Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Hum Genet ; 139(4): 499-512, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31980904

RESUMEN

CHD8, which encodes Chromodomain helicase DNA-binding protein 8, is one of a few well-established Autism Spectrum Disorder (ASD) genes. Over 60 mutations have been reported in subjects with variable phenotypes, but little is known concerning genotype-phenotype correlations. We have identified four novel de novo mutations in Chinese subjects: two nonsense variants (c.3562C>T/p.Arg1188X, c.2065C>A/p.Glu689X), a splice site variant (c.4818-1G>A) and a missense variant (c.3502T>A/p.Tyr1168Asn). Three of these were identified from a 445-member ASD cohort by ASD gene panel sequencing of the 96 subjects who remained negative after molecular testing for copy number variation, Rett syndrome, FragileX and tuberous sclerosis complex (TSC). The fourth (p.Glu689X) was detected separately by diagnostic trio exome sequencing. We used diagnostic instruments and a comprehensive review of phenotypes, including prenatal and postnatal growth parameters, developmental milestones, and dysmorphic features to compare these four subjects. In addition to autism, they also presented with prenatal onset macrocephaly, intellectual disability, overgrowth during puberty, sleep disorder, and dysmorphic features, including broad forehead with prominent supraorbital ridges, flat nasal bridge, telecanthus and large ears. For further comparison, we compiled a comprehensive list of CHD8 variants from the literature and databases, which revealed constitutive and somatic truncating variants in the HELIC (Helicase-C) domain in ASD and in cancer patients, respectively, but not in the general population. Furthermore, HELIC domain mutations were associated with a severe phenotype defined by a greater number of clinical features, lower verbal IQ, and a prominent, consistent pattern of overgrowth as measured by weight, height and head circumference. Overall, this study adds to the ASD-associated loss-of-function mutations in CHD8 and highlights the clinical importance of the HELIC domain of CHD8.


Asunto(s)
Trastorno del Espectro Autista/genética , Codón sin Sentido , Proteínas de Unión al ADN/genética , Síndrome del Cromosoma X Frágil/genética , Trastornos del Desarrollo del Lenguaje/genética , Mutación Missense , Fenotipo , Síndrome de Rett/genética , Factores de Transcripción/genética , Esclerosis Tuberosa/genética , Trastorno del Espectro Autista/enzimología , Niño , Femenino , Síndrome del Cromosoma X Frágil/enzimología , Humanos , Trastornos del Desarrollo del Lenguaje/enzimología , Masculino , Dominios Proteicos , Síndrome de Rett/enzimología , Esclerosis Tuberosa/enzimología
2.
Mol Neurobiol ; 56(12): 8277-8295, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31214863

RESUMEN

MeCP2 is an X-linked gene; its mutation causes Rett Syndrome (RTT), a severe neurodevelopmental disability that affects mainly girls. Acting as a transcription factor, the MeCP2 protein is able to regulate several hormone-related genes, such as the thyroid hormones (TH), which are known to play an important role in the development of the central nervous system (CNS). Although only a few studies have associated RTT and TH, TH deficit can lead to neurological deregulation by triggering functional deficiencies during adulthood. Here, we used human-induced pluripotent stem cell (iPSC) to generate MeCP2-knockout neuronal progenitor cells and adult neurons. Using this cellular model, we then investigated the expression of genes associated with TH homeostasis, such as the TH transporters (LAT1, LAT2, MCT8, MCT10, and OATP4A1) and deiodinases (DIO1, 2, and 3). Then, we treated the neural cells with THs and analyzed the expression of several genes related to neurodevelopment and functional maintenance. Our results showed that several TH-related genes, such as deiodinases, are altered in RTT samples when compared to WT cells. Moreover, the treatment of the neural cells with THs increased the amount of MAP2 and synapsin-1 expression in RTT cells. Our work provided evidences that TH homeostasis is compromised in RTT-derived neural cells, which could be an important factor to contribute to the imbalance in the neurodevelopmental phenotype presented in this syndrome and can lead us to better understand other neurodevelopmental diseases.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Yoduro Peroxidasa/genética , Proteínas de Transporte de Membrana/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Neuronas/metabolismo , Hormonas Tiroideas/metabolismo , Humanos , Yoduro Peroxidasa/metabolismo , Cariotipificación , Masculino , Proteínas de Transporte de Membrana/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Síndrome de Rett/enzimología , Síndrome de Rett/genética
3.
J Clin Invest ; 125(8): 3163-77, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26214522

RESUMEN

The X-linked neurological disorder Rett syndrome (RTT) presents with autistic features and is caused primarily by mutations in a transcriptional regulator, methyl CpG-binding protein 2 (MECP2). Current treatment options for RTT are limited to alleviating some neurological symptoms; hence, more effective therapeutic strategies are needed. We identified the protein tyrosine phosphatase PTP1B as a therapeutic candidate for treatment of RTT. We demonstrated that the PTPN1 gene, which encodes PTP1B, was a target of MECP2 and that disruption of MECP2 function was associated with increased levels of PTP1B in RTT models. Pharmacological inhibition of PTP1B ameliorated the effects of MECP2 disruption in mouse models of RTT, including improved survival in young male (Mecp2-/y) mice and improved behavior in female heterozygous (Mecp2-/+) mice. We demonstrated that PTP1B was a negative regulator of tyrosine phosphorylation of the tyrosine kinase TRKB, the receptor for brain-derived neurotrophic factor (BDNF). Therefore, the elevated PTP1B that accompanies disruption of MECP2 function in RTT represents a barrier to BDNF signaling. Inhibition of PTP1B led to increased tyrosine phosphorylation of TRKB in the brain, which would augment BDNF signaling. This study presents PTP1B as a mechanism-based therapeutic target for RTT, validating a unique strategy for treating the disease by modifying signal transduction pathways with small-molecule drugs.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Síndrome de Rett/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Ratones Endogámicos CBA , Ratones Mutantes , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Receptor trkB/genética , Receptor trkB/metabolismo , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Síndrome de Rett/patología , Transducción de Señal/genética
4.
Eur J Hum Genet ; 23(9): 1171-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25424712

RESUMEN

Rett syndrome (RTT), a neurodevelopmental disorder that predominantly affects females, is primarily caused by variants in MECP2. Variants in other genes such as CDKL5 and FOXG1 are usually associated with individuals who manifest distinct phenotypes that may overlap with RTT. Individuals with phenotypes suggestive of RTT are typically screened for variants in MECP2 and then subsequently the other genes dependent on the specific phenotype. Even with this screening strategy, there are individuals in whom no causative variant can be identified, suggesting that there are other novel genes that contribute to the RTT phenotype. Here we report a de novo deletion of protein tyrosine phosphatase, non-receptor type 4 (PTPN4) in identical twins with a RTT-like phenotype. We also demonstrate the reduced expression of Ptpn4 in a Mecp2 null mouse model of RTT, as well as the activation of the PTPN4 promoter by MeCP2. Our findings suggest that PTPN4 should be considered for addition to the growing list of genes that warrant screening in individuals with a RTT-like phenotype.


Asunto(s)
Eliminación de Gen , Proteína 2 de Unión a Metil-CpG/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 4/genética , Síndrome de Rett/genética , Adolescente , Animales , Cerebelo/enzimología , Cerebelo/patología , Cromosomas Humanos Par 2/química , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Genotipo , Hipocampo/enzimología , Hipocampo/patología , Humanos , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Ratones Transgénicos , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 4/deficiencia , Síndrome de Rett/enzimología , Síndrome de Rett/patología , Gemelos Monocigóticos
5.
Neuropharmacology ; 80: 70-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24495398

RESUMEN

Epigenetic regulation has been long considered to be a critical mechanism in the control of key aspects of cellular functions such as cell division, growth, and cell fate determination. Exciting recent developments have demonstrated that epigenetic mechanisms can also play necessary roles in the nervous system by regulating, for example, neuronal gene expression, DNA damage, and genome stability. Despite the fact that postmitotic neurons are developmentally less active then dividing cells, epigenetic regulation appears to provide means of both long-lasting and very dynamic regulation of neuronal function. Growing evidence indicates that epigenetic mechanisms in the central nervous system (CNS) are important for regulating not only specific aspects of individual neuronal metabolism but also for maintaining function of neuronal circuits and regulating their behavioral outputs. Multiple reports demonstrated that higher-level cognitive behaviors, such as learning and memory, are subject to a sophisticated epigenetic control, which includes interplay between multiple mechanisms of neuronal chromatin modification. Experiments with animal models have demonstrated that various epigenetic manipulations can affect cognition in different ways, from severe dysfunction to substantial improvement. In humans, epigenetic dysregulation has been known to underlie a number of disorders that are accompanied by mental impairment. Here, we review some of the epigenetic mechanisms that regulate cognition and how their disruption may contribute to cognitive dysfunctions. Due to the fact that histone acetylation and DNA methylation are some of the best-studied and critically important epigenomic modifications our research team has particularly strong expertise in, in this review, we are going to concentrate on histone acetylation, as well as DNA methylation/hydroxymethylation, in the mammalian CNS. Additional epigenetic modifications, not surveyed here, are being discussed in depth in the other review articles in this issue of Neuropharmacology.


Asunto(s)
Sistema Nervioso Central/metabolismo , Trastornos del Conocimiento/metabolismo , Cognición , Epigénesis Genética , Modelos Biológicos , Neuronas/metabolismo , 5-Metilcitosina/análogos & derivados , Acetilación , Animales , Sistema Nervioso Central/enzimología , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/etiología , Citosina/análogos & derivados , Citosina/metabolismo , Metilación de ADN , Regulación de la Expresión Génica , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Aprendizaje , Memoria , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología , Procesamiento Proteico-Postraduccional , Síndrome de Rett/enzimología , Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatología , Síndrome de Rubinstein-Taybi/enzimología , Síndrome de Rubinstein-Taybi/metabolismo , Síndrome de Rubinstein-Taybi/fisiopatología
6.
Neuromolecular Med ; 15(1): 218-25, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23242510

RESUMEN

Rett syndrome is a severe neurodevelopmental disorder, almost exclusively affecting females and characterized by a wide spectrum of clinical manifestations. Both the classic and atypical forms of Rett syndrome are primarily due to mutations in the methyl-CpG-binding protein 2 (MECP2) gene. Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have been identified in patients with atypical Rett syndrome, X-linked infantile spasms sharing common features of generally early-onset seizures and mental retardation. CDKL5 is known as serine/threonine protein kinase 9 (STK9) and is mapped to the Xp22 region. It has a conserved serine/threonine kinase domain within its amino terminus and a large C-terminal region. Disease-causing mutations are distributed in both the amino terminal domain and in the large C-terminal domain. We have screened the CDKL5 gene in 44 patients with atypical Rett syndrome who had tested negative for MECP2 gene mutations and have identified 6 sequence variants, out of which three were novel and three known mutations. Two of these novel mutations p.V966I and p.A1011V were missense and p.H589H a silent mutation. Other known mutations identified were p.V999M, p.Q791P and p.T734A. Sequence homology for all the mutations revealed that the two mutations (p.Q791P and p.T734A) were conserved across species. This indicated the importance of these residues in structure and function of the protein. The damaging effects of these mutations were analysed in silico using PolyPhen-2 online software. The PolyPhen-2 scores of p.Q791P and p.T734A were 0.998 and 0.48, revealing that these mutations could be deleterious and might have potential functional effect. All other mutations had a low score suggesting that they might not alter the activity of CDKL5. We have also analysed the position of the mutations in the CDKL5 protein and found that all the mutations were present in the C-terminal domain of the protein. The C-terminal domain is required for cellular localization through protein-protein interaction; any mutations in this domain might alter this function of the protein. This is the first report from India showing the mutation in CDKL5 gene in Indian cases of Rett syndrome. Our study emphasizes the role of CDKL5 mutation screening in cases of atypical Rett syndrome with congenital seizure variant.


Asunto(s)
Cromosomas Humanos X/genética , Mutación Missense , Mutación Puntual , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Rett/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia Conservada , Femenino , Humanos , India/epidemiología , Recién Nacido , Proteína 2 de Unión a Metil-CpG/genética , Modelos Moleculares , Datos de Secuencia Molecular , Fenotipo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Estructura Terciaria de Proteína , Síndrome de Rett/enzimología , Síndrome de Rett/etnología , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Vertebrados/genética , Inactivación del Cromosoma X
7.
Neuropsychopharmacology ; 37(5): 1152-63, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22157810

RESUMEN

RhoGTPases are crucial molecules in neuronal plasticity and cognition, as confirmed by their role in non-syndromic mental retardation. Activation of brain RhoGTPases by the bacterial cytotoxic necrotizing factor 1 (CNF1) reshapes the actin cytoskeleton and enhances neurotransmission and synaptic plasticity in mouse brains. We evaluated the effects of a single CNF1 intracerebroventricular inoculation in a mouse model of Rett syndrome (RTT), a rare neurodevelopmental disorder and a genetic cause of mental retardation, for which no effective therapy is available. Fully symptomatic MeCP2-308 male mice were evaluated in a battery of tests specifically tailored to detect RTT-related impairments. At the end of behavioral testing, brain sections were immunohistochemically characterized. Magnetic resonance imaging and spectroscopy (MRS) were also applied to assess morphological and metabolic brain changes. The CNF1 administration markedly improved the behavioral phenotype of MeCP2-308 mice. CNF1 also dramatically reversed the evident signs of atrophy in astrocytes of mutant mice and restored wt-like levels of this cell population. A partial rescue of the overexpression of IL-6 cytokine was also observed in RTT brains. CNF1-induced brain metabolic changes detected by MRS analysis involved markers of glial integrity and bioenergetics, and point to improved mitochondria functionality in CNF1-treated mice. These results clearly indicate that modulation of brain RhoGTPases by CNF1 may constitute a totally innovative therapeutic approach for RTT and, possibly, for other disorders associated with mental retardation.


Asunto(s)
Astrocitos/fisiología , Síndrome de Rett/enzimología , Síndrome de Rett/patología , Proteínas de Unión al GTP rho/metabolismo , Análisis de Varianza , Animales , Astrocitos/efectos de los fármacos , Toxinas Bacterianas/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Condicionamiento Psicológico/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Proteínas de Escherichia coli/uso terapéutico , Conducta Exploratoria/efectos de los fármacos , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Inyecciones Intraventriculares , Interleucina-6/metabolismo , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Destreza Motora/efectos de los fármacos , Destreza Motora/fisiología , Fenotipo , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética
8.
Biosci Rep ; 31(5): 333-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21070191

RESUMEN

Mutations in MECP2 (methyl-CpG-binding protein 2) are linked to the severe postnatal neurodevelopmental disorder RTT (Rett syndrome). MeCP2 was originally characterized as a transcriptional repressor that preferentially bound methylated DNA; however, recent results indicate MeCP2 is a multifunctional protein. MeCP2 binding is now associated with certain expressed genes and involved in nuclear organization as well, indicating that its gene regulatory function is context-dependent. In addition, MeCP2 is proposed to regulate mRNA splicing and a mouse model for RTT shows aberrant mRNA splicing. To further understand MeCP2 and potential roles in RTT pathogenesis, we have employed a biochemical approach to identify the MeCP2 protein complexes present in the mammalian brain. We show that MeCP2 exists in at least four biochemically distinct pools in the brain and characterize one novel brain-derived MeCP2 complex that contains the splicing factor Prpf3 (pre-mRNA processing factor 3). MeCP2 directly interacts with Prpf3 in vitro and in vivo and many MECP2 RTT truncations disrupt the MeCP2-Prpf3 complex. In addition, MeCP2 and Prpf3 associate in vivo with mRNAs from genes known to be expressed when their promoters are associated with MeCP2. These results support a role for MeCP2 in mRNA biogenesis and suggest an additional mechanism for RTT pathophysiology.


Asunto(s)
Encéfalo/enzimología , Proteína 2 de Unión a Metil-CpG/genética , Procesamiento Proteico-Postraduccional/genética , ARN Mensajero/biosíntesis , Animales , Línea Celular , Regulación Enzimológica de la Expresión Génica/fisiología , Ratones , Regiones Promotoras Genéticas/fisiología , Multimerización de Proteína/genética , Empalme del ARN/genética , Factores de Empalme de ARN , Ratas , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Síndrome de Rett/fisiopatología , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo
9.
Twin Res Hum Genet ; 13(2): 168-78, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20397747

RESUMEN

Rett syndrome (RTT) is a severe neurodevelopmental disorder affecting females almost exclusively and is characterized by a wide spectrum of clinical manifestations. Mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene have been found in up to 95% of classical RTT cases and a lesser proportion of atypical cases. Recently, mutations in another X-linked gene, CDKL5 (cyclin-dependent kinase-like 5) have been found to cause atypical RTT, in particular the early onset seizure (Hanefeld variant) and one female with autism. In this study we screened several cohorts of children for CDKL5 mutations, totaling 316 patients, including individuals with a clinical diagnosis of RTT but who were negative for MECP2 mutations (n=102), males with X-linked mental retardation (n=9), patients with West syndrome (n=52), patients with autism (n=59), patients with epileptic encephalopathy (n=33), patients with Aicardi syndrome (n=7) and other patients with intellectual disability with or without seizures (n=54). In all, seven polymorphic variations and four de novo mutations (c.586C>T [p.S196L]; c.58G>C [p.G20R]; c.2504delC [p.P835fs]; deletion of exons 1-3) were identified, and in all instances of the latter the clinical phenotype was that of an epileptic encephalopathy. These results suggest that pathogenic CDKL5 mutations are unlikely to be identified in the absence of severe early-onset seizures and highlight the importance of screening for large intragenic and whole gene deletions.


Asunto(s)
Trastornos del Conocimiento/genética , Quinasa 5 Dependiente de la Ciclina/genética , Mutación , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Convulsiones/genética , Secuencia de Aminoácidos , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/enzimología , Estudios de Cohortes , Quinasa 5 Dependiente de la Ciclina/metabolismo , Femenino , Pruebas Genéticas , Humanos , Masculino , Datos de Secuencia Molecular , Síndrome de Rett/enzimología , Convulsiones/diagnóstico , Convulsiones/enzimología
10.
Biochem Biophys Res Commun ; 394(2): 285-90, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20193660

RESUMEN

Rett syndrome is a neurodevelopmental disorder caused by Mecp2 gene mutations. In RTT patients and Mecp2-null (Mecp2(-/Y)) mice, norepinephrine (NE) content drops significantly, which may play a role in breathing arrhythmia, sleep disorders and sudden death. However, the underlying mechanisms for the NE defect are not fully understood. The NE defect may result from decreased NE biosynthesis, loss of catecholaminergic neurons or both. Although deficiency in tyrosine hydroxylase (TH) has been demonstrated, it is possible that dopamine beta-hydroxylase (DBH), the critical enzyme converting dopamine to NE, is also affected. To test these possibilities, we studied DBH expressions in pontine catecholaminergic neurons of Mecp2(-/Y) mice identified with breathing abnormalities. In comparison to the wild type, Mecp2(-/Y) mice at 2months of age showed approximately 50% decrease in the expressions of DBH and TH, at both protein and mRNA levels in the locus coeruleus (LC) region. Consistently, DBH and TH immunoreactivity was markedly decreased in LC neurons of Mecp2(-/Y) mice. No evidence was found for selective deficiency in TH- or DBH-containing neurons in Mecp2(-/Y) mice, as almost all TH-positive cells expressed DBH. By counting TH-immunoreactive cells in the LC, we found that the Mecp2(-/Y) mice lost only approximately 5% of the catecholaminergic neurons as compared to wild-type, although their LC volume shrank by approximately 15%. These results strongly suggest that the NE defect in Mecp2(-/Y) mice is likely to result from deficient expression of not only TH but also DBH without significant loss of catecholaminergic neurons in the LC.


Asunto(s)
Dopamina beta-Hidroxilasa/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Neuronas/enzimología , Norepinefrina/deficiencia , Puente/enzimología , Síndrome de Rett/enzimología , Animales , Catecolaminas/metabolismo , Dopamina beta-Hidroxilasa/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Norepinefrina/biosíntesis , Anomalías del Sistema Respiratorio/enzimología , Anomalías del Sistema Respiratorio/genética , Síndrome de Rett/genética , Transcripción Genética , Tirosina 3-Monooxigenasa/deficiencia
11.
Hum Mol Genet ; 18(23): 4590-602, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19740913

RESUMEN

Mutations in the human X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have been shown to cause severe neurodevelopmental disorders including infantile spasms, encephalopathy, West-syndrome and an early-onset variant of Rett syndrome. CDKL5 is a serine/threonine kinase whose involvement in Rett syndrome can be inferred by its ability to directly bind and mediate phosphorylation of MeCP2. However, it remains to be elucidated how CDKL5 exerts its function. Here, we report that CDKL5 localizes to specific nuclear foci referred to as nuclear speckles in both cell lines and tissues. These sub-nuclear structures are traditionally considered as storage/modification sites of pre-mRNA splicing factors. Interestingly, we provide evidence that CDKL5 regulates the dynamic behaviour of nuclear speckles. Indeed, CDKL5 overexpression leads to nuclear speckle disassembly, and this event is strictly dependent on its kinase activity. Conversely, its down-regulation affects nuclear speckle morphology leading to abnormally large and uneven speckles. Similar results were obtained for primary adult fibroblasts isolated from CDKL5-mutated patients. Altogether, these findings indicate that CDKL5 controls nuclear speckle morphology probably by regulating the phosphorylation state of splicing regulatory proteins. Nuclear speckles are dynamic sites that can continuously supply splicing factors to active transcription sites, where splicing occurs. Notably, we proved that CDKL5 influences alternative splicing, at least as proved in heterologous minigene assays. In conclusion, we provide evidence that CDKL5 is involved indirectly in pre-mRNA processing, by controlling splicing factor dynamics. These findings identify a biological process whose disregulation might affect neuronal maturation and activity in CDKL5-related disorders.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Empalme del ARN , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Adolescente , Animales , Línea Celular , Núcleo Celular/química , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/química , Fibroblastos/metabolismo , Humanos , Lactante , Ratones , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Rett/metabolismo
12.
Am J Med Genet A ; 149A(4): 722-5, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19253388

RESUMEN

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have recently been reported in patients with severe neurodevelopmental disorder characterized by early-onset seizures, infantile spasms, severe psychomotor impairment and very recently, in patients with Rett syndrome (RTT)-like phenotype. Although the involvement of CDKL5 in specific biological pathways and its neurodevelopmental role have not been completely elucidated, the CDKL5 appears to be physiologically related to the MECP2 gene. Here we report on the clinical and CDKL5 molecular investigation in a very unusual RTT case, with severe, early-neurological involvement in which we have shown in a previous report, a novel P388S MECP2 mutation [Conforti et al. (2003); Am J Med Genet A 117A: 184-187]. The patient has had severe psychomotor delay since the first month of life and infantile spasms since age 5 months. Moreover, at age 5 years the patient suddenly presented with renal failure. The severe pattern of symptoms in our patient, similar to a CDKL5 phenotype, prompted us to perform an analysis of the CDKL5, which revealed a novel missense mutation never previously described. The X-inactivation assay was non-informative. In conclusion, this report reinforces the observation that the CDKL5 phenotype overlaps with RTT and that CDKL5 analysis is recommended in patients with a seizure disorder commencing during the first months of life.


Asunto(s)
Mutación Missense , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Adolescente , Edad de Inicio , Cromosomas Humanos X/genética , Análisis Mutacional de ADN , Epilepsia/enzimología , Epilepsia/genética , Femenino , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Fenotipo
13.
Biochem Biophys Res Commun ; 377(4): 1162-7, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18977197

RESUMEN

DNA methyltransferase 1 (Dnmt1) is an enzyme that recognizes and methylates hemimethylated CpG after DNA replication to maintain methylation patterns. Although the N-terminal region of Dnmt1 is known to interact with various proteins, such as methyl-CpG-binding protein 2 (MeCP2), the associations of protein kinases with this region have not been reported. In the present study, we found that a 110-kDa protein kinase in mouse brain could bind to the N-terminal domain of Dnmt1. This 110-kDa kinase was identified as cyclin-dependent kinase-like 5 (CDKL5) by LC-MS/MS analysis. CDKL5 and Dnmt1 were found to colocalize in nuclei and appeared to interact with each other. Catalytically active CDKL5, CDKL5(1-352), phosphorylated the N-terminal region of Dnmt1 in the presence of DNA. Considering that defects in the MeCP2 or CDKL5 genes cause Rett syndrome, we propose that the interaction between Dnmt1 and CDKL5 may contribute to the pathogenic processes of Rett syndrome.


Asunto(s)
Encéfalo/enzimología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Núcleo Celular/enzimología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Análisis Mutacional de ADN , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Síndrome de Rett/enzimología , Eliminación de Secuencia
14.
Auton Neurosci ; 136(1-2): 82-9, 2007 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17544925

RESUMEN

Methyl-CpG-binding protein 2 is a transcription factor that is involved in gene silencing. It is mutated in the majority of cases of Rett syndrome. This X-linked neurodevelopmental disorder is reported to involve abnormalities in autonomic cardiovascular regulation. As an initial step in understanding the basis for these abnormalities we have characterized autonomic cardiovascular function in Mecp2 deficient mice. Arterial pressure waves were recorded in freely moving animals using telemetry. Baseline blood pressure and pulse interval (PI) as well as indices of heart rate variability (HRV): standard deviation of PI (SDNN), range encompassing 90% of PIs (PI90) and standard deviation of adjacent PIs (SDSD) were similar in Mecp2(+/+) and Mecp2(+/-) animals. Spectral analysis of mean arterial pressure (MAP) and PI in the frequency domain showed similar relative power in low frequency 1 (LF1, 08-0.4 Hz), low frequency 2 (LF2, 0.4-1.0 Hz), middle frequency (MF, 1-3 Hz) and high frequency (HF, 3.0-10.0 Hz) bands. Autonomic blockade with atropine or propranolol as well as elevation in ambient temperature to 32 degrees C resulted in changes in blood pressure, PI and HRV that did not differ between the strains. Atropine, propranolol and elevated temperature resulted in similar changes in both MAP and PI spectral power. Baroreceptor function was tested using intravenous injections of nitroprusside followed by phenylephrine. Maximum gain was not different. These results do reveal any disturbance of autonomic cardiovascular regulation in the Mecp2 deficient mouse genotype.


Asunto(s)
Arritmias Cardíacas/genética , Enfermedades del Sistema Nervioso Autónomo/genética , Enfermedades Cardiovasculares/genética , Predisposición Genética a la Enfermedad/genética , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Antagonistas Adrenérgicos beta/farmacología , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Sistema Nervioso Autónomo/fisiopatología , Enfermedades del Sistema Nervioso Autónomo/enzimología , Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Barorreflejo/efectos de los fármacos , Barorreflejo/fisiología , Presión Sanguínea/genética , Temperatura Corporal/fisiología , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/fisiopatología , Antagonistas Colinérgicos/farmacología , Femenino , Corazón/inervación , Corazón/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/genética , Hipertermia Inducida , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Rett/complicaciones , Síndrome de Rett/enzimología
15.
Clin Genet ; 70(1): 29-33, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16813600

RESUMEN

The CDKL5 gene has been implicated in infantile spasms and more recently in a Rett syndrome-like phenotype. We report a case of a young girl presenting generalized convulsions at 10 days of life. Subsequent mutation analysis by denaturing high-performance liquid chromatography of MECP2 and CDKL5 genes revealed heterozygosity for a c.47_48insAGG insertion in exon 1 of MECP2 and heterozygosity for a new nonsense mutation p.Q834X and a new missense variant p.V999M in the CDKL5 gene. Co-segregation analysis showed that the nonsense mutation was a de novo mutation and that the insertion and the missense variant were also found in the asymptomatic mother. In the absence of skewed X inactivation in the mother, it is likely that these last two variants are not pathogenic. Reverse transcription-polymerase chain reaction from lymphoblastoid cells of the patient showed only the transcript without the nonsense and missense variations suggesting decreased stability of mature mRNA by nonsense-mediated decay. These data also suggest an occurrence of the de novo mutation in maternal germ line cells. Moreover, this report reinforces the observation that the CDKL5 phenotype overlaps with Rett syndrome and that CDKL5 gene analysis is recommended in females with a seizure disorder commencing in the first weeks of life.


Asunto(s)
Mutación de Línea Germinal , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Secuencia de Bases , Preescolar , Codón sin Sentido , ADN Complementario/genética , Exones , Femenino , Humanos , Masculino , Mutación Missense , Linaje , Fenotipo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Convulsiones/genética , Inactivación del Cromosoma X
16.
Hum Mol Genet ; 14(24): 3775-86, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16330482

RESUMEN

X-linked cyclin-dependent kinase-like 5 (CDKL5 or STK9) has recently been implicated in atypical Rett and X-linked West syndromes, severe neurological disorders associated with mental retardation, loss of communication and motor skills and infantile spasms and seizures in predominantly females. Besides CDKL5, these disease phenotypes are also linked to mutations in the MECP2 and ARX genes. Here, we have expressed and characterized CDKL5 and its mutant forms. CDKL5 is a 118 kDa protein that is widely distributed in all tissues, with highest levels in brain, thymus and testes. Whole mount embryo staining reveals CDKL5 to be ubiquitous. Within cells, CDKL5 is localized primarily in the nucleus. Removal of the C-terminal domain increases CDKL5 expression, enhances autophosphorylation activity and causes perinuclear localization, indicating that the C-terminus regulates CDKL5 function. Although we detect MeCP2 but not ARX binding to CDKL5, our results suggest that neither of these proteins are direct substrates of the CDKL5 kinase. Finally, the CDKL5 mutations associated with the disease phenotype cause loss of kinase activity as assessed by autophosphorylation. These results suggest that inactivation of the CDKL5 kinase can lead to severe neurodevelopmental disorders.


Asunto(s)
Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Síndrome de Rett/enzimología , Espasmos Infantiles/enzimología , Secuencias de Aminoácidos , Animales , Encéfalo/enzimología , Núcleo Celular/metabolismo , Células Cultivadas , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Masculino , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Síndrome de Rett/genética , Espasmos Infantiles/genética , Testículo/enzimología , Factores de Transcripción/metabolismo
17.
Neuropediatrics ; 33(2): 105-8, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12075494

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder that almost exclusively affects girls. Recently mutations in MECP2, that encodes the methyl CpG binding protein 2 (MeCP2), have been found to cause RTT. MeCP2 has a role in gene silencing. It binds to methylated cytosine in the DNA and recruits histone deacetylases. We studied the methylation pattern of the promoters of two X chromosomal genes, G6 PD and SYBL1, in patients with RTT and in a control group. Both genes undergo X inactivation which correlates with promoter methylation. A 1 : 1 ratio of methylated versus non-methylated alleles was expected. In the control group a median ratio of 47 : 53 of methylated to non-methylated alleles was found at the G6 PD promoter locus. In 22 patients with RTT the median ratio was significantly different, 33 : 67 (p < 0.0001). Analysis of the SYBL1 promoter revealed an almost identical median ratio of methylated versus non-methylated alleles (RTT 47 : 53; control 49 : 51), however, the range was wider in the RTT group (RTT 23 : 77 to 56 : 44; control 43 : 57 to 55 : 45). There was no apparent correlation between G6 PD promoter methylation status and mutations in the MeCP2 gene or the severity of the clinical phenotype in our patient group. The finding of reduced methylation at the G6 PD promoter is an interesting finding and suggests that there could be widespread dysregulation of X chromosomal genes in Rett syndrome.


Asunto(s)
Proteínas Cromosómicas no Histona , Glucosafosfato Deshidrogenasa/genética , Metiltransferasas/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Represoras , Síndrome de Rett/enzimología , Síndrome de Rett/genética , Fragmentación del ADN/genética , Metilación de ADN , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Compensación de Dosificación (Genética) , Exones , Humanos , Proteínas de la Membrana/genética , Proteína 2 de Unión a Metil-CpG , Proteínas R-SNARE , Síndrome de Rett/diagnóstico , Índice de Severidad de la Enfermedad , Cromosoma X/genética
18.
Brain Dev ; 23 Suppl 1: S58-61, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11738843

RESUMEN

The current status of neurobiological and neurochemical research on Rett syndrome is reviewed, and correlations are developed with previously described neurophysiological, neuroimaging, neuropathological, and immunohistochemical changes. We review the abnormalities reported in the biogenic amine neurotransmitters/receptor systems, and of beta-phenylethylamine, an endogenous amine synthesized by the decarboxylation of phenylalanine in dopaminergic neurons of the nigrostriatal system. We also discuss the roles of other neurotransmitters, including beta-endorphin and substance P, and neurotrophic factors, including nerve growth factors. Recently, DNA mutations in the methyl-CpG binding protein 2, mapped to Xq28, have been identified in some patients with Rett syndrome. The multiple abnormalities in the various neurotransmitters/receptor systems explain the pervasive effects of Rett syndrome.


Asunto(s)
Química Encefálica/genética , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/fisiopatología , Neurotransmisores/deficiencia , Síndrome de Rett/enzimología , Síndrome de Rett/fisiopatología , Adolescente , Adulto , Sistema Nervioso Central/crecimiento & desarrollo , Niño , Preescolar , Enzimas/deficiencia , Enzimas/genética , Femenino , Humanos , Neuronas/enzimología , Neuronas/patología , Neurotransmisores/biosíntesis , Neurotransmisores/genética
19.
Brain Dev ; 23 Suppl 1: S122-6, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11738857

RESUMEN

We immunohistochemically examined neurotransmitter systems, which function in the brainstem and are involved in neuronal organization of respiration, in an autopsy brain from a patient with Rett syndrome (RS). Immunoreactivity (IR) for tyrosine hydroxylase, a functional marker for catecholaminergic neurons, was severely reduced in the locus ceruleus, while that for tryptophan hydroxylase involved in serotonin synthesis was spared in the raphe nuclei. In the brainstem, IR for substance P (SP) was reduced in the parabrachial complex and that for methionine-enkephalin (met-enk) was affected in the parabrachial, hypoglossal, dorsal vagal and solitary nuclei. In addition, expressions of these neuropeptides were also disturbed in the basal ganglia. A widespread altered expression of antagonistic neuropeptides, SP and met-enk, may be involved in the pathogenesis of RS, especially in its respiratory manifestation.


Asunto(s)
Ganglios Basales/metabolismo , Tronco Encefálico/enzimología , Regulación hacia Abajo/genética , Neuropéptidos/deficiencia , Neurotransmisores/deficiencia , Trastornos Respiratorios/enzimología , Síndrome de Rett/enzimología , Adolescente , Ganglios Basales/patología , Ganglios Basales/fisiopatología , Tronco Encefálico/patología , Tronco Encefálico/fisiopatología , Catecolaminas/biosíntesis , Encefalina Metionina/deficiencia , Encefalina Metionina/genética , Enzimas/deficiencia , Enzimas/genética , Femenino , Humanos , Inmunohistoquímica , Locus Coeruleus/enzimología , Locus Coeruleus/patología , Locus Coeruleus/fisiopatología , Neuropéptidos/genética , Neurotransmisores/genética , Núcleos del Rafe/enzimología , Núcleos del Rafe/patología , Núcleos del Rafe/fisiopatología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/patología , Síndrome de Rett/patología , Síndrome de Rett/fisiopatología , Serotonina/biosíntesis , Sustancia P/deficiencia , Sustancia P/genética , Triptófano Hidroxilasa/efectos de los fármacos , Triptófano Hidroxilasa/genética , Tirosina 3-Monooxigenasa/efectos de los fármacos , Tirosina 3-Monooxigenasa/genética
20.
Hum Mol Genet ; 10(10): 1085-92, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11331619

RESUMEN

Rett syndrome (RTT) is a mostly sporadic disorder of developmental regression, with loss of speech and purposeful hand use, microcephaly and seizures. It affects 1 in 10 000-15 000 females. RTT is caused by mutations in the MECP2 gene, which is located in Xq28 and subject to X inactivation. MECP2 encodes a methyl-CpG-binding protein that binds to 5-methyl-cytosine in DNA through its methyl-binding domain. Recruitment of a transcriptional silencing complex through MeCP2's transcriptional repression domain results in histone deacetylation and chromatin condensation. To study the effects of two common truncating RTT mutations (R168X and 803delG), we examined mutant MeCP2 expression and global histone acetylation levels in clonal cell cultures from a female RTT patient with the mutant R168X allele on the active X chromosome, as well as in cells from a male hemizygous for the frameshift mutation 803delG (V288X). Both mutant alleles generated stable RNA transcripts, but no intact MeCP2 protein was detected with an antibody against the C-terminal region of MeCP2. Western blots with antibodies against acetylated histones H3 and H4 revealed that H4, but not H3, was hyperacetylated. By using antibodies against individual acetylated lysine residues, the observed H4 hyperacetylation was attributed to increased acetylation of lysine 16. Therefore, expression of endogenous truncating MECP2 alleles, in the absence of wild-type MeCP2 protein, is specifically associated with an increase in the mono-acetylated histone isoform H4K16. This observed effect may result in over-expression of MeCP2 target genes and, thus, play a role in the pathogenesis of RTT.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Proteínas Represoras , Síndrome de Rett/genética , Acetilación , Alelos , Western Blotting , Línea Celular , Análisis Mutacional de ADN , Proteínas de Unión al ADN/inmunología , Femenino , Mutación del Sistema de Lectura , Humanos , Lisina/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG , Mutación , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Síndrome de Rett/enzimología , Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...