , Pulmón/metabolismo , Proteínas de la Membrana/genética , Proteómica , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética
15.
Chem Biol ; 22(12): 1588-96, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26670081

RESUMEN

Bromodomains are involved in transcriptional regulation through the recognition of acetyl lysine modifications on diverse proteins. Selective pharmacological modulators of bromodomains are lacking, although the largely hydrophobic nature of the pocket makes these modules attractive targets for small-molecule inhibitors. This work describes the structure-based design of a highly selective inhibitor of the CREB binding protein (CBP) bromodomain and its use in cell-based transcriptional profiling experiments. The inhibitor downregulated a number of inflammatory genes in macrophages that were not affected by a selective BET bromodomain inhibitor. In addition, the CBP bromodomain inhibitor modulated the mRNA level of the regulator of G-protein signaling 4 (RGS4) gene in neurons, suggesting a potential therapeutic opportunity for CBP inhibitors in the treatment of neurological disorders.


Asunto(s)
Proteína de Unión a CREB/antagonistas & inhibidores , Diseño de Fármacos , Bibliotecas de Moléculas Pequeñas/química , Proteína de Unión a CREB/genética , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Estructura Terciaria de Proteína , Proteínas RGS/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Transcriptoma
16.
Invest Ophthalmol Vis Sci ; 56(4): 2737-48, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25813999

RESUMEN

PURPOSE: The pathophysiology of ocular hypertension (OH) leading to primary open-angle glaucoma shares many features with a secondary form of OH caused by treatment with glucocorticoids, but also exhibits distinct differences. In this study, a pharmacogenomics approach was taken to discover candidate genes for this disorder. METHODS: A genome-wide association study was performed, followed by an independent candidate gene study, using a cohort enrolled from patients treated with off-label intravitreal triamcinolone, and handling change in IOP as a quantitative trait. RESULTS: An intergenic quantitative trait locus (QTL) was identified at chromosome 6p21.33 near the 5' end of HCG22 that attained the accepted statistical threshold for genome-level significance. The HCG22 transcript, encoding a novel mucin protein, was expressed in trabecular meshwork cells, and expression was stimulated by IL-1, and inhibited by triamcinolone acetate and TGF-ß. Bioinformatic analysis defined the QTL as an approximately 4 kilobase (kb) linkage disequilibrium block containing 10 common single nucleotide polymorphisms (SNPs). Four of these SNPs were identified in the National Center for Biotechnology Information (NCBI) GTEx eQTL browser as modifiers of HCG22 expression. Most are predicted to disrupt or improve motifs for transcription factor binding, the most relevant being disruption of the glucocorticoid receptor binding motif. A second QTL was identified within the predicted signal peptide of the HCG22 encoded protein that could affect its secretion. Translation, O-glycosylation, and secretion of the predicted HCG22 protein was verified in cultured trabecular meshwork cells. CONCLUSIONS: Identification of two independent QTLs that could affect expression of the HCG22 mucin gene product via two different mechanisms (transcription or secretion) is highly suggestive of a role in steroid-induced OH.


Asunto(s)
Regulación de la Expresión Génica , Presión Intraocular/efectos de los fármacos , Mucinas/genética , Hipertensión Ocular/genética , ARN Mensajero/genética , Triamcinolona/efectos adversos , Adulto , Femenino , Estudios de Seguimiento , Estudio de Asociación del Genoma Completo , Genotipo , Glucocorticoides/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Mucinas/biosíntesis , Hipertensión Ocular/inducido químicamente , Hipertensión Ocular/metabolismo , Malla Trabecular/metabolismo
17.
Reprod Toxicol ; 45: 77-86, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24434083

RESUMEN

Many of the commonly observed reproductive toxicities associated with therapeutic compounds can be traced to a disruption of the steroidogenic pathway. We sought to develop an in vitro assay that would predict reproductive toxicity and be high throughput in nature. H295R cells, previously validated as having an intact and functional steroidogenic pathway, were treated with 83 known-positive and 79 known-negative proprietary and public-domain compounds. The assay measured the expression of the key enzymes STAR, 3ßHSD2, CYP17A1, CYP11B2, CYP19A1, CYP21A2, and CYP11A1 and the hormones DHEA, progesterone, testosterone, and cortisol. We found that a Random Forest model yielded a receiver operating characteristic area under the curve (ROC AUC) of 0.845, with sensitivity of 0.724 and specificity of 0.758 for predicting in vivo reproductive toxicity with this in vitro assay system.


Asunto(s)
Evaluación Preclínica de Medicamentos , Modelos Biológicos , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Línea Celular Tumoral , Colforsina/toxicidad , Sistema Enzimático del Citocromo P-450/metabolismo , Deshidroepiandrosterona/metabolismo , Humanos , Hidrocortisona/metabolismo , Imidazoles/toxicidad , Modelos Estadísticos , Fosfoproteínas/metabolismo , Progesterona/metabolismo , Testosterona/metabolismo
18.
Toxicol Sci ; 140(2): 481-92, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24848797

RESUMEN

Isoniazid (INH), the mainstay therapeutic for tuberculosis infection, has been associated with rare but serious hepatotoxicity in the clinic. However, the mechanisms underlying inter-individual variability in the response to this drug have remained elusive. A genetically diverse mouse population model in combination with a systems biology approach was utilized to identify transcriptional changes, INH-responsive metabolites, and gene variants that contribute to the liver response in genetically sensitive individuals. Sensitive mouse strains developed severe microvesicular steatosis compared with corresponding vehicle control mice following 3 days of oral treatment with INH. Genes involved in mitochondrial dysfunction were enriched among liver transcripts altered with INH treatment. Those associated with INH treatment and susceptibility to INH-induced steatosis in the liver included apolipoprotein A-IV, lysosomal-associated membrane protein 1, and choline phosphotransferase 1. These alterations were accompanied by metabolomic changes including reduced levels of glutathione and the choline metabolites betaine and phosphocholine, suggesting that oxidative stress and reduced lipid export may additionally contribute to INH-induced steatosis. Finally, genome-wide association mapping revealed that polymorphisms in perilipin 2 were linked to increased triglyceride levels following INH treatment, implicating a role for inter-individual differences in lipid packaging in the susceptibility to INH-induced steatosis. Taken together, our data suggest that INH-induced steatosis is caused by not one, but multiple events involving lipid retention in the livers of genetically sensitive individuals. This work also highlights the value of using a mouse diversity panel to investigate drug-induced responses across a diverse population.


Asunto(s)
Antituberculosos/efectos adversos , Hígado Graso/inducido químicamente , Isoniazida/efectos adversos , Biología de Sistemas , Animales , Colesterol/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Proteínas de la Membrana/genética , Metabolómica , Ratones , Ratones Endogámicos , Perilipina-2 , Polimorfismo de Nucleótido Simple
19.
BMC Pharmacol Toxicol ; 14: 46, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-24010585

RESUMEN

Drug-induced cardiac toxicity has been implicated in 31% of drug withdrawals in the USA. The fact that the risk for cardiac-related adverse events goes undetected in preclinical studies for so many drugs underscores the need for better, more predictive in vitro safety screens to be deployed early in the drug discovery process. Unfortunately, many questions remain about the ability to accurately translate findings from simple cellular systems to the mechanisms that drive toxicity in the complex in vivo environment. In this study, we analyzed translatability of cardiotoxic effects for a diverse set of drugs from rodents to two different cell systems (rat heart tissue-derived cells (H9C2) and primary rat cardiomyocytes (RCM)) based on their transcriptional response. To unravel the altered pathway, we applied a novel computational systems biology approach, the Causal Reasoning Engine (CRE), to infer upstream molecular events causing the observed gene expression changes. By cross-referencing the cardiotoxicity annotations with the pathway analysis, we found evidence of mechanistic convergence towards common molecular mechanisms regardless of the cardiotoxic phenotype. We also experimentally verified two specific molecular hypotheses that translated well from in vivo to in vitro (Kruppel-like factor 4, KLF4 and Transforming growth factor beta 1, TGFB1) supporting the validity of the predictions of the computational pathway analysis. In conclusion, this work demonstrates the use of a novel systems biology approach to predict mechanisms of toxicity such as KLF4 and TGFB1 that translate from in vivo to in vitro. We also show that more complex in vitro models such as primary rat cardiomyocytes may not offer any advantage over simpler models such as immortalized H9C2 cells in terms of translatability to in vivo effects if we consider the right endpoints for the model. Further assessment and validation of the generated molecular hypotheses would greatly enhance our ability to design predictive in vitro cardiotoxicity assays.


Asunto(s)
Biología Computacional/métodos , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Corazón/efectos de los fármacos , Modelos Cardiovasculares , Preparaciones Farmacéuticas , Adenosina Trifosfato/metabolismo , Animales , Causalidad , Biología Computacional/estadística & datos numéricos , Evaluación Preclínica de Medicamentos/estadística & datos numéricos , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Valor Predictivo de las Pruebas , Ratas , Factor de Crecimiento Transformador beta1/genética
20.
Mol Autism ; 4(1): 45, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24238429

RESUMEN

BACKGROUND: Austism spectrum disorder (ASD) is a heterogeneous behavioral disorder or condition characterized by severe impairment of social engagement and the presence of repetitive activities. The molecular etiology of ASD is still largely unknown despite a strong genetic component. Part of the difficulty in turning genetics into disease mechanisms and potentially new therapeutics is the sheer number and diversity of the genes that have been associated with ASD and ASD symptoms. The goal of this work is to use shRNA-generated models of genetic defects proposed as causative for ASD to identify the common pathways that might explain how they produce a core clinical disability. METHODS: Transcript levels of Mecp2, Mef2a, Mef2d, Fmr1, Nlgn1, Nlgn3, Pten, and Shank3 were knocked-down in mouse primary neuron cultures using shRNA constructs. Whole genome expression analysis was conducted for each of the knockdown cultures as well as a mock-transduced culture and a culture exposed to a lentivirus expressing an anti-luciferase shRNA. Gene set enrichment and a causal reasoning engine was employed to identify pathway level perturbations generated by the transcript knockdown. RESULTS: Quantification of the shRNA targets confirmed the successful knockdown at the transcript and protein levels of at least 75% for each of the genes. After subtracting out potential artifacts caused by viral infection, gene set enrichment and causal reasoning engine analysis showed that a significant number of gene expression changes mapped to pathways associated with neurogenesis, long-term potentiation, and synaptic activity. CONCLUSIONS: This work demonstrates that despite the complex genetic nature of ASD, there are common molecular mechanisms that connect many of the best established autism candidate genes. By identifying the key regulatory checkpoints in the interlinking transcriptional networks underlying autism, we are better able to discover the ideal points of intervention that provide the broadest efficacy across the diverse population of autism patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA