Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Psychiatry ; 23(9): 1900-1910, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-28848234

RESUMEN

Alcohol use disorder (AUD) is a common and chronic disorder with substantial effects on personal and public health. The underlying pathophysiology is poorly understood but strong evidence suggests significant roles of both genetic and epigenetic components. Given that alcohol affects many organ systems, we performed a cross-tissue and cross-phenotypic analysis of genome-wide methylomic variation in AUD using samples from 3 discovery, 4 replication, and 2 translational cohorts. We identified a differentially methylated region in the promoter of the proprotein convertase subtilisin/kexin 9 (PCSK9) gene that was associated with disease phenotypes. Biological validation showed that PCSK9 promoter methylation is conserved across tissues and positively correlated with expression. Replication in AUD datasets confirmed PCSK9 hypomethylation and a translational mouse model of AUD showed that alcohol exposure leads to PCSK9 downregulation. PCSK9 is primarily expressed in the liver and regulates low-density lipoprotein cholesterol (LDL-C). Our finding of alcohol-induced epigenetic regulation of PCSK9 represents one of the underlying mechanisms between the well-known effects of alcohol on lipid metabolism and cardiovascular risk, with light alcohol use generally being protective while chronic heavy use has detrimental health outcomes.


Asunto(s)
Alcoholismo/genética , Proproteína Convertasa 9/efectos de los fármacos , Proproteína Convertasa 9/genética , Adulto , Alcoholismo/fisiopatología , Animales , LDL-Colesterol/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Epigenómica/métodos , Etanol/efectos adversos , Etanol/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Humanos , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Ratones , Fenotipo , Regiones Promotoras Genéticas/genética , Proproteína Convertasa 9/fisiología , Ratas , Ratas Wistar
2.
Pharmacogenomics J ; 14(3): 303-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24126707

RESUMEN

Two commonly prescribed treatments for opioid addiction are methadone and buprenorphine. Although these drugs show some efficacy in treating opioid dependence, treatment response varies among individuals. It is likely that genetic factors have a role in determining treatment outcome. This study analyses the pharmacogenetic association of six polymorphisms in OPRD1, the gene encoding the delta-opioid receptor, on treatment outcome in 582 opioid addicted European Americans randomized to either methadone or buprenorphine/naloxone (Suboxone) over the course of a 24-week open-label clinical trial. Treatment outcome was assessed as the number of missed or opioid-positive urine drug screens over the 24 weeks. In the total sample, no single-nucleotide polymorphisms (SNPs) in OPRD1 were significantly associated with treatment outcome in either treatment arm. However, sex-specific analyses revealed two intronic SNPs (rs581111 and rs529520) that predicted treatment outcome in females treated with buprenorphine. Females with the AA or AG genotypes at rs581111 had significantly worse outcomes than those with the GG genotype when treated with buprenorphine (P=0.03, relative risk (RR)=1.67, 95% confidence interval (CI) 1.06-2.1). For rs529520, females with the AA genotype had a significantly worse outcome than those with the CC genotype when (P=0.006, RR=2.15, 95% CI 1.3-2.29). No significant associations were detected in males. These findings suggest that rs581111 and rs52920 may be useful when considering treatment options for female opioid addicts, however, confirmation in an independent sample is warranted.


Asunto(s)
Buprenorfina/uso terapéutico , Variación Genética , Trastornos Relacionados con Opioides/tratamiento farmacológico , Receptores Opioides delta/genética , Femenino , Humanos , Trastornos Relacionados con Opioides/genética , Población Blanca
3.
Mol Psychiatry ; 19(1): 129-39, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23337945

RESUMEN

Emotional behavior is in part heritable and often disrupted in psychopathology. Identification of specific genetic variants that drive this heritability may provide important new insight into molecular and neurobiological mechanisms involved in emotionality. Our results demonstrate that the presynaptic vesicular monoamine transporter 1 (VMAT1) Thr136Ile (rs1390938) polymorphism is functional in vitro, with the Ile allele leading to increased monoamine transport into presynaptic vesicles. Moreover, we show that the Thr136Ile variant predicts differential responses in emotional brain circuits consistent with its effects in vitro. Lastly, deep sequencing of bipolar disorder (BPD) patients and controls identified several rare novel VMAT1 variants. The variant Phe84Ser was only present in individuals with BPD and leads to marked increase monoamine transport in vitro. Taken together, our data show that VMAT1 polymorphisms influence monoamine signaling, the functional response of emotional brain circuits and risk for psychopathology.


Asunto(s)
Síntomas Afectivos/genética , Emociones/fisiología , Polimorfismo Genético/genética , Proteínas de Transporte Vesicular de Monoaminas/genética , Adolescente , Síntomas Afectivos/patología , Animales , Monoaminas Biogénicas/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Línea Celular Transformada , Chlorocebus aethiops , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Transfección , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Adulto Joven
5.
Pharmacogenomics J ; 13(5): 464-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22907732

RESUMEN

Variation in genes involved in serotonergic signaling is thought to be associated with antidepressant treatment response in generalized anxiety disorder (GAD). We examined a possible interaction between the serotonin transporter gene (SLC6A4) 5-HTTLPR/rs25531 haplotype and the serotonin 2A receptor gene (HTR2A) single-nucleotide polymorphism (SNP) rs7997012 in antidepressant treatment outcome in GAD. Patients diagnosed with GAD received venlafaxine XR treatment as part of an 18-month relapse prevention study. Genotypes obtained for the 5-HTTLPR/rs25531 (La/La, La/S or S/S) haplotype and rs7997012 SNP (G or A) in the European American population (n=112) were used for pharmacogenetic analysis. Our data show that subjects with genotypes La/La+G/G or La/La+G/A (n=28) had significantly lower Hamilton Anxiety Scale (HAM-A) scores than those with genotypes La/S+A/A or S/S+A/A (n=12) at 6 months (HAM-A difference=10.7; P<0.0001). Single-marker analysis only showed HAM-A differences of 4.3 (5-HTTLPR/rs25531: La/La versus La/S+S/S) and 4.8 (rs7997012: G/G+G/A versus A/A), showing for the first time a significant gene-gene interaction between these markers.


Asunto(s)
Antidepresivos/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/genética , Ciclohexanoles/uso terapéutico , Receptor de Serotonina 5-HT2A/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Biomarcadores Farmacológicos/metabolismo , Método Doble Ciego , Haplotipos , Humanos , Farmacogenética/métodos , Polimorfismo de Nucleótido Simple , Clorhidrato de Venlafaxina , Población Blanca/genética
6.
Drug Alcohol Depend ; 127(1-3): 122-8, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22795689

RESUMEN

BACKGROUND: Addiction susceptibility and treatment responsiveness are greatly influenced by genetic factors. Sequence variation in genes involved in the mechanisms of drug action have the potential to influence addiction risk and treatment outcome. The opioid receptor system is involved in mediating the rewarding effects of cocaine and opioids. The µ-opioid receptor (MOR) has traditionally been considered the primary target for opioid addiction. The MOR, however, interacts with and is regulated by many known MOR interacting proteins (MORIPs), including the δ-opioid receptor (DOR). METHODS: The present study evaluated the contribution of OPRD1, the gene encoding the DOR, to the risk of addiction to opioids and cocaine. The association of OPRD1 polymorphisms with both opioid addiction (OA) and cocaine addiction (CA) was analyzed in African American (OA n=336, CA n=503) and European American (OA n=1007, CA n=336) populations. RESULTS: The primary finding of this study is an association of rs678849 with cocaine addiction in African Americans (allelic p=0.0086). For replication purposes, this SNP was analyzed in a larger independent population of cocaine addicted African Americans and controls and the association was confirmed (allelic p=4.53 × 10(-5); n=993). By performing a meta-analysis on the expanded populations, the statistical evidence for an association was substantially increased (allelic p=8.5 × 10(-7)) (p-values non-FDR corrected). CONCLUSION: The present study suggests that polymorphisms in OPRD1 are relevant for cocaine addiction in the African American population and provides additional support for a broad role for OPRD1 variants in drug dependence.


Asunto(s)
Negro o Afroamericano/genética , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Opioides/genética , Polimorfismo de Nucleótido Simple/genética , Receptores Opioides delta/genética , Población Blanca/genética , Estudios de Casos y Controles , Trastornos Relacionados con Cocaína/diagnóstico , Femenino , Humanos , Masculino , Trastornos Relacionados con Opioides/diagnóstico , Vigilancia de la Población/métodos
7.
Neuroscience ; 232: 32-44, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23201251

RESUMEN

Vesicular monoamine transporters (VMAT) are involved in presynaptic storage and release of neurotransmitters. While it was thought initially that only VMAT2 is brain expressed and VMAT1 is present only in the periphery, recent data have challenged the exclusive expression of VMAT2 in the brain. To further elucidate the role of VMAT1 brain expression and its potential role in neuropsychiatric disorders, we have investigated mice lacking VMAT1. Comparison of wildtype and knock-out (KO) mice using qPCR and immunohistochemistry documents the expression of VMAT1 in the brain. Deletion of VMAT1 leads to increased hippocampal apoptosis and reduced neurogenesis as assessed by caspase-3-labeling and 5-bromo-deoxy-uridine-labeling. Behavioral data show that mice lacking VMAT1 have neurocognitive deficits. VMAT2 expression is not altered in VMAT1 KO mice, suggesting a distinct role of VMAT1. Our data support VMAT1 brain expression and suggest that VMAT1 plays a key role in survival of hippocampal neurons and thus might contribute to neurocognitive deficits observed in neuropsychiatric disorders.


Asunto(s)
Encéfalo/fisiopatología , Trastornos del Conocimiento/fisiopatología , Discriminación en Psicología/fisiología , Neuronas/patología , Percepción Espacial/fisiología , Proteínas de Transporte Vesicular de Monoaminas/deficiencia , Animales , Apoptosis/fisiología , Encéfalo/patología , Caspasa 3/metabolismo , Trastornos del Conocimiento/patología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Masculino , Ratones Noqueados , Neurogénesis/fisiología , Neuronas/fisiología , ARN Mensajero/metabolismo , Reconocimiento en Psicología/fisiología , Sinaptofisina/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/genética , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
8.
Pharmacogenomics J ; 13(1): 21-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22006095

RESUMEN

Generalized anxiety disorder (GAD) is a chronic psychiatric disorder with significant morbidity and mortality. Antidepressant drugs are the preferred choice for treatment; however, treatment response is often variable. Several studies in major depression have implicated a role of the serotonin receptor gene (HTR2A) in treatment response to antidepressants. We tested the hypothesis that the genetic polymorphism rs7997012 in the HTR2A gene predicts treatment outcome in GAD patients treated with venlafaxine XR. Treatment response was assessed in 156 patients that participated in a 6-month open-label clinical trial of venlafaxine XR for GAD. Primary analysis included Hamilton Anxiety Scale (HAM-A) reduction at 6 months. Secondary outcome measure was the Clinical Global Impression of Improvement (CGI-I) score at 6 months. Genotype and allele frequencies were compared between groups using χ(2) contingency analysis. The frequency of the G-allele differed significantly between responders (70%) and nonresponders (56%) at 6 months (P=0.05) using the HAM-A scale as outcome measure. Similarly, using the CGI-I as outcome, the G-allele was significantly associated with improvement (P=0.01). Assuming a dominant effect of the G-allele, improvement differed significantly between groups (P=0.001, odds ratio=4.72). Similar trends were observed for remission although not statistically significant. We show for the first time a pharmacogenetic effect of the HTR2A rs7997012 variant in anxiety disorders, suggesting that pharmacogenetic effects cross diagnostic categories. Our data document that individuals with the HTR2A rs7997012 single nucleotide polymorphism G-allele have better treatment outcome over time. Future studies with larger sample sizes are necessary to further characterize this effect in treatment response to antidepressants in GAD.


Asunto(s)
Antidepresivos/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/genética , Ciclohexanoles/uso terapéutico , Receptores de Serotonina 5-HT2/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Resultado del Tratamiento , Clorhidrato de Venlafaxina , Adulto Joven
9.
Genes Brain Behav ; 11(4): 415-23, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22443215

RESUMEN

Genetic factors are believed to account for 30-50% of the risk for cocaine and heroin addiction. Dynorphin peptides, derived from the prodynorphin (PDYN) precursor, bind to opioid receptors, preferentially the kappa-opioid receptor, and may mediate the aversive effects of drugs of abuse. Dynorphin peptides produce place aversion in animals and produce dysphoria in humans. Cocaine and heroin have both been shown to increase expression of PDYN in brain regions relevant for drug reward and use. Polymorphisms in PDYN are therefore hypothesized to increase risk for addiction to drugs of abuse. In this study, 3 polymorphisms in PDYN (rs1022563, rs910080 and rs1997794) were genotyped in opioid-addicted [248 African Americans (AAs) and 1040 European Americans (EAs)], cocaine-addicted (1248 AAs and 336 EAs) and control individuals (674 AAs and 656 EAs). Sex-specific analyses were also performed as a previous study identified PDYN polymorphisms to be more significantly associated with female opioid addicts. We found rs1022563 to be significantly associated with opioid addiction in EAs [P = 0.03, odds ratio (OR) = 1.31; false discovery rate (FDR) corrected q-value]; however, when we performed female-specific association analyses, the OR increased from 1.31 to 1.51. Increased ORs were observed for rs910080 and rs199774 in female opioid addicts also in EAs. No statistically significant associations were observed with cocaine or opioid addiction in AAs. These data show that polymorphisms in PDYN are associated with opioid addiction in EAs and provide further evidence that these risk variants may be more relevant in females.


Asunto(s)
Trastornos Relacionados con Cocaína/genética , Encefalinas/genética , Dependencia de Heroína/genética , Polimorfismo de Nucleótido Simple , Precursores de Proteínas/genética , Adolescente , Adulto , Negro o Afroamericano/genética , Alelos , Conducta Adictiva/genética , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Factores Sexuales , Población Blanca/genética
10.
Genes Brain Behav ; 10(3): 309-15, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21129161

RESUMEN

We mapped the quantitative trait loci (QTL) that contribute to the robust difference in maximal electroshock seizure threshold (MEST) between C57BLKS/J (BKS) and C57BL10S/J (B10S) mice. BKS, B10S, BKS × B10S F1 and BKS × B10S F2 intercross mice were tested for MEST at 8-9 weeks of age. Results of F2 testing showed that, in this cross, MEST is a continuously distributed trait determined by polygenic inheritance. Mice from the extremes of the trait distribution were genotyped using microarray technology. MEST correlated significantly with body weight and sex; however, because of the high correlation between these factors, the QTL mapping was conditioned on sex alone. A sequential series of statistical analyses was used to map QTLs including single-point, multipoint and multilocus methods. Two QTLs reached genome-wide levels of significance based upon an empirically determined permutation threshold: chromosome 6 (LOD = 6.0 at ∼69 cM) and chromosome 8 (LOD = 5.7 at ∼27 cM). Two additional QTLs were retained in a multilocus regression model: chromosome 3 (LOD = 2.1 at ∼68 cM) and chromosome 5 (LOD = 2.7 at ∼73 cM). Together the four QTLs explain one third of the total phenotypic variance in the mapping population. Lack of overlap between the major MEST QTLs mapped here in BKS and B10S mice and those mapped previously in C57BL/6J and DBA/2J mice (strains that are closely related to BKS and B10S) suggest that BKS and B10S represent a new polygenic mouse model for investigating susceptibility to seizures.


Asunto(s)
Mapeo Cromosómico/métodos , Epilepsia/genética , Predisposición Genética a la Enfermedad/genética , Sitios de Carácter Cuantitativo/genética , Animales , Química Encefálica/genética , Modelos Animales de Enfermedad , Estimulación Eléctrica/efectos adversos , Estimulación Eléctrica/métodos , Epilepsia/fisiopatología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
11.
Genes Brain Behav ; 10(1): 57-68, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20731721

RESUMEN

Aggressive behaviors are disabling, treatment refractory, and sometimes lethal symptoms of several neuropsychiatric disorders. However, currently available treatments for patients are inadequate, and the underlying genetics and neurobiology of aggression is only beginning to be elucidated. Inbred mouse strains are useful for identifying genomic regions, and ultimately the relevant gene variants (alleles) in these regions, that affect mammalian aggressive behaviors, which, in turn, may help to identify neurobiological pathways that mediate aggression. The BALB/cJ inbred mouse strain exhibits relatively high levels of intermale aggressive behaviors and shows multiple brain and behavioral phenotypes relevant to neuropsychiatric syndromes associated with aggression. The A/J strain shows very low levels of aggression. We hypothesized that a cross between BALB/cJ and A/J inbred strains would reveal genomic loci that influence the tendency to initiate intermale aggressive behavior. To identify such loci, we conducted a genomewide scan in an F2 population of 660 male mice bred from BALB/cJ and A/J inbred mouse strains. Three significant loci on chromosomes 5, 10 and 15 that influence aggression were identified. The chromosome 5 and 15 loci are completely novel, and the chromosome 10 locus overlaps an aggression locus mapped in our previous study that used NZB/B1NJ and A/J as progenitor strains. Haplotype analysis of BALB/cJ, NZB/B1NJ and A/J strains showed three positional candidate genes in the chromosome 10 locus. Future studies involving fine genetic mapping of these loci as well as additional candidate gene analysis may lead to an improved biological understanding of mammalian aggressive behaviors.


Asunto(s)
Agresión/fisiología , Agresión/psicología , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/fisiología , Cruzamientos Genéticos , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Genotipo , Haplotipos , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Fenotipo , Especificidad de la Especie
12.
Physiol Genomics ; 42A(1): 1-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20571108

RESUMEN

To confirm seizure susceptibility (SZS) quantitative trait loci (QTLs) on chromosome (chr) 15 identified previously using C57BL/6J (B6) and DBA/2J (D2) mice and to refine their genomic map position, we studied a set of three congenic strains in which overlapping segments of chr 15 from D2 were transferred onto the B6 background. We measured thresholds for generalized electroshock seizure (GEST) and maximal electroshock seizure (MEST) in congenic strains and B6-like littermates and also tested their responses to kainic acid (KA) and pentylenetetrazol (PTZ). Results document that MEST is significantly lower in strains 15M and 15D, which harbor medial and distal (telomeric) segments of chr 15 (respectively) from D2, compared with strain 15P, which harbors the proximal (acromeric) segment of chr 15 from D2, and with control littermates. Congenic strains 15P and 15M exhibited greater KA SZS compared with strain 15D and B6-like controls. All congenic strains were similar to controls with regard to PTZ SZS. Taken together, results suggest there are multiple SZS QTLs on chr 15 and that two QTLs harbor gene variants that affect MEST and KA SZS independently. The MEST QTL is refined to a 19 Mb region flanked by rs13482630 and D15Mit159. This interval contains 350 genes, 183 of which reside in areas where the polymorphism rate between B6 and D2 is high. The KA QTL interval spans a 65 Mb region flanked by markers D15Mit13 and rs31271969. It harbors 83 genes in highly polymorphic areas, 310 genes in all. Complete dissection of these loci will lead to identification of genetic variants that influence SZS in mice and provide a better understanding of seizure biology.


Asunto(s)
Cromosomas de los Mamíferos/genética , Predisposición Genética a la Enfermedad/genética , Sitios de Carácter Cuantitativo/genética , Convulsiones/genética , Animales , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
13.
Am J Med Genet B Neuropsychiatr Genet ; 139B(1): 51-3, 2005 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-16152572

RESUMEN

Recent studies have indicated that the brain-derived neurotrophic factor (BDNF) gene is involved in the etiology of bipolar disorder (BPD). Two family-based association studies showed that the Val allele of the functional polymorphism Val66Met in the BDNF gene is associated with BPD; however, others could not confirm the results. Here we performed a replication study in an independent sample and tested the hypothesis that the Val66 allele in the BDNF gene confers susceptibility to bipolar I disorder (BPI). Six hundred twenty-one patients with BPI and 998 control subjects were genotyped for the Val66Met polymorphism. All cases and controls were of European descent. All BPI patients had a positive family history of affective disorder. The frequency of the Val allele was significantly increased in BPI patient when compared to controls (chi2 = 4.8; df = 1; P = 0.028; two-sided; OR = 1.22; 95% CI: 1.02-1.47). Results confirm previous findings and suggest that the Val allele increases risk for BPI in patients of European descent. Further studies are necessary to elucidate the involvement of the BDNF gene in the pathophysiology of BPD.


Asunto(s)
Trastorno Bipolar/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Polimorfismo Genético , Valina/genética , Adulto , Alelos , Sustitución de Aminoácidos , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Metionina/genética , Población Blanca
14.
Epilepsy Res ; 63(2-3): 113-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15725393

RESUMEN

PURPOSE: Quantitative trait loci (QTL) mapping in mice revealed a seizure-related QTL (Szs1), for which the inward-rectifying potassium channel Kcnj10 is the most compelling candidate gene. Association analysis of the human KCNJ10 gene identified a common KCNJ10 missense variation (Arg271Cys) that influences susceptibility to focal and generalized epilepsies. The present replication study tested the initial finding that the KCNJ10 Cys271 allele is associated with seizure resistance to common syndromes of idiopathic generalized epilepsy (IGE). METHODS: The study sample comprised 563 German IGE patients and 660 healthy population controls. To search for seizure type-specific effects, two IGE subgroups were formed, comprising 258 IGE patients with typical absences (IAE group) and 218 patients with juvenile myoclonic epilepsy (JME group). A TaqMan nuclease assay was used to genotype the KCNJ10 single nucleotide polymorphism c.1037C > T (dbSNP: rs1130183) that alters amino acid at position 271 from arginine to cysteine. RESULTS: Replication analysis revealed a significant decrease of the Cys271 allele frequency in 446 IGE patients compared to controls (chi2 = 3.52, d.f. = 1, P = 0.030, one-sided; OR(Cys271+) = 0.69; 95% CI: 0.50-0.95). Among the IGE subgroups, lack of the Cys271 allele was accentuated in the JME group (chi2 = 5.20, d.f. = 1, P = 0.011, one-sided). CONCLUSION: Our results support previous evidence that the common KCNJ10 Arg271Cys missense variation influences seizure susceptibility of common IGE syndromes.


Asunto(s)
Alelos , Epilepsia Generalizada/genética , Predisposición Genética a la Enfermedad , Canales de Potasio de Rectificación Interna/genética , Arginina/genética , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Cisteína/genética , Análisis Mutacional de ADN/métodos , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Mutación Missense , Polimorfismo Genético
15.
Epilepsy Res ; 58(2-3): 175-83, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15120748

RESUMEN

PURPOSE: Our research program uses genetic linkage and association analysis to identify human seizure sensitivity and resistance alleles. Quantitative trait loci mapping in mice led to identification of genetic variation in the potassium ion channel gene Kcnj10, implicating it as a putative seizure susceptibility gene. The purpose of this work was to translate these animal model data to a human genetic association study. METHODS: We used single stranded conformation polymorphism (SSCP) electrophoresis, DNA sequencing and database searching (NCBI) to identify variation in the human KCNJ10 gene. Restriction fragment length polymorphism (RFLP) analysis, SSCP and Pyrosequencing were used to genotype a single nucleotide polymorphism (SNP, dbSNP rs#1130183) in KCNJ10 in epilepsy patients (n = 407) and unrelated controls (n = 284). The epilepsy group was comprised of patients with refractory mesial temporal lobe epilepsy (n = 153), childhood absence (n = 84), juvenile myoclonic (n = 111) and idiopathic generalized epilepsy not otherwise specified (IGE-NOS, n = 59) and all were of European ancestry. RESULTS: SNP rs#1130183 (C > T) alters amino acid 271 (of 379) from an arginine to a cysteine (R271C). The C allele (Arg) is common with conversion to the T allele (Cys) occurring twice as often in controls compared to epilepsy patients. Contingency analysis documented a statistically significant association between seizure resistance and allele frequency, Mantel-Haenszel chi square = 5.65, d.f. = 1, P = 0.017, odds ratio 0.52, 95% CI 0.33-0.82. CONCLUSION: The T allele of SNP rs#1130183 is associated with seizure resistance when common forms of focal and generalized epilepsy are analyzed as a group. These data suggest that this missense variation in KCNJ10 (or a nearby variation) is related to general seizure susceptibility in humans.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Convulsiones/genética , Distribución de Chi-Cuadrado , Intervalos de Confianza , Frecuencia de los Genes/genética , Genotipo , Humanos , Oportunidad Relativa , Sitios de Carácter Cuantitativo/genética
16.
Genomics ; 75(1-3): 35-42, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11472065

RESUMEN

We conducted a quantitative trait locus (QTL) mapping study to dissect the multifactorial nature of maximal electroshock seizure threshold (MEST) in C57BL/6 (B6) and DBA/2 (D2) mice. MEST determination involved a standard paradigm in which 8- to 12-week-old mice received one shock per day with a daily incremental increase in electrical current until a maximal seizure (tonic hindlimb extension) was induced. Mean MEST values in parental strains were separated by over five standard deviation units, with D2 mice showing lower values than B6 mice. The distribution of MEST values in B6xD2 F2 intercrossed mice spanned the entire phenotypic range defined by parental strains. Statistical mapping yielded significant evidence for QTLs on chromosomes 1, 2, 5, and 15, which together explained over 60% of the phenotypic variance in the model. The chromosome 1 QTL represents a locus of major effect, accounting for about one-third of the genetic variance. Experiments involving a congenic strain (B6.D2-Mtv7(a)/Ty) enabled more precise mapping of the chromosome 1 QTL and indicate that it lies in the genetic interval between markers D1Mit145 and D1Mit17. These results support the hypothesis that the distal portion of chromosome 1 harbors a gene(s) that has a fundamental role in regulating seizure susceptibility.


Asunto(s)
Electrochoque , Umbral del Dolor , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Epilepsia/genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genotipo , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Repeticiones de Microsatélite , Modelos Estadísticos , Fenotipo , Polimorfismo Genético , Carácter Cuantitativo Heredable , Factores Sexuales
17.
Epilepsia ; 42(6): 782-4, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11422336

RESUMEN

PURPOSE: We attempted to confirm recent findings of Kanemoto et al. that demonstrated a positive association (p < 0.017) between a polymorphism in the promoter region of the interleukin 1-beta (IL-1beta) gene and the clinical phenotype of temporal lobe epilepsy with hippocampal sclerosis (TLE+HS). METHODS: We determined the frequency of this polymorphism in a group of 61 TLE+HS patients of European ancestry and compared it with that found in 119 ethnically matched control subjects. RESULTS: Analysis of genotype and allele frequencies showed no statistically significant difference in the distribution of the polymorphism between the two groups (p = 0.10). CONCLUSIONS: These data suggest that this IL-1beta promoter polymorphism does not act as a strong susceptibility factor for TLE+HS in a population of individuals of European ancestry.


Asunto(s)
Epilepsia del Lóbulo Temporal/genética , Variación Genética , Interleucina-1/genética , Fragmentos de Péptidos/genética , Epilepsia del Lóbulo Temporal/diagnóstico , Epilepsia del Lóbulo Temporal/epidemiología , Etnicidad/genética , Europa (Continente)/etnología , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Genotipo , Hipocampo/patología , Humanos , Interleucina-1beta , Fenotipo , Polimorfismo Genético/genética , Regiones Promotoras Genéticas/genética , Esclerosis , Estados Unidos/epidemiología , Población Blanca/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...