Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 213: 29-37, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22521588

RESUMEN

Oligodendrocytes generate large amounts of myelin by extension of their cell membranes. Though lipid is the major component of myelin, detailed lipid metabolism in the maintenance of myelin is not understood. We reported previously that miR-32 might be involved in myelin maintenance (Shin et al., 2009). Here we demonstrate a novel role for miR-32 in oligodendrocyte function and development through the regulation of SLC45A3 (solute carrier family 45, member 3) and other downstream targets such as CLDN-11. miR-32 is highly expressed in the myelin-enriched regions of the brain and mature oligodendrocytes, and it promotes myelin protein expression. We found that miR-32 directly regulates the expression of SLC45A3 by binding to the complementary sequence on the 3'UTR of cldn11 and slc45a3. As a myelin-enriched putative sugar transporter, SLC45A3 enhances intracellular glucose levels and the synthesis of long-chain fatty acids. Therefore, overexpression of SLC45A3 triggers neutral lipid accumulation. Interestingly, both overexpression and suppression of SLC45A3 reduces myelin protein expression in mature oligodendrocytes and alters oligodendrocyte morphology, indicating that tight regulation of SLC45A3 expression is necessary for the proper maintenance of myelin proteins and structure. Taken together, our data suggest that miR-32 and its downstream target SLC45A3 play important roles in myelin maintenance by modulating glucose and lipid metabolism and myelin protein expression in oligodendrocytes.


Asunto(s)
Metabolismo de los Lípidos/genética , Proteínas de Transporte de Membrana/biosíntesis , MicroARNs/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Animales , Western Blotting , Regulación de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , MicroARNs/genética , Microscopía Electrónica de Transmisión , Proteínas de Transporte de Monosacáridos/genética , Mutagénesis Sitio-Dirigida , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Cephalalgia ; 30(2): 170-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19489890

RESUMEN

The association between the clinical use of nitroglycerin (NTG) and headache has led to the examination of NTG as a model trigger for migraine and related headache disorders, both in humans and laboratory animals. In this study in mice, we hypothesized that NTG could trigger behavioural and physiological responses that resemble a common manifestation of migraine in humans. We report that animals exhibit a dose-dependent and prolonged NTG-induced thermal and mechanical allodynia, starting 30-60 min after intraperitoneal injection of NTG at 5-10 mg/kg. NTG administration also induced Fos expression, an anatomical marker of neuronal activity in neurons of the trigeminal nucleus caudalis and cervical spinal cord dorsal horn, suggesting that enhanced nociceptive processing within the spinal cord contributes to the increased nociceptive behaviour. Moreover, sumatriptan, a drug with relative specificity for migraine, alleviated the NTG-induced allodynia. We also tested whether NTG reduces the threshold for cortical spreading depression (CSD), an event considered to be the physiological substrate of the migraine aura. We found that the threshold of CSD was unaffected by NTG, suggesting that NTG stimulates migraine mechanisms that are independent of the regulation of cortical excitability.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Nitroglicerina/toxicidad , Antagonistas del Receptor de Serotonina 5-HT1/farmacología , Sumatriptán/farmacología , Vasodilatadores/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Depresión de Propagación Cortical/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Calor , Hiperalgesia/inducido químicamente , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Estimulación Física , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
3.
Neurology ; 68(21): 1782-9, 2007 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-17515540

RESUMEN

BACKGROUND: Paroxysmal nonkinesigenic dyskinesia (PNKD) is a rare disorder characterized by episodic hyperkinetic movement attacks. We have recently identified mutations in the MR-1 gene causing familial PNKD. METHODS: We reviewed the clinical features of 14 kindreds with familial dyskinesia that was not clearly induced by movement or during sleep. Of these 14 kindreds, 8 had MR-1 mutations and 6 did not. RESULTS: Patients with PNKD with MR-1 mutations had their attack onset in youth (infancy and early childhood). Typical attacks consisted of a mixture of chorea and dystonia in the limbs, face, and trunk, and typical attack duration lasted from 10 minutes to 1 hour. Caffeine, alcohol, and emotional stress were prominent precipitants. Attacks had a favorable response to benzodiazepines, such as clonazepam and diazepam. Attacks in families without MR-1 mutations were more variable in their age at onset, precipitants, clinical features, and response to medications. Several were induced by persistent exercise. CONCLUSIONS: Paroxysmal nonkinesigenic dyskinesia (PNKD) should be strictly defined based on age at onset and ability to precipitate attacks with caffeine and alcohol. Patients with this clinical presentation (which is similar to the phenotype initially reported by Mount and Reback) are likely to harbor myofibrillogenesis regulator 1 (MR-1) gene mutations. Other "PNKD-like" families exist, but atypical features suggests that these subjects are clinically distinct from PNKD and do not have MR-1 mutations. Some may represent paroxysmal exertional dyskinesia.


Asunto(s)
Corea/genética , Corea/fisiopatología , Predisposición Genética a la Enfermedad/genética , Proteínas Musculares/genética , Mutación/genética , Adolescente , Adulto , Edad de Inicio , Cafeína/efectos adversos , Niño , Preescolar , Corea/metabolismo , Análisis Mutacional de ADN , Distonía/genética , Distonía/metabolismo , Distonía/fisiopatología , Etanol/efectos adversos , Femenino , Marcadores Genéticos/genética , Pruebas Genéticas , Genotipo , Humanos , Masculino , Linaje , Penetrancia , Estrés Psicológico/complicaciones
4.
Neurology ; 68(17): 1382-9, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17452583

RESUMEN

BACKGROUND: Familial adult myoclonic epilepsy (FAME) is associated with myoclonus, tremor, and rare seizures, and is a nonprogressive disorder linked to the FAME 1 locus. A similar disorder has been linked to the FAME 2 locus. METHODS: Seventeen patients from two families with myoclonus and epilepsy were evaluated clinically and underwent EEG, EMG, jerk-locked averaging, and MRI scanning. Three had responses to magnetic stimulation assessed. Linkage was assessed for microsatellite markers across the FAME 1 and 2 loci. RESULTS: The median age at onset was 20 years, with many patients having frequent seizures, cognitive impairment, and cerebellar dysfunction. Electrophysiologic features of cortical myoclonus were typically present, but photosensitivity was uncommon. MRI frequently demonstrated cerebellar atrophy. Pathology of a single case showed Purkinje cell loss, dentate atrophy, and neuronal loss and gliosis in the olives and pallidum. Analysis of genotypes for markers at the FAME 1 and FAME 2 loci excluded these as the region containing the same locus in one family, but only the FAME 2 locus was excluded in the other family. CONCLUSIONS: This form of familial adult myoclonic epilepsy does not show linkage to either of the known familial adult myoclonic epilepsy loci, and is characterized in some members by frequent seizures, cerebellar ataxia, dementia, and progression of the disease. This may represent a new form of progressive myoclonus and epilepsy, which we have termed familial adult myoclonic epilepsy type 3.


Asunto(s)
Epilepsias Mioclónicas/genética , Adolescente , Adulto , Edad de Inicio , Atrofia , Encéfalo/patología , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/patología , Trastornos del Conocimiento/epidemiología , Trastornos del Conocimiento/genética , Progresión de la Enfermedad , Disartria/genética , Electroencefalografía , Electromiografía , Epilepsias Mioclónicas/clasificación , Epilepsias Mioclónicas/epidemiología , Epilepsias Mioclónicas/patología , Epilepsias Mioclónicas/fisiopatología , Epilepsia Tónico-Clónica/genética , Potenciales Evocados , Femenino , Genes Dominantes , Ligamiento Genético , Gliosis/patología , Humanos , Escala de Lod , Imagen por Resonancia Magnética , Masculino , Repeticiones de Microsatélite , Nistagmo Patológico/genética , Linaje , Células de Purkinje/patología , Sudáfrica , Estimulación Magnética Transcraneal
5.
Cell ; 128(1): 59-70, 2007 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-17218255

RESUMEN

Circadian rhythms are endogenous oscillations of physiological and behavioral phenomena with period length of approximately 24 hr. A mutation in human Period 2 (hPER2), a gene crucial for resetting the central clock in response to light, is associated with familial advanced sleep phase syndrome (FASPS), an autosomal dominant condition with early morning awakening and early sleep times. The FASPS hPER2 S662G mutation resulted in PER2 being hypophosphorylated by casein kinase I (CKI) in vitro. We generated transgenic mice carrying the FASPS hPER2 S662G mutation and faithfully recapitulate the human phenotype. We show that phosphorylation at S662 leads to increased PER2 transcription and suggest that phosphorylation at another site leads to PER2 degradation. Altering CKIdelta dosage modulates the S662 phenotype demonstrating that CKIdelta can regulate period through PER2 in vivo. Modeling a naturally occurring human variant in mice has yielded novel insights into PER2 regulation.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Mutación/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Relojes Biológicos/genética , Quinasa de la Caseína I/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/genética , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Péptidos/química , Péptidos/metabolismo , Proteínas Circadianas Period , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina/genética , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Artículo en Inglés | MEDLINE | ID: mdl-18419283

RESUMEN

Biological rhythms govern the ebb and flow of life on planet Earth. Animals have an internal timekeeping mechanism that precisely regulates 24-hour rhythms of body function and behavior and synchronizes them to the day/night cycle. Circadian pacemakers trigger behavioral and physiological processes that dictate our daily rhythms. Despite the importance of the circadian clock to all aspects of our physiology and behavior, the opportunity to probe the human circadian clock only recently became possible with the recognition of Mendelian circadian variants in people (familial advanced sleep phase syndrome, FASPS). We have now cloned several genes and identified mutations causing FASPS. Study of these genes and the proteins they encode and engineering of the human mutations into mouse models are allowing study of this fascinating phenotype and yielding novel insights into circadian regulation in humans. Ultimately, such work will allow us to understand the similarities and differences between the human clock and those of model organisms. In addition, recent studies have also linked disruption of the circadian clock with numerous ailments, including cancer, cardiovascular diseases, asthma, and learning disorders. Thus, studying the molecular mechanism of human circadian rhythmicity will have an enormous impact on our understanding of human health and disease. It should also lead to new strategies for pharmacological manipulation of the human clock to improve the treatment of jet lag, various clock-related sleep and psychiatric disorders, and other human diseases.


Asunto(s)
Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Envejecimiento/fisiología , Animales , Caseína Cinasa 1 épsilon/genética , Caseína Cinasa 1 épsilon/fisiología , Clonación Molecular , Dosificación de Gen , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteínas Circadianas Period , Fenotipo , Trastornos del Sueño del Ritmo Circadiano/genética , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
7.
Neurology ; 66(11): 1703-10, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16769944

RESUMEN

BACKGROUND: The Andersen-Tawil syndrome (ATS) is a potassium ion channelopathy caused by mutations in the KCNJ2 gene. It is characterized by periodic paralysis, cardiac arrhythmias, and distinctive features; the effect of KCNJ2 mutations on the CNS has never been studied. OBJECTIVE: To define a potential CNS phenotype in ATS using standardized methods. METHODS: Ten subjects with KCNJ2 mutations and their unaffected siblings were evaluated at the University of California San Francisco General Clinical Research Center. A comprehensive battery of neurocognitive tests was administered to ATS subjects and their unaffected siblings, followed by pairwise analysis of the resultant differences in scores. An EEG was obtained for all ATS subjects. RESULTS: There was no EEG evidence of subclinical seizure activity in any subject. ATS subjects universally had more school difficulties than their siblings, despite similar IQ between the two groups. On formal neurocognitive testing, there was no difference between ATS subjects and their siblings on tests of verbal and visual memory. Assessment of executive functioning revealed ATS subjects scored 1.93 points lower than their siblings on tests of Design Fluency (95% CI -3.46, 0.01; p = 0.052) and made 1.9 more errors (95% CI 0.46, 2.54; p = 0.005). Subjects with ATS scored an average of 5 points lower than their siblings on tests of matrix reasoning (95% CI -8.67, -1.33; p = 0.008). On tests of general ability, ATS subjects achieved much lower scores than their siblings, with an average difference of 9.13 points for reading (95% CI -12.46, 3.21; p = 0.056) and 23.4 points for mathematics (95% CI -42.53, -4.22; p = 0.017). CONCLUSION: Mutations in KCNJ2 are associated with a distinct neurocognitive phenotype, characterized by deficits in executive function and abstract reasoning.


Asunto(s)
Síndrome de Andersen/diagnóstico , Síndrome de Andersen/genética , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/genética , Pruebas Neuropsicológicas , Canales de Potasio de Rectificación Interna/genética , Adolescente , Adulto , Síndrome de Andersen/clasificación , Niño , Trastornos del Conocimiento/clasificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
8.
Am J Med Genet A ; 140(4): 312-21, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16419128

RESUMEN

Andersen-Tawil syndrome (ATS) is an autosomal dominant multisystem disorder characterized by developmental, cardiac, and neuromuscular abnormalities. Approximately 70% of patients have mutations in KCNJ2, resulting in dysfunction of the inward-rectifying potassium channel Kir2.1. Variable expression complicates the diagnosis of ATS, which in many cases, is not made until years after the first recognized symptom. To better define the distinctive clinical features of ATS and facilitate earlier diagnosis, we conducted a prospective, standardized evaluation of 10 subjects with confirmed KCNJ2 mutations. Detailed anthropometric, neurological, and cardiac evaluations were performed. Using this approach, we identified novel skeletal and dental findings and proposed additional diagnostic criteria for ATS dysmorphology.


Asunto(s)
Anomalías Múltiples/genética , Síndrome de Andersen/genética , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Anomalías Múltiples/diagnóstico , Adolescente , Adulto , Síndrome de Andersen/patología , Antropometría , Arritmias Cardíacas/genética , Niño , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Enfermedades del Sistema Nervioso/genética , Linaje , Fenotipo , Estudios Prospectivos , Anomalías Dentarias/genética
9.
Brain ; 129(Pt 1): 8-17, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16195244

RESUMEN

Periodic paralyses (PPs) are rare inherited channelopathies that manifest as abnormal, often potassium (K)-sensitive, muscle membrane excitability leading to episodic flaccid paralysis. Hypokalaemic (HypoPP) and hyperkalaemic PP and Andersen-Tawil syndrome are genetically heterogeneous. Over the past decade mutations in genes encoding three ion channels, CACN1AS, SCN4A and KCNJ2, have been identified and account for at least 70% of the identified cases of PP and several allelic disorders. No prospective clinical studies have followed sufficiently large cohorts with characterized molecular lesions to draw precise conclusions. We summarize current knowledge of the clinical diagnosis, molecular genetics, genotype-phenotype correlations, pathophysiology and treatment in the PPs. We focus on unresolved issues including (i) Are there additional ion channel defects in cases without defined mutations? (ii) What is the mechanism for depolarization-induced weakness in Hypo PP? and finally (iii) Will detailed electrophysiological studies be able to correctly identify specific channel mutations? Understanding the pathophysiology of the potassium-sensitive PPs ought to reduce genetic complexity, allow subjects to be stratified during future clinical trials and increase the likelihood of observing true clinical effects. Ideally, therapy for the PPs will prevent attacks, avoid permanent weakness and improve quality of life. Moreover, understanding the skeletal muscle channelopathies will hopefully lead to insights into the more common central nervous system channel diseases such as migraine and epilepsy.


Asunto(s)
Parálisis Periódicas Familiares , Animales , Inhibidores de Anhidrasa Carbónica/uso terapéutico , Genotipo , Humanos , Activación del Canal Iónico , Ratones , Ratones Noqueados , Modelos Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Parálisis Periódicas Familiares/diagnóstico , Parálisis Periódicas Familiares/tratamiento farmacológico , Parálisis Periódicas Familiares/genética , Fenotipo , Potasio/metabolismo , Potasio/uso terapéutico , Canales de Potasio/genética , Canales de Potasio/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo
10.
Genomics ; 85(5): 582-90, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15820310

RESUMEN

The human ortholog of the gene responsible for audiogenic seizure susceptibility in Frings and BUB/BnJ mice (mouse gene symbol Mass1) recently was shown to underlie Usher syndrome type IIC (USH2C). Here we report that the Mass1frings mutation is responsible for the early onset hearing impairment of BUB/BnJ mice. We found highly significant linkage of Mass1 with ABR threshold variation among mice from two backcrosses involving BUB/BnJ mice with mice of strains CAST/EiJ and MOLD/RkJ. We also show an additive effect of the Cdh23 locus in modulating the progression of hearing loss in backcross mice. Together, these two loci account for more than 70% of the total ABR threshold variation among the backcross mice at all ages. The modifying effect of the strain-specific Cdh23ahl variant may account for the hearing and audiogenic seizure differences observed between Frings and BUB/BnJ mice, which share the Mass1frings mutation. During postnatal cochlear development in BUB/BnJ mice, stereocilia bundles develop abnormally and remain immature and splayed into adulthood, corresponding with the early onset hearing impairment associated with Mass1frings. Progressive base-apex hair cell degeneration occurs at older ages, corresponding with the age-related hearing loss associated with Cdh23ahl. The molecular basis and pathophysiology of hearing loss suggest BUB/BnJ and Frings mice as models to study cellular and molecular mechanisms underlying USH2C auditory pathology.


Asunto(s)
Cóclea/ultraestructura , Modelos Animales de Enfermedad , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Mutación/genética , Receptores Acoplados a Proteínas G/genética , Animales , Cruzamientos Genéticos , Cartilla de ADN , Electroforesis en Gel de Agar , Potenciales Evocados Auditivos del Tronco Encefálico , Ratones , Ratones Mutantes , Repeticiones de Microsatélite/genética , Microscopía Electrónica de Rastreo
11.
Neurology ; 63(12): 2280-7, 2004 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-15623687

RESUMEN

BACKGROUND: Paroxysmal kinesigenic dyskinesia (PKD) is a rare disorder characterized by short episodes of involuntary movement attacks triggered by sudden voluntary movements. Although a genetic basis is suspected in idiopathic cases, the gene has not been discovered. Establishing strict diagnostic criteria will help genetic studies. METHODS: The authors reviewed the clinical features of 121 affected individuals, who were referred for genetic study with a presumptive diagnosis of idiopathic PKD. RESULTS: The majority (79%) of affected subjects had a distinctive homogeneous phenotype. The authors propose the following diagnostic criteria for idiopathic PKD based on this phenotype: identified trigger for the attacks (sudden movements), short duration of attacks (<1 minute), lack of loss of consciousness or pain during attacks, antiepileptic drug responsiveness, exclusion of other organic diseases, and age at onset between 1 and 20 years if there is no family history (age at onset may be applied less stringently in those with family history). In comparing familial and sporadic cases, sporadic cases were more frequently male, and infantile convulsions were more common in the familial kindreds. Females had a higher remission rate than males. An infantile-onset group with a different set of characteristics was identified. A clear kinesigenic trigger was not elicited in all cases, antiepileptic response was not universal, and some infants had attacks while asleep. CONCLUSIONS: The diagnosis of idiopathic paroxysmal kinesigenic dyskinesia (PKD) can be made based on historical features. The correct diagnosis has implications for treatment and prognosis, and the diagnostic scheme may allow better focus in the search for the PKD gene(s).


Asunto(s)
Corea/diagnóstico , Adolescente , Adulto , Edad de Inicio , Anticonvulsivantes/uso terapéutico , Niño , Preescolar , Corea/tratamiento farmacológico , Corea/epidemiología , Corea/genética , Comorbilidad , Trastornos Distónicos/epidemiología , Temblor Esencial/epidemiología , Salud de la Familia , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Trastornos Migrañosos/epidemiología , Movimiento , Fenotipo , Embarazo , Complicaciones del Embarazo/epidemiología , Remisión Espontánea , Espasmos Infantiles/epidemiología
12.
Neurology ; 63(9): 1647-55, 2004 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-15534250

RESUMEN

BACKGROUND: Periodic paralyses and paramyotonia congenita are rare disorders causing disabling weakness and myotonia. Mutations in sodium, calcium, and potassium channels have been recognized as causing disease. OBJECTIVE: To analyze the clinical phenotype of patients with and without discernible genotype and to identify other mutations in ion channel genes associated with disease. METHODS: The authors have reviewed clinical data in patients with a diagnosis of hypokalemic periodic paralysis (56 kindreds, 71 patients), hyperkalemic periodic paralysis (47 kindreds, 99 patients), and paramyotonia congenita (24 kindreds, 56 patients). For those patients without one of the classically known mutations, the authors analyzed the entire coding region of the SCN4A, KCNE3, and KCNJ2 genes and portions of the coding region of the CACNA1S gene in order to identify new mutations. RESULTS: Mutations were identified in approximately two thirds of kindreds with periodic paralysis or paramyotonia congenita. The authors found differences between the disorders and between those with and without identified mutations in terms of age at onset, frequency of attacks, duration of attacks, fixed proximal weakness, precipitants of attacks, myotonia, electrophysiologic studies, serum potassium levels, muscle biopsy, response to potassium administration, and response to treatment with acetazolamide. CONCLUSIONS: Hypokalemic periodic paralysis, hyperkalemic periodic paralysis, and paramyotonia congenita may be distinguished based on clinical data. This series of 226 patients (127 kindreds) confirms some clinical features of this disorder with notable exceptions: In this series, patients without mutations had a less typical clinical presentation including an older age at onset, no changes in diet as a precipitant, and absence of vacuolar myopathy on muscle biopsy.


Asunto(s)
Parálisis Periódica Hipopotasémica/diagnóstico , Trastornos Miotónicos/diagnóstico , Parálisis Periódica Hiperpotasémica/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Parálisis Periódica Hipopotasémica/genética , Masculino , Persona de Mediana Edad , Trastornos Miotónicos/genética , Canal de Sodio Activado por Voltaje NAV1.4 , Parálisis Periódica Hiperpotasémica/genética , Fenotipo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio con Entrada de Voltaje/genética , Canales de Sodio/genética
13.
Neurology ; 60(11): 1811-6, 2003 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-12796536

RESUMEN

BACKGROUND: Mutations in KCNJ2, the gene encoding the inward-rectifying K+ channel Kir2.1, cause the cardiac, skeletal muscle, and developmental phenotypes of Andersen-Tawil syndrome (ATS; also known as Andersen syndrome). Although pathogenic mechanisms have been proposed for select mutations, a common mechanism has not been identified. METHODS: Seventeen probands presenting with symptoms characteristic of ATS were evaluated clinically and screened for mutations in KCNJ2. The results of mutation analysis were combined with those from previously studied subjects to assess the frequency with which KCNJ2 mutations cause ATS. RESULTS: Mutations in KCNJ2 were discovered in nine probands. These included six novel mutations (D71N, T75R, G146D, R189I, G300D, and R312C) as well as previously reported mutations R67W and R218W. Six probands possessed mutations of residues implicated in binding membrane-associated phosphatidylinositol 4,5-bisphosphate (PIP2). In total, mutations in PIP(2)-related residues accounted for disease in 18 of 29 (62%) reported KCNJ2 -based probands with ATS. Also reported is that mutation R67W causes the full clinical triad in two unrelated males. CONCLUSIONS: The novel mutations corresponding to residues involved in Kir2.1 channel-PIP2 interactions presented here as well as the overall frequency of mutations occurring in these residues indicate that defects in PIP2 binding constitute a major pathogenic mechanism of ATS. Furthermore, screening KCNJ2 in patients with the complex phenotypes of ATS was found to be invaluable in establishing or confirming a disease diagnosis as mutations in this gene can be identified in the majority of patients.


Asunto(s)
Anomalías Múltiples/genética , Arritmias Cardíacas/genética , Mutación , Parálisis/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Rectificación Interna/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Arritmias Cardíacas/diagnóstico , Sitios de Unión , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Debilidad Muscular/genética , Parálisis/diagnóstico , Linaje , Fenotipo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/metabolismo , Síndrome
14.
Neurology ; 58(8): 1266-72, 2002 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-11971097

RESUMEN

BACKGROUND: Mutations in the human skeletal muscle sodium channels are associated with hyperKPP, hypoKPP, paramyotonia congenita, and potassium-aggravated myotonia. This article describes the clinical manifestations of a patient with hyperKPP carrying a mutation (L689I) occurring in the linker DIIS4-S5 and its functional expression in a mammalian system. OBJECTIVE: To correlate the clinical manifestations of hyperkalemic periodic paralysis (hyperKPP) with the functional expression of a sodium channel mutation. METHODS: The mutation was introduced into a mammalian expression vector and expressed in the human embryonic kidney 293 cells. The functional expression of the L689I and that of the wild-type channels was monitored using the whole cell voltage-clamp technique. RESULTS: There was no change in the kinetics of fast inactivation, and inactivation curves were indistinguishable from that of wild-type channels. However, the L689I mutation caused a hyperpolarizing shift in the voltage dependence of activation and the mutant channels showed an impaired slow inactivation process. In addition, the mutant channels have a larger persistent current at -40 mV where window current may occur. CONCLUSIONS: The L689I mutation has similar effects to the T704M mutation and causes hyperKPP in this family. Because both of these hyperKPP mutations cause episodic muscle weakness, and because patients harboring another mutation (I693T) also can have episodic weakness, it is hypothesized that mutations occurring in this region of the sodium channel may cause episodic weakness through an impaired slow inactivation process coupled with enhanced activation.


Asunto(s)
Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Parálisis/genética , Parálisis/fisiopatología , Canales de Sodio/genética , Canales de Sodio/fisiología , Adulto , Sustitución de Aminoácidos , Línea Celular , ADN/genética , Electrofisiología , Femenino , Humanos , Hiperpotasemia/fisiopatología , Masculino , Potenciales de la Membrana/fisiología , Mutagénesis , Mutación/genética , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
15.
Hum Mol Genet ; 10(24): 2821-31, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11734547

RESUMEN

Spinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder characterized by ataxia and selective neuronal cell loss caused by the expansion of a translated CAG repeat encoding a polyglutamine tract in ataxin-7, the SCA7 gene product. To gain insight into ataxin-7 function and to decipher the molecular mechanisms of neurodegeneration in SCA7, a two-hybrid assay was performed to identify ataxin-7 interacting proteins. Herein, we show that ataxin-7 interacts with the ATPase subunit S4 of the proteasomal 19S regulatory complex. The ataxin-7/S4 association is modulated by the length of the polyglutamine tract whereby S4 shows a stronger association with the wild-type allele of ataxin-7. We demonstrate that endogenous ataxin-7 localizes to discrete nuclear foci that also contain additional components of the proteasomal complex. Immunohistochemical analyses suggest alterations either of the distribution or the levels of S4 immunoreactivity in neurons that degenerate in SCA7 brains. Immunoblot analyses demonstrate reduced levels of S4 in SCA7 cerebella without evident alterations in the levels of other proteasome subunits. These results suggest a role for S4 and ubiquitin-mediated proteasomal proteolysis in the molecular pathogenesis of SCA7.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-7 , Encéfalo/metabolismo , Células COS , Núcleo Celular/metabolismo , ADN Complementario , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Péptido Hidrolasas/metabolismo , Péptidos/metabolismo , Pruebas de Precipitina , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Transporte de Proteínas , Ataxias Espinocerebelosas/patología , Técnicas del Sistema de Dos Híbridos
16.
Neuron ; 31(6): 913-27, 2001 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-11580893

RESUMEN

Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant disorder caused by a CAG repeat expansion. To determine the mechanism of neurotoxicity, we produced transgenic mice and observed a cone-rod dystrophy. Nuclear inclusions were present, suggesting that the disease pathway involves the nucleus. When yeast two-hybrid assays indicated that cone-rod homeobox protein (CRX) interacts with ataxin-7, we performed further studies to assess this interaction. We found that ataxin-7 and CRX colocalize and coimmunoprecipitate. We observed that polyglutamine-expanded ataxin-7 can dramatically suppress CRX transactivation. In SCA7 transgenic mice, electrophoretic mobility shift assays indicated reduced CRX binding activity, while RT-PCR analysis detected reductions in CRX-regulated genes. Our results suggest that CRX transcription interference accounts for the retinal degeneration in SCA7 and thus may provide an explanation for how cell-type specificity is achieved in this polyglutamine repeat disease.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Péptidos/química , Transactivadores/antagonistas & inhibidores , Repeticiones de Trinucleótidos , Factores de Edad , Animales , Ataxina-7 , Línea Celular , Núcleo Celular/ultraestructura , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo/química , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Perfilación de la Expresión Génica , Genes Sintéticos , Proteínas de Homeodominio/fisiología , Humanos , Sustancias Macromoleculares , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Priones/genética , Regiones Promotoras Genéticas , Unión Proteica , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Transmisión Sináptica , Transactivadores/fisiología , Activación Transcripcional , Transfección , Transgenes , Técnicas del Sistema de Dos Híbridos
17.
Neuron ; 31(4): 537-44, 2001 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-11545713

RESUMEN

Frings mice are a model of generalized epilepsy and have seizures in response to loud noises. This phenotype is due to the autosomal recessive inheritance of a single gene on mouse chromosome 13. Here we report the fine genetic and physical mapping of the locus. Sequencing of the region led to identification of a novel gene; mutant mice are homozygous for a single base pair deletion that leads to premature termination of the encoded protein. Interestingly, the mRNA levels of this gene in various tissues are so low that the cDNA has eluded detection by standard library screening approaches. Study of the MASS1 protein will lead to new insights into regulation of neuronal excitability and a new pathway through which dysfunction can lead to epilepsy.


Asunto(s)
Epilepsia Refleja/genética , Proteínas de la Membrana/genética , Ratones Mutantes Neurológicos/genética , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Clonación Molecular , ADN Complementario , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular
18.
Ann Neurol ; 50(3): 417-20, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11558801

RESUMEN

A novel mutation in a family with hypokalemic periodic paralysis is described. The mutation R672S is located in the voltage sensor segment S4 of domain II in the SCN4A gene encoding the human skeletal muscle voltage-gated sodium channel. Functional expression of the R672S channels in human embryonic kidney 293 cells revealed a small but significant hyperpolarizing shift in the steady-state fast inactivation, and a dramatic enhancement in channel slow inactivation. These two defects are mainly due to a slow recovery of the mutant channels from fast and/or slow inactivation. Our data may help explain the mechanism underlying hypokalemic periodic paralysis and the patient's worsening from acetazolamide.


Asunto(s)
Acetazolamida/efectos adversos , Inhibidores de Anhidrasa Carbónica/efectos adversos , Parálisis Periódica Hipopotasémica/metabolismo , Mutación/genética , Bloqueadores de los Canales de Sodio , Canales de Sodio/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adolescente , Adulto , Línea Celular , Femenino , Humanos , Parálisis Periódica Hipopotasémica/genética , Parálisis Periódica Hipopotasémica/fisiopatología , Masculino , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Linaje , Canales de Sodio/metabolismo
19.
Gene ; 273(1): 89-96, 2001 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-11483364

RESUMEN

Several years ago, we initiated a long-term project of cloning new human ATP-binding cassette (ABC) transporters and linking them to various disease phenotypes. As one of the results of this project, we present two new members of the human ABCC subfamily, ABCC11 and ABCC12. These two new human ABC transporters were fully characterized and mapped to the human chromosome 16q12. With the addition of these two genes, the complete human ABCC subfamily has 12 identified members (ABCC1-12), nine from the multidrug resistance-like subgroup, two from the sulfonylurea receptor subgroup, and the CFTR gene. Phylogenetic analysis determined that ABCC11 and ABCC12 are derived by duplication, and are most closely related to the ABCC5 gene. Genetic variation in some ABCC subfamily members is associated with human inherited diseases, including cystic fibrosis (CFTR/ABCC7), Dubin-Johnson syndrome (ABCC2), pseudoxanthoma elasticum (ABCC6) and familial persistent hyperinsulinemic hypoglycemia of infancy (ABCC8). Since ABCC11 and ABCC12 were mapped to a region harboring gene(s) for paroxysmal kinesigenic choreoathetosis, the two genes represent positional candidates for this disorder.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Cromosomas Humanos Par 16 , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Mapeo Cromosómico , Clonación Molecular , Humanos , Datos de Secuencia Molecular , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Filogenia
20.
Neurogenetics ; 3(2): 83-90, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11354830

RESUMEN

Expansion of polymorphic CAG repeats encoding polyglutamine cause at least eight inherited neurodegenerative diseases, including Huntington disease and the spinocerebellar ataxias. However, the pathways by which proteins containing expanded polyglutamine tracts cause disease remain unclear. To gain insight into the function of the SCA7 gene product, ataxin-7, as well as its contribution to cell death in spinocerebellar ataxia type 7 (SCA7), polyclonal antibodies were generated and ataxin-7 expression was examined within neuronal tissues from controls and three SCA7 patients. Immunoblotting demonstrates that ataxin-7 is widely expressed but that expression levels vary between tissues. Immunohistochemical analyses indicate that ataxin-7 is expressed within neurons both affected and unaffected in SCA7 pathology and that subcellular localization varies depending upon the neuronal subtype. Additionally, ataxin-7 staining was detected throughout control retina, including intense staining within the cell bodies and photosensitive outer segments of cone photoreceptors. Anti-ataxin-7 antibodies revealed intranuclear inclusions within surviving inferior olivary and cortical pyramidal neurons, as well as within surviving photoreceptor and ganglion cells of SCA7 patients harboring either 42 or 66 CAG repeats at the SCA7 locus. In contrast, inclusion formation was not detected within neurons of a patient with 41 repeats. This study broadens the current understanding of ataxin-7 localization and incorporates for the first time analysis of late-onset SCA7 patients where polyglutamine tract lengths are relatively shorter and disease course less severe than in previously described infantile-onset cases.


Asunto(s)
Encéfalo/patología , Cerebelo/patología , Proteínas del Tejido Nervioso/genética , Degeneraciones Espinocerebelosas/genética , Repeticiones de Trinucleótidos , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Ataxina-7 , Femenino , Humanos , Inmunohistoquímica , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/química , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Valores de Referencia , Retina/patología , Médula Espinal/patología , Degeneraciones Espinocerebelosas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...