Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 300(5): 107263, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582451

RESUMEN

Synapse formation depends on the coordinated expression and regulation of scaffold proteins. The JNK family kinases play a role in scaffold protein regulation, but the nature of this functional interaction in dendritic spines requires further investigation. Here, using a combination of biochemical methods and live-cell imaging strategies, we show that the dynamics of the synaptic scaffold molecule SAP102 are negatively regulated by JNK inhibition, that SAP102 is a direct phosphorylation target of JNK3, and that SAP102 regulation by JNK is restricted to neurons that harbor mature synapses. We further demonstrate that SAP102 and JNK3 cooperate in the regulated trafficking of kainate receptors to the cell membrane. Specifically, we observe that SAP102, JNK3, and the kainate receptor subunit GluK2 exhibit overlapping expression at synaptic sites and that modulating JNK activity influences the surface expression of the kainate receptor subunit GluK2 in a neuronal context. We also show that SAP102 participates in this process in a JNK-dependent fashion. In summary, our data support a model in which JNK-mediated regulation of SAP102 influences the dynamic trafficking of kainate receptors to postsynaptic sites, and thus shed light on common pathophysiological mechanisms underlying the cognitive developmental defects associated with diverse mutations.


Asunto(s)
Espinas Dendríticas , Receptor de Ácido Kaínico GluK2 , Receptores de Ácido Kaínico , Animales , Humanos , Ratas , Membrana Celular/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Hipocampo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/genética , Neuronas/metabolismo , Neuropéptidos , Fosforilación , Transporte de Proteínas , Receptores de Ácido Kaínico/metabolismo , Receptores de Ácido Kaínico/genética , Sinapsis/metabolismo , Células Cultivadas
2.
PLoS Biol ; 20(3): e3001503, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35312684

RESUMEN

Recent advances in imaging technology have highlighted that scaffold proteins and receptors are arranged in subsynaptic nanodomains. The synaptic membrane-associated guanylate kinase (MAGUK) scaffold protein membrane protein palmitoylated 2 (MPP2) is a component of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-associated protein complexes and also binds to the synaptic cell adhesion molecule SynCAM 1. Using superresolution imaging, we show that-like SynCAM 1-MPP2 is situated at the periphery of the postsynaptic density (PSD). In order to explore MPP2-associated protein complexes, we used a quantitative comparative proteomics approach and identified multiple γ-aminobutyric acid (GABA)A receptor subunits among novel synaptic MPP2 interactors. In line with a scaffold function for MPP2 in the assembly and/or modulation of intact GABAA receptors, manipulating MPP2 expression had effects on inhibitory synaptic transmission. We further show that GABAA receptors are found together with MPP2 in a subset of dendritic spines and thus highlight MPP2 as a scaffold that serves as an adaptor molecule, linking peripheral synaptic elements critical for inhibitory regulation to central structures at the PSD of glutamatergic synapses.


Asunto(s)
Proteínas de la Membrana , Densidad Postsináptica , Proteínas de la Membrana/metabolismo , Densidad Postsináptica/metabolismo , Receptores AMPA/metabolismo , Receptores de GABA-A , Sinapsis/metabolismo
3.
Nature ; 486(7402): 256-60, 2012 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-22699619

RESUMEN

Autism spectrum disorders comprise a range of neurodevelopmental disorders characterized by deficits in social interaction and communication, and by repetitive behaviour. Mutations in synaptic proteins such as neuroligins, neurexins, GKAPs/SAPAPs and ProSAPs/Shanks were identified in patients with autism spectrum disorder, but the causative mechanisms remain largely unknown. ProSAPs/Shanks build large homo- and heteromeric protein complexes at excitatory synapses and organize the complex protein machinery of the postsynaptic density in a laminar fashion. Here we demonstrate that genetic deletion of ProSAP1/Shank2 results in an early, brain-region-specific upregulation of ionotropic glutamate receptors at the synapse and increased levels of ProSAP2/Shank3. Moreover, ProSAP1/Shank2(-/-) mutants exhibit fewer dendritic spines and show reduced basal synaptic transmission, a reduced frequency of miniature excitatory postsynaptic currents and enhanced N-methyl-d-aspartate receptor-mediated excitatory currents at the physiological level. Mutants are extremely hyperactive and display profound autistic-like behavioural alterations including repetitive grooming as well as abnormalities in vocal and social behaviours. By comparing the data on ProSAP1/Shank2(-/-) mutants with ProSAP2/Shank3αß(-/-) mice, we show that different abnormalities in synaptic glutamate receptor expression can cause alterations in social interactions and communication. Accordingly, we propose that appropriate therapies for autism spectrum disorders are to be carefully matched to the underlying synaptopathic phenotype.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Trastorno Autístico/genética , Conducta Animal/fisiología , Proteínas del Tejido Nervioso/genética , Agitación Psicomotora/genética , Animales , Trastorno Autístico/patología , Espinas Dendríticas/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Agitación Psicomotora/patología , Receptores Ionotrópicos de Glutamato/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba , Vocalización Animal/fisiología
4.
Hum Genet ; 132(4): 461-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23329067

RESUMEN

The c-Jun N-terminal kinases (JNKs) are stress-activated serine-threonine kinases that have recently been linked to various neurological disorders. We previously described a patient with intellectual disability (ID) and seizures (Patient 1), carrying a de novo chromosome translocation affecting the CNS-expressed MAPK10/JNK3 gene. Here, we describe a second ID patient (Patient 2) with a similar translocation that likewise truncates MAPK10/JNK3, highlighting a role for JNK3 in human brain development. We have pinpointed the breakpoint in Patient 2, which is just distal to that in Patient 1. In both patients, the rearrangement resulted in a predicted protein interrupted towards the C-terminal end of the kinase domain. We demonstrate that these truncated proteins, although capable of weak interaction with various known JNK scaffolds, are not capable of phosphorylating the classical JNK target c-Jun in vitro, which suggests that the patient phenotype potentially arises from partial loss of JNK3 function. We next investigated JNK3-binding partners to further explore potential disease mechanisms. We identified PSD-95, SAP102 and SHANK3 as novel interaction partners for JNK3, and we demonstrate that JNK3 and PSD-95 exhibit partially overlapping expression at synaptic sites in cultured hippocampal neurons. Moreover, JNK3, like JNK1, is capable of phosphorylating PSD-95 in vitro, whereas disease-associated mutant JNK3 proteins do not. We conclude that reduced JNK3 activity has potentially deleterious effects on neuronal function via altered regulation of a set of post-synaptic proteins.


Asunto(s)
Secuencia de Aminoácidos/genética , Discapacidad Intelectual/genética , Proteína Quinasa 10 Activada por Mitógenos/genética , Convulsiones/metabolismo , Eliminación de Secuencia , Translocación Genética , Adolescente , Animales , Células COS , Chlorocebus aethiops , Homólogo 4 de la Proteína Discs Large , Femenino , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Convulsiones/genética , Convulsiones/patología , Sinapsis/genética , Sinapsis/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Sci Rep ; 10(1): 5709, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32235845

RESUMEN

Scaffold proteins are responsible for structural organisation within cells; they form complexes with other proteins to facilitate signalling pathways and catalytic reactions. The scaffold protein connector enhancer of kinase suppressor of Ras 2 (CNK2) is predominantly expressed in neural tissues and was recently implicated in X-linked intellectual disability (ID). We have investigated the role of CNK2 in neurons in order to contribute to our understanding of how CNK2 alterations might cause developmental defects, and we have elucidated a functional role for CNK2 in the molecular processes that govern morphology of the postsynaptic density (PSD). We have also identified novel CNK2 interaction partners and explored their functional interdependency with CNK2. We focussed on the novel interaction partner TRAF2- and NCK-interacting kinase TNIK, which is also associated with ID. Both CNK2 and TNIK are expressed in neuronal dendrites and concentrated in dendritic spines, and staining with synaptic markers indicates a clear postsynaptic localisation. Importantly, our data highlight that CNK2 plays a role in directing TNIK subcellular localisation, and in neurons, CNK2 participates in ensuring that this multifunctional kinase is present in the correct place at desirable levels. In summary, our data indicate that CNK2 expression is critical for modulating PSD morphology; moreover, our study highlights that CNK2 functions as a scaffold with the potential to direct the localisation of regulatory proteins within the cell. Importantly, we describe a novel link between CNK2 and the regulatory kinase TNIK, and provide evidence supporting the idea that alterations in CNK2 localisation and expression have the potential to influence the behaviour of TNIK and other important regulatory molecules in neurons.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Espinas Dendríticas/metabolismo , Neuronas/metabolismo , Densidad Postsináptica/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Cricetulus , Hipocampo/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Sinapsis/metabolismo
6.
Amyotroph Lateral Scler ; 10(3): 162-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18985462

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating disorder of the central nervous system that leads to progressive loss of upper and lower motor neurons. Most cases are sporadic and of unknown aetiology. In this study, we screened 72 patients with sporadic ALS for the presence of DNA copy number variations, in order to identify novel candidate disease genes. We have used sub-megabase resolution BAC array comparative genomic hybridization to detect genomic imbalances in our ALS patient cohort. Aberrations with potential relevance for disease aetiology were verified by oligo array CGH. In 72 patients with sporadic ALS, we identified a total of six duplications and five deletions that scored above our threshold. Nine of these 11 variations were smaller than 1Mb, and five were observed exclusively in ALS patients. In conclusion, non-polymorphic sub-microscopic duplications and deletions observable by array CGH are frequent in patients with sporadic ALS. Analysis of such aberrations serves as a starting point in deciphering the aetiology of this complex disease, given that affected genes can be considered candidates for influencing disease susceptibility.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Hibridación Genómica Comparativa , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Esclerosis Amiotrófica Lateral/fisiopatología , Dosificación de Gen , Predisposición Genética a la Enfermedad , Humanos
7.
Elife ; 82019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30864948

RESUMEN

PSD-95 MAGUK family scaffold proteins are multi-domain organisers of synaptic transmission that contain three PDZ domains followed by an SH3-GK domain tandem. This domain architecture allows coordinated assembly of protein complexes composed of neurotransmitter receptors, synaptic adhesion molecules and downstream signalling effectors. Here we show that binding of monomeric CRIPT-derived PDZ3 ligands to the third PDZ domain of PSD-95 induces functional changes in the intramolecular SH3-GK domain assembly that influence subsequent homotypic and heterotypic complex formation. We identify PSD-95 interactors that differentially bind to the SH3-GK domain tandem depending on its conformational state. Among these interactors, we further establish the heterotrimeric G protein subunit Gnb5 as a PSD-95 complex partner at dendritic spines of rat hippocampal neurons. The PSD-95 GK domain binds to Gnb5, and this interaction is triggered by CRIPT-derived PDZ3 ligands binding to the third PDZ domain of PSD-95, unraveling a hierarchical binding mechanism of PSD-95 complex formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Homólogo 4 de la Proteína Discs Large/química , Homólogo 4 de la Proteína Discs Large/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Multimerización de Proteína , Sinapsis/química , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Hipocampo/citología , Humanos , Unión Proteica , Conformación Proteica , Dominios Proteicos
8.
eNeuro ; 5(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30023428

RESUMEN

Autism spectrum disorders (ASDs) are neurodevelopmental disorders with a strong genetic etiology. Since mutations in human SHANK genes have been found in patients with autism, genetic mouse models are used for a mechanistic understanding of ASDs and the development of therapeutic strategies. SHANKs are scaffold proteins in the postsynaptic density of mammalian excitatory synapses with proposed functions in synaptogenesis, regulation of dendritic spine morphology, and instruction of structural synaptic plasticity. In contrast to all studies so far on the function of SHANK proteins, we have previously observed enhanced synaptic plasticity in Shank2 Δex7-/- mice. In a series of experiments, we now reproduce these results, further explore the synaptic phenotype, and directly compare our model to the independently generated Shank2 Δex6-7-/- mice. Minimal stimulation experiments reveal that Shank2 Δex7-/- mice possess an excessive fraction of silent (i.e., α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, short, AMPA receptor lacking) synapses. The synaptic maturation deficit emerges during the third postnatal week and constitutes a plausible mechanistic explanation for the mutants' increased capacity for long-term potentiation, both in vivo and in vitro. A direct comparison with Shank2 Δex6-7-/- mice adds weight to the hypothesis that both mouse models show a different set of synaptic phenotypes, possibly due to differences in their genetic background. These findings add to the diversity of synaptic phenotypes in neurodevelopmental disorders and further support the supposed existence of "modifier genes" in the expression and inheritance of ASDs.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Potenciación a Largo Plazo , Proteínas del Tejido Nervioso/fisiología , Sinapsis/fisiología , Animales , Trastorno del Espectro Autista/genética , Modelos Animales de Enfermedad , Hipocampo/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Receptores AMPA/fisiología
9.
FEBS Open Bio ; 7(9): 1234-1245, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28904854

RESUMEN

Synaptic α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors are essential mediators of neurotransmission in the central nervous system. Shisa9/cysteine-knot AMPAR modulating protein 44 (CKAMP44) is a transmembrane protein recently found to be present in AMPA receptor-associated protein complexes. Here, we show that the cytosolic tail of Shisa9/CKAMP44 interacts with multiple scaffold proteins that are important for regulating synaptic plasticity in central neurons. We focussed on the interaction with the scaffold protein PICK1, which facilitates the formation of a tripartite complex with the protein kinase C (PKC) and thereby regulates phosphorylation of Shisa9/CKAMP44 C-terminal residues. This work has implications for our understanding of how PICK1 modulates AMPAR-mediated transmission and plasticity and also highlights a novel function of PKC.

10.
Sci Rep ; 6: 35283, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27756895

RESUMEN

At neuronal synapses, multiprotein complexes of trans-synaptic adhesion molecules, scaffold proteins and neurotransmitter receptors assemble to essential building blocks required for synapse formation and maintenance. Here we describe a novel role for the membrane-associated guanylate kinase (MAGUK) protein MPP2 (MAGUK p55 subfamily member 2) at synapses of rat central neurons. Through interactions mediated by its C-terminal SH3-GK domain module, MPP2 binds to the abundant postsynaptic scaffold proteins PSD-95 and GKAP and localises to postsynaptic sites in hippocampal neurons. MPP2 also colocalises with the synaptic adhesion molecule SynCAM1. We demonstrate that the SynCAM1 C-terminus interacts directly with the MPP2 PDZ domain and that MPP2 does not interact in this manner with other highly abundant postsynaptic transmembrane proteins. Our results highlight a previously unexplored role for MPP2 at postsynaptic sites as a scaffold that links SynCAM1 cell adhesion molecules to core proteins of the postsynaptic density.


Asunto(s)
Moléculas de Adhesión Celular/genética , Inmunoglobulinas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Homólogo 4 de la Proteína Discs Large/genética , Homólogo 4 de la Proteína Discs Large/metabolismo , Hipocampo/metabolismo , Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Fosforilación , Unión Proteica/genética , Ratas , Proteínas Asociadas a SAP90-PSD95/genética , Proteínas Asociadas a SAP90-PSD95/metabolismo , Sinapsis/genética , Dominios Homologos src/genética
11.
Chem Biol ; 20(8): 1044-54, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973190

RESUMEN

To examine the scaffolding properties of PSD-95, we have taken advantage of established ligand/PDZ domain interactions and developed a cell-based assay for investigating protein complex formation. This assay enables quantitative analysis of PDZ domain-mediated protein clustering using bimolecular fluorescence complementation (BiFC). Two nonfluorescent halves of EYFP were fused to C-terminal PDZ ligand sequences to generate probes that sense for PDZ domain binding grooves of adjacent (interacting) molecules. When these probes are brought into proximity by the PDZ domains of a multiprotein scaffold, a functional fluorescent EYFP molecule can be detected. We have used this system to examine the properties of selected PSD-95 variants and thereby delineated regions of importance for PSD-95 complex formation. Further analysis led to the finding that PSD-95 multimerization is PDZ domain-mediated and promoted by ligand binding.


Asunto(s)
Guanilato-Quinasas/metabolismo , Dominios PDZ , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Citometría de Flujo/métodos , Fluorescencia , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Guanilato-Quinasas/química , Ligandos , Microscopía Confocal/métodos , Modelos Moleculares , Unión Proteica , Multimerización de Proteína
12.
Hum Genet ; 121(3-4): 501-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17211639

RESUMEN

We report on three unrelated mentally disabled patients, each carrying a de novo balanced translocation that truncates the autism susceptibility candidate 2 (AUTS2) gene at 7q11.2. One of our patients shows relatively mild mental retardation; the other two display more profound disorders. One patient is also physically disabled, exhibiting urogenital and limb malformations in addition to severe mental retardation. The function of AUTS2 is presently unknown, but it has been shown to be disrupted in monozygotic twins with autism and mental retardation, both carrying a translocation t(7;20)(q11.2;p11.2) (de la Barra et al. in Rev Chil Pediatr 57:549-554, 1986; Sultana et al. in Genomics 80:129-134, 2002). Given the overlap of this autism/mental retardation (MR) phenotype and the MR-associated disorders in our patients, together with the fact that mapping of the additional autosomal breakpoints involved did not disclose obvious candidate disease genes, we ascertain with this study that AUTS2 mutations are clearly linked to autosomal dominant mental retardation.


Asunto(s)
Cromosomas Humanos Par 7 , Genes Dominantes , Discapacidad Intelectual/genética , Mutación , Anomalías Múltiples/genética , Adulto , Trastorno Autístico/genética , Niño , Rotura Cromosómica , Femenino , Humanos , Masculino , Translocación Genética
13.
Hum Genet ; 118(5): 559-67, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16249883

RESUMEN

We have investigated the breakpoints in a male child with pharmacoresistant epileptic encephalopathy and a de novo balanced translocation t(Y;4)(q11.2;q21). By fluorescence in situ hybridisation, we have identified genomic clones from both chromosome 4 and chromosome Y that span the breakpoints. Precise mapping of the chromosome 4 breakpoint indicated that the c-Jun N-terminal kinase 3 (JNK3) gene is disrupted in the patient. This gene is predominantly expressed in the central nervous system, and it plays an established role in both neuronal differentiation and apoptosis. Expression studies in the patient lymphoblastoid cell line show that the truncated JNK3 protein is expressed, i.e. the disrupted transcript is not immediately subject to nonsense-mediated mRNA decay, as is often the case for truncated mRNAs or those harbouring premature termination codons. Over-expression studies with the mutant protein in various cell lines, including neural cells, indicate that both its solubility and cellular localisation differ from that of the wild-type JNK3. It is plausible, therefore, that the presence of the truncated JNK3 disrupts normal JNK3 signal transduction in neuronal cells. JNK3 is one of the downstream effectors of the GTPase-regulated MAP kinase cascade, several members of which have been implicated in cognitive function. In addition, two known JNK3-interacting proteins, beta-arrestin 2 and JIP3, play established roles in neurite outgrowth and neurological development. These interactions are likely affected by a truncated JNK3 protein, and thereby provide an explanation for the link between alterations in MAP kinase signal transduction and brain disorders.


Asunto(s)
Encefalopatías/genética , Sistema Nervioso Central/metabolismo , Epilepsia/genética , Proteína Quinasa 10 Activada por Mitógenos/genética , Secuencia de Bases , Western Blotting , Cromosomas Humanos Par 4 , Cromosomas Humanos Y , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , ARN Mensajero/genética , Índice de Severidad de la Enfermedad , Translocación Genética
14.
Hum Genet ; 120(2): 171-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16783569

RESUMEN

We report on a large family in which a novel X-linked recessive mental retardation (XLMR) syndrome comprising macrocephaly and ciliary dysfunction co-segregates with a frameshift mutation in the OFD1 gene. Mutations of OFD1 have been associated with oral-facial-digital type 1 syndrome (OFD1S) that is characterized by X-chromosomal dominant inheritance and lethality in males. In contrast, the carrier females of our family were clinically inconspicuous, and the affected males suffered from severe mental retardation, recurrent respiratory tract infections and macrocephaly. All but one of the affected males died from respiratory problems in infancy; and impaired ciliary motility was confirmed in the index patient by high-speed video microscopy examination of nasal epithelium. This family broadens the phenotypic spectrum of OFD1 mutations in an unexpected way and sheds light on the complexity of the underlying disease mechanisms.


Asunto(s)
Trastornos de la Motilidad Ciliar/genética , Anomalías Craneofaciales/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteínas/genética , Alelos , Northern Blotting , Niño , Mapeo Cromosómico , Trastornos de la Motilidad Ciliar/patología , Anomalías Craneofaciales/patología , ADN Complementario , Femenino , Mutación del Sistema de Lectura , Genes Recesivos , Heterocigoto , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/patología , Linaje , Síndrome
15.
Hum Genet ; 117(6): 536-44, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16133170

RESUMEN

We have investigated the chromosome abnormalities in a female patient exhibiting a severe cognitive disability associated with complete agenesis of the corpus callosum and microcephaly. The patient carries a balanced de novo translocation t(2;14)(p22;q12), together with a neighbouring 720 kb inversion in chromosome 14q12. By combined fluorescence in situ hybridisation and Southern hybridisation, the distal inversion breakpoint on chromosome 14 was mapped to a region harbouring genes and ESTs derived predominantly from brain tissue. RT-PCR studies indicated that these transcripts comprise the 3' ends of novel splice variants of the winged helix transcription factor FOXG1B (also referred to in previous studies as FOXG1A and FOXG1C, as well as Brain Factor 1), the mouse orthologue of which is essential for normal development of the telencephalon. Analysis of these novel FOXG1B transcripts indicated that they are all disrupted by the breakpoint in the patient. Moreover, we have identified novel orthologous Foxg1 transcripts in the mouse and other vertebrates, which validates the functional importance of these variants and provides a direct genetic link between the patient phenotype and that of the heterozygous Foxg1 knockout mice. These results, together with previously published studies on patients with similar disorders and proximal 14q deletions, strongly suggest that several disorders associated with malformations of the human brain may be directly caused by mutations or alterations in the FOXG1B gene.


Asunto(s)
Agenesia del Cuerpo Calloso , Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Proteínas del Tejido Nervioso/genética , Anomalías Múltiples/genética , Animales , Células Cultivadas , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 2 , Femenino , Factores de Transcripción Forkhead , Humanos , Recién Nacido , Ratones , Datos de Secuencia Molecular , Transfección , Translocación Genética
16.
Hum Mol Genet ; 11(7): 815-21, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11929854

RESUMEN

Friedreich ataxia is an inherited disorder caused by decreased expression of frataxin protein. Increasing evidence suggests that this protein might detoxify reactive oxygen species (ROS) by an unknown mechanism. Here we demonstrate that transgenic overexpression of human frataxin increases cellular antioxidant defense via activation of glutathione peroxidase and elevation of reduced thiols, thereby reducing the incidence of malignant transformation induced by ROS, as observed by soft agar assays and tumour formation in nude mice. These findings expand the understanding of antioxidant properties of frataxin, and tentatively suggest a role in the early induction of cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Ataxia de Friedreich/genética , Glutatión Peroxidasa/metabolismo , Proteínas de Unión a Hierro , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Especies Reactivas de Oxígeno/metabolismo , Células 3T3 , Animales , Regulación de la Expresión Génica , Técnicas In Vitro , Ratones , Microscopía Confocal , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Transgenes , Frataxina
17.
Am J Hum Genet ; 73(6): 1341-54, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14628291

RESUMEN

Nonsyndromic X-linked mental retardation (MRX) is defined by an X-linked inheritance pattern of low IQ, problems with adaptive behavior, and the absence of additional specific clinical features. The 13 MRX genes identified to date account for less than one-fifth of all MRX, suggesting that numerous gene defects cause the disorder in other families. In a female patient with severe nonsyndromic mental retardation and a de novo balanced translocation t(X;7)(p11.3;q11.21), we have cloned the DNA fragment that contains the X-chromosomal and the autosomal breakpoint. In silico sequence analysis provided no indication of a causative role for the chromosome 7 breakpoint in mental retardation (MR), whereas, on the X chromosome, a zinc-finger gene, ZNF41, was found to be disrupted. Expression studies indicated that ZNF41 transcripts are absent in the patient cell line, suggesting that the mental disorder in this patient results from loss of functional ZNF41. Moreover, screening of a panel of patients with MRX led to the identification of two other ZNF41 mutations that were not found in healthy control individuals. A proline-to-leucine amino acid exchange is present in affected members of one family with MRX. A second family carries an intronic splice-site mutation that results in loss of specific ZNF41 splice variants. Wild-type ZNF41 contains a highly conserved transcriptional repressor domain that is linked to mechanisms of chromatin remodeling, a process that is defective in various other forms of MR. Our results suggest that ZNF41 is critical for cognitive development; further studies aim to elucidate the specific mechanisms by which ZNF41 alterations lead to MR.


Asunto(s)
Cromosomas Humanos X/genética , Trastornos del Conocimiento/genética , Proteínas de Unión al ADN/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mutación/genética , Secuencia de Bases , Northern Blotting , Southern Blotting , Ensamble y Desensamble de Cromatina/genética , Cromatografía Líquida de Alta Presión , Mapeo Cromosómico , Cartilla de ADN , Femenino , Expresión Génica/genética , Humanos , Hibridación Fluorescente in Situ , Factores de Transcripción de Tipo Kruppel , Datos de Secuencia Molecular , Linaje , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA