Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Cell Neurosci ; 14: 1, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32038177

RESUMEN

The epilepsy of infancy with migrating focal seizures (EIMFS; previously called Malignant migrating partial seizures of infancy) are early-onset epileptic encephalopathies (EOEE) that associate multifocal ictal discharges and profound psychomotor retardation. EIMFS have a genetic origin and are mostly caused by de novo mutations in the KCNT1 gene, and much more rarely in the KCNT2 gene. KCNT1 and KCNT2 respectively encode the KNa1.1 (Slack) and KNa1.2 (Slick) subunits of the sodium-dependent voltage-gated potassium channel KNa. Functional analyses of the corresponding mutant homomeric channels in vitro suggested gain-of-function effects. Here, we report two novel, de novo truncating mutations of KCNT2: one mutation is frameshift (p.L48Qfs43), is situated in the N-terminal domain, and was found in a patient with EOEE (possibly EIMFS); the other mutation is nonsense (p.K564*), is located in the C-terminal region, and was found in a typical EIMFS patient. Using whole-cell patch-clamp recordings, we have analyzed the functional consequences of those two novel KCNT2 mutations on reconstituted KNa1.2 homomeric and KNa1.1/KNa1.2 heteromeric channels in transfected chinese hamster ovary (CHO) cells. We report that both mutations significantly impacted on KNa function; notably, they decreased the global current density of heteromeric channels by ~25% (p.K564*) and ~55% (p.L48Qfs43). Overall our data emphasize the involvement of KCNT2 in EOEE and provide novel insights into the role of heteromeric KNa channel in the severe KCNT2-related epileptic phenotypes. This may have important implications regarding the elaboration of future treatment.

2.
Front Cell Neurosci ; 12: 55, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29559892

RESUMEN

Congenital cytomegalovirus (CMV) infections represent one leading cause of neurodevelopmental disorders. Recently, we reported on a rat model of CMV infection of the developing brain in utero, characterized by early and prominent infection and alteration of microglia-the brain-resident mononuclear phagocytes. Besides their canonical function against pathogens, microglia are also pivotal to brain development. Here we show that CMV infection of the rat fetal brain recapitulated key postnatal phenotypes of human congenital CMV including increased mortality, sensorimotor impairment reminiscent of cerebral palsy, hearing defects, and epileptic seizures. The possible influence of early microglia alteration on those phenotypes was then questioned by pharmacological targeting of microglia during pregnancy. One single administration of clodronate liposomes in the embryonic brains at the time of CMV injection to deplete microglia, and maternal feeding with doxycyxline throughout pregnancy to modify microglia in the litters' brains, were both associated with dramatic improvements of survival, body weight gain, sensorimotor development and with decreased risk of epileptic seizures. Improvement of microglia activation status did not persist postnatally after doxycycline discontinuation; also, active brain infection remained unchanged by doxycycline. Altogether our data indicate that early microglia alteration, rather than brain CMV load per se, is instrumental in influencing survival and the neurological outcomes of CMV-infected rats, and suggest that microglia might participate in the neurological outcome of congenital CMV in humans. Furthermore this study represents a first proof-of-principle for the design of microglia-targeted preventive strategies in the context of congenital CMV infection of the brain.

3.
Methods Mol Biol ; 1677: 129-135, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28986869

RESUMEN

The functional study of reconstituted NMDA receptors (NMDARs) in host cells requires that the corresponding vectors for the expression of the NMDAR subunits are co-transfected with high efficiency. Magnetofection™ is a technology used to deliver nucleic acids to cells. It is driven and site-specifically guided by the attractive forces of magnetic fields acting on magnetic nanoparticles that are associated with nucleic acid vectors. In magnetofection™, cationic lipids form self-assembled complexes with the nucleic acid vectors of interest. Those complexes are then associated with magnetic nanoparticles that are concentrated at the surface of cultured cells by applying a permanent magnetic field. Magnetofection™ is a simple method to transfect cultured cells with high transfection rates. Satisfactory expression levels are obtained with very low amounts of nucleic acid vector. Moreover, incubation time with host cells is less than 1 h, as compared with the several hours needed with standard transfection assays.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Vectores Genéticos/genética , Células HEK293 , Humanos , Nanopartículas de Magnetita , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Transfección
4.
Front Cell Neurosci ; 11: 155, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28611597

RESUMEN

Genetic variants of the glutamate activated N-methyl-D-aspartate (NMDA) receptor (NMDAR) subunit GluN2A are associated with the hyperexcitable states manifested by epileptic seizures and interictal discharges in patients with disorders of the epilepsy-aphasia spectrum (EAS). The variants found in sporadic cases and families are of different types and include microdeletions encompassing the corresponding GRIN2A gene as well as nonsense, splice-site and missense GRIN2A defects. They are located at different functional domains of GluN2A and no clear genotype-phenotype correlation has emerged yet. Moreover, GluN2A variants may be associated with phenotypic pleiotropy. Deciphering the consequences of pathogenic GRIN2A variants would surely help in better understanding of the underlying mechanisms. This emphasizes the need for functional studies to unravel the basic functional properties of each specific NMDAR variant. In the present study, we have used patch-clamp recordings to evaluate kinetic changes of mutant NMDARs reconstituted after co-transfection of cultured cells with the appropriate expression vectors. Three previously identified missense variants found in patients or families with disorders of the EAS and situated in the N-terminal domain (p.Ile184Ser) or in the ligand-binding domain (p.Arg518His and p.Ala716Thr) of GluN2A were studied in both the homozygous and heterozygous conditions. Relative surface expression and current amplitude were significantly reduced for NMDARs composed of mutant p.Ile184Ser and p.Arg518His, but not p.Ala716His, as compared with wild-type (WT) NMDARs. Amplitude of whole-cell currents was still drastically decreased when WT and mutant p.Arg518His-GluN2A subunits were co-expressed, suggesting a dominant-negative mechanism. Activation times were significantly decreased in both homozygous and heterozygous conditions for the two p.Ile184Ser and p.Arg518His variants, but not for p.Ala716His. Deactivation also significantly increased for p.Ile184Ser variant in the homozygous but not the heterozygous state while it was increased for p.Arg518His in both states. Our data indicate that p.Ile184Ser and p.Arg518His GluN2A variants both impacted on NMDAR function, albeit differently, whereas p.Ala716His did not significantly influence NMDAR kinetics, hence partly questioning its direct and strong pathogenic role. This study brings new insights into the functional impact that GRIN2A variants might have on NMDAR kinetics, and provides a mechanistic explanation for the neurological manifestations seen in the corresponding human spectrum of disorders.

5.
PLoS One ; 11(7): e0160176, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27472761

RESUMEN

BACKGROUND: Congenital cytomegalovirus infections are a leading cause of neurodevelopmental disorders in human and represent a major health care and socio-economical burden. In contrast with this medical importance, the pathophysiological events remain poorly known. Murine models of brain cytomegalovirus infection, mostly neonatal, have brought recent insights into the possible pathogenesis, with convergent evidence for the alteration and possible involvement of brain immune cells. OBJECTIVES AND METHODS: In order to confirm and expand those findings, particularly concerning the early developmental stages following infection of the fetal brain, we have created a model of in utero cytomegalovirus infection in the developing rat brain. Rat cytomegalovirus was injected intraventricularly at embryonic day 15 (E15) and the brains analyzed at various stages until the first postnatal day, using a combination of gene expression analysis, immunohistochemistry and multicolor flow cytometry experiments. RESULTS: Rat cytomegalovirus infection was increasingly seen in various brain areas including the choroid plexi and the ventricular and subventricular areas and was prominently detected in CD45low/int, CD11b+ microglial cells, in CD45high, CD11b+ cells of the myeloid lineage including macrophages, and in CD45+, CD11b- lymphocytes and non-B non-T cells. In parallel, rat cytomegalovirus infection of the developing rat brain rapidly triggered a cascade of pathophysiological events comprising: chemokines upregulation, including CCL2-4, 7 and 12; infiltration by peripheral cells including B-cells and monocytes at E17 and P1, and T-cells at P1; and microglia activation at E17 and P1. CONCLUSION: In line with previous findings in neonatal murine models and in human specimen, our study further suggests that neuroimmune alterations might play critical roles in the early stages following cytomegalovirus infection of the brain in utero. Further studies are now needed to determine which role, whether favorable or detrimental, those putative double-edge swords events actually play.


Asunto(s)
Encéfalo/embriología , Infecciones por Citomegalovirus/patología , Microglía/patología , Muromegalovirus/patogenicidad , Animales , Linaje de la Célula , Infecciones por Citomegalovirus/inmunología , Citometría de Flujo , Activación de Macrófagos , Microglía/inmunología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Eur J Hum Genet ; 24(12): 1761-1770, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27352968

RESUMEN

Genetic generalized epilepsy (GGE), formerly known as idiopathic generalized epilepsy, is the most common form of epilepsy and is thought to have predominant genetic etiology. GGE are clinically characterized by absence, myoclonic, or generalized tonic-clonic seizures with electroencephalographic pattern of bilateral, synchronous, and symmetrical spike-and-wave discharges. Despite their strong heritability, the genetic basis of generalized epilepsies remains largely elusive. Nevertheless, recent advances in genetic technology have led to the identification of numerous genes and genomic defects in various types of epilepsies in the past few years. In the present study, we performed whole-exome sequencing in a family with GGE consistent with the diagnosis of eyelid myoclonia with absences. We found a nonsense variant (c.196C>T/p.(Arg66*)) in RORB, which encodes the beta retinoid-related orphan nuclear receptor (RORß), in four affected family members. In addition, two de novo variants (c.218T>C/p.(Leu73Pro); c.1249_1251delACG/p.(Thr417del)) were identified in sporadic patients by trio-based exome sequencing. We also found two de novo deletions in patients with behavioral and cognitive impairment and epilepsy: a 52-kb microdeletion involving exons 5-10 of RORB and a larger 9q21-microdeletion. Furthermore, we identified a patient with intellectual disability and a balanced translocation where one breakpoint truncates RORB and refined the phenotype of a recently reported patient with RORB deletion. Our data support the role of RORB gene variants/CNVs in neurodevelopmental disorders including epilepsy, and especially in generalized epilepsies with predominant absence seizures.


Asunto(s)
Discapacidades del Desarrollo/genética , Epilepsia Generalizada/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Adulto , Niño , Puntos de Rotura del Cromosoma , Deleción Cromosómica , Codón sin Sentido , Discapacidades del Desarrollo/diagnóstico , Epilepsia Generalizada/diagnóstico , Exoma , Exones , Femenino , Humanos , Masculino , Linaje , Síndrome , Translocación Genética
7.
Epilepsia ; 55(2): 370-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24372385

RESUMEN

OBJECTIVES: Rolandic epilepsies (REs) represent the most frequent epilepsy in childhood. Patients may experience cognitive, speech, language, reading, and behavioral issues. The genetic origin of REs has long been debated. The participation of rare copy number variations (CNVs) in the pathophysiology of various human epilepsies has been increasingly recognized. However, no systematic search for microdeletions or microduplications has been reported in RE so far. METHODS: Array comparative genomic hybridization (aCGH) and quantitative polymerase chain reaction (qPCR) were used to analyze the genomic status of a series of 47 unrelated RE patients who displayed various types of electroclinical manifestations. RESULTS: Thirty rare CNVs were detected in 21 RE patients. Two CNVs were de novo, 12 were inherited, and 16 were of unknown inheritance. Each CNV was unique to one given patient, except for a 16p11.2 duplication found in two patients. The CNVs of highest interest comprised or disrupted strong candidate or confirmed genes for epileptic and other neurodevelopmental disorders, including BRWD3, GRIN2A, KCNC3, PRKCE, PRRT2, SHANK1, and TSPAN7. SIGNIFICANCE: Patients with REs showed rare microdeletions and microduplications with high frequency and heterogeneity. Whereas only a subset of all genomic alterations found here may actually participate in the phenotype, the novel de novo events as well as several inherited CNVs contain or disrupt genes, some of which are likely to influence the emergence, the presentation, or the comorbidity of RE. The future screening of cohorts of larger size will help in detecting more de novo or recurrent events and in appreciating the possible enrichment of specific CNVs in patients with RE.


Asunto(s)
Epilepsia Rolándica/diagnóstico , Epilepsia Rolándica/genética , Estudios de Asociación Genética/métodos , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Receptores de N-Metil-D-Aspartato/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Variaciones en el Número de Copia de ADN/genética , Femenino , Heterogeneidad Genética , Humanos , Masculino
8.
Nat Genet ; 45(9): 1073-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23933818

RESUMEN

Epilepsy-aphasia syndromes (EAS) are a group of rare, severe epileptic encephalopathies of unknown etiology with a characteristic electroencephalogram (EEG) pattern and developmental regression particularly affecting language. Rare pathogenic deletions that include GRIN2A have been implicated in neurodevelopmental disorders. We sought to delineate the pathogenic role of GRIN2A in 519 probands with epileptic encephalopathies with diverse epilepsy syndromes. We identified four probands with GRIN2A variants that segregated with the disorder in their families. Notably, all four families presented with EAS, accounting for 9% of epilepsy-aphasia cases. We did not detect pathogenic variants in GRIN2A in other epileptic encephalopathies (n = 475) nor in probands with benign childhood epilepsy with centrotemporal spikes (n = 81). We report the first monogenic cause, to our knowledge, for EAS. GRIN2A mutations are restricted to this group of cases, which has important ramifications for diagnostic testing and treatment and provides new insights into the pathogenesis of this debilitating group of conditions.


Asunto(s)
Síndrome de Landau-Kleffner/genética , Mutación , Receptores de N-Metil-D-Aspartato/genética , Electroencefalografía , Femenino , Humanos , Síndrome de Landau-Kleffner/diagnóstico , Masculino , Linaje , Fenotipo
9.
Nat Genet ; 45(9): 1061-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23933820

RESUMEN

Epileptic encephalopathies are severe brain disorders with the epileptic component contributing to the worsening of cognitive and behavioral manifestations. Acquired epileptic aphasia (Landau-Kleffner syndrome, LKS) and continuous spike and waves during slow-wave sleep syndrome (CSWSS) represent rare and closely related childhood focal epileptic encephalopathies of unknown etiology. They show electroclinical overlap with rolandic epilepsy (the most frequent childhood focal epilepsy) and can be viewed as different clinical expressions of a single pathological entity situated at the crossroads of epileptic, speech, language, cognitive and behavioral disorders. Here we demonstrate that about 20% of cases of LKS, CSWSS and electroclinically atypical rolandic epilepsy often associated with speech impairment can have a genetic origin sustained by de novo or inherited mutations in the GRIN2A gene (encoding the N-methyl-D-aspartate (NMDA) glutamate receptor α2 subunit, GluN2A). The identification of GRIN2A as a major gene for these epileptic encephalopathies provides crucial insights into the underlying pathophysiology.


Asunto(s)
Epilepsias Parciales/genética , Síndrome de Landau-Kleffner/genética , Mutación , Receptores de N-Metil-D-Aspartato/genética , Sustitución de Aminoácidos , Línea Celular , Electroencefalografía , Femenino , Expresión Génica , Genotipo , Humanos , Masculino , Linaje , Fenotipo
10.
Brain ; 136(Pt 8): 2457-73, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23831613

RESUMEN

Altered development of the human cerebral cortex can cause severe malformations with often intractable focal epileptic seizures and may participate in common pathologies, notably epilepsy. This raises important conceptual and therapeutic issues. Two missense mutations in the sushi repeat-containing protein SRPX2 had been previously identified in epileptic disorders with or without structural developmental alteration of the speech cortex. In the present study, we aimed to decipher the precise developmental role of SRPX2, to have a better knowledge on the consequences of its mutations, and to start addressing therapeutic issues through the design of an appropriate animal model. Using an in utero Srpx2 silencing approach, we show that SRPX2 influences neuronal migration in the developing rat cerebral cortex. Wild-type, but not the mutant human SRPX2 proteins, rescued the neuronal migration phenotype caused by Srpx2 silencing in utero, and increased alpha-tubulin acetylation. Following in utero Srpx2 silencing, spontaneous epileptiform activity was recorded post-natally. The neuronal migration defects and the post-natal epileptic consequences were prevented early in embryos by maternal administration of tubulin deacetylase inhibitor tubacin. Hence epileptiform manifestations of developmental origin could be prevented in utero, using a transient and drug-based therapeutic protocol.


Asunto(s)
Anilidas/farmacología , Movimiento Celular/genética , Corteza Cerebral/metabolismo , Epilepsia/genética , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Proteínas de la Membrana/genética , Neuronas/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Epilepsia/metabolismo , Silenciador del Gen , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
11.
Pigment Cell Melanoma Res ; 26(4): 592-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23582189

RESUMEN

Expression of organic anion transporting polypeptides (OATP) transporters can be modified with potential incidence in cancers, yet they have not been considered in melanoma. Here, we demonstrate transcriptional and protein expression of OATP members in human melanoma cell lines with sodium-independent organic anion uptake activity. Importantly, uptake of different organic anions over 24 h led to a common resistance signal to apoptotic cell death, induced further by cisplatin in 24 h. The mechanism is not dependent on the transport of cisplatin by the OATP, as it is not an OATP substrate. The resistance signal was modulated by PKC, disclosing it as signal mediator. This study suggests that OATP, which can be constantly activated by endobiotics, may contribute to melanoma chemotherapeutic resistance, thereby justifying the development of OATP targeting strategies.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Cisplatino/farmacología , Resistencia a Antineoplásicos , Melanoma/metabolismo , Transportadores de Anión Orgánico/química , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Melanocitos/metabolismo , Péptidos/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
12.
Neurology ; 79(21): 2097-103, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23077017

RESUMEN

OBJECTIVE: Whole genome sequencing and the screening of 103 families recently led us to identify PRRT2 (proline-rich-transmembrane protein) as the gene causing infantile convulsions (IC) with paroxysmal kinesigenic dyskinesia (PKD) (PKD/IC syndrome, formerly ICCA). There is interfamilial and intrafamilial variability and the patients may have IC or PKD. Association of IC with hemiplegic migraine (HM) has also been reported. In order to explore the mutational and clinical spectra, we analyzed 34 additional families with either typical PKD/IC or PKD/IC with migraine. METHODS: We performed Sanger sequencing of all PRRT2 coding exons and of exon-intron boundaries in the probands and in their relatives whenever appropriate. RESULTS: Two known and 2 novel PRRT2 mutations were detected in 18 families. The p.R217Pfs*8 recurrent mutation was found in ≈50% of typical PKD/IC, and the unreported p.R145Gfs*31 in one more typical family. PRRT2 mutations were also found in PKD/IC with migraine: p.R217Pfs*8 cosegregated with PKD associated with HM in one family, and was also detected in one IC patient having migraine with aura, in related PKD/IC familial patients having migraine without aura, and in one sporadic migraineur with abnormal MRI. Previously reported p.R240X was found in one patient with PKD with migraine without aura. The novel frameshift p.S248Afs*65 was identified in a PKD/IC family member with IC and migraine with aura. CONCLUSIONS: We extend the spectrum of PRRT2 mutations and phenotypes to HM and to other types of migraine in the context of PKD/IC, and emphasize the phenotypic pleiotropy seen in patients with PRRT2 mutations.


Asunto(s)
Discinesias/diagnóstico , Discinesias/genética , Epilepsia Benigna Neonatal/diagnóstico , Epilepsia Benigna Neonatal/genética , Ligamiento Genético/genética , Proteínas de la Membrana/genética , Trastornos Migrañosos/diagnóstico , Trastornos Migrañosos/genética , Proteínas del Tejido Nervioso/genética , Convulsiones/diagnóstico , Convulsiones/genética , Secuencia de Bases , Corea/diagnóstico , Corea/epidemiología , Corea/genética , Discinesias/epidemiología , Epilepsia Benigna Neonatal/epidemiología , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Trastornos Migrañosos/epidemiología , Datos de Secuencia Molecular , Mutación/genética , Linaje , Convulsiones/epidemiología
13.
Cell Rep ; 1(1): 2-12, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22832103

RESUMEN

Paroxysmal kinesigenic dyskinesia with infantile convulsions (PKD/IC) is an episodic movement disorder with autosomal-dominant inheritance and high penetrance, but the causative genetic mutation is unknown. We have now identified four truncating mutations involving the gene PRRT2 in the vast majority (24/25) of well-characterized families with PKD/IC. PRRT2 truncating mutations were also detected in 28 of 78 additional families. PRRT2 encodes a proline-rich transmembrane protein of unknown function that has been reported to interact with the t-SNARE, SNAP25. PRRT2 localizes to axons but not to dendritic processes in primary neuronal culture, and mutants associated with PKD/IC lead to dramatically reduced PRRT2 levels, leading ultimately to neuronal hyperexcitability that manifests in vivo as PKD/IC.


Asunto(s)
Distonía/complicaciones , Distonía/genética , Proteínas de la Membrana/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Convulsiones/complicaciones , Convulsiones/genética , Alelos , Secuencia de Aminoácidos , Animales , Sistema Nervioso Central/metabolismo , Segregación Cromosómica/genética , Variaciones en el Número de Copia de ADN/genética , Femenino , Genoma Humano/genética , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/química , Linaje , Fenotipo , Unión Proteica/genética , Ratas , Alineación de Secuencia , Análisis de Secuencia de ADN , Especificidad de la Especie , Proteína 25 Asociada a Sinaptosomas/metabolismo
14.
Curr Pharm Des ; 17(19): 1914-23, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21711233

RESUMEN

As a key component of the plasminogen activation system, uPAR, the receptor for the plasminogen activator of the urokinase type, is involved in many physiological and pathological processes. Besides its classical roles, there has been increased evidence that uPAR or uPAR-associated pathways, participate in the development, in the functioning and in the pathology of the central nervous system. Qualitative and quantitative changes in the expressions of uPAR and of its canonical ligand uPA have been observed in a large variety of epileptic disorders, either in human or in animal models, as well as in other brain diseases (stroke and brain trauma, multiple sclerosis, Alzheimer's disease, cerebral malaria, HIV-associated leukoencephalopathy and encephalitis). The variety of such pathological conditions and the different brain areas and cell types involved, likely reflects the wide range and the complexity of the multiple and somehow intertwined pathophysiological mechanisms related with uPAR. In the mouse, the knock-out of the Upar-encoding gene (Plaur) leads to significant and nearly complete loss in parvalbumin-containing interneurons during brain development. This is associated with increased susceptibility to spontaneous and chemically-induced seizures and with increased anxiety and impaired social interactions. The recent identification of the novel uPAR ligand SRPX2 (Sushi repeat protein, X-linked 2) and the regulation of both the SRPX2 and PLAUR genes by transcription factor FOXP2 has shed novel and exciting insights into the role of uPAR-related molecular networks in rolandic epilepsy, in developmental verbal dyspraxia, in perisylvian polymicrogyria, and generally in disorders of the speech areas and circuits. uPAR, its regulators and partners, as well as other proteins containing Ly-6/uPAR/alpha-neurotoxin domains, represent key entry points for present and future studies not only on speech-related disorders but also on epilepsy and autism spectrum disorders.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Trastornos de la Comunicación/metabolismo , Epilepsia/metabolismo , Trastornos del Lenguaje/metabolismo , Trastornos Mentales/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Animales , Humanos
15.
PLoS One ; 5(10): e13750, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21060786

RESUMEN

BACKGROUND: Benign infantile convulsions and paroxysmal dyskinesia are episodic cerebral disorders that can share common genetic bases. They can be co-inherited as one single autosomal dominant trait (ICCA syndrome); the disease ICCA gene maps at chromosome 16p12-q12. Despite intensive and conventional mutation screening, the ICCA gene remains unknown to date. The critical area displays highly complicated genomic architecture and is the site of deletions and duplications associated with various diseases. The possibility that the ICCA syndrome is related to the existence of large-scale genomic alterations was addressed in the present study. METHODOLOGY/PRINCIPAL FINDINGS: A combination of whole genome and dedicated oligonucleotide array comparative genomic hybridization coupled with quantitative polymerase chain reaction was used. Low copy number of a region corresponding to a genomic variant (Variation_7105) located at 16p11 nearby the centromere was detected with statistical significance at much higher frequency in patients from ICCA families than in ethnically matched controls. The genomic variant showed no apparent difference in size and copy number between patients and controls, making it very unlikely that the genomic alteration detected here is ICCA-specific. Furthermore, no other genomic alteration that would directly cause the ICCA syndrome in those nine families was detected in the ICCA critical area. CONCLUSIONS/SIGNIFICANCE: Our data excluded that inherited genomic deletion or duplication events directly cause the ICCA syndrome; rather, they help narrowing down the critical ICCA region dramatically and indicate that the disease ICCA genetic defect lies very close to or within Variation_7105 and hence should now be searched in the corresponding genomic area and its surrounding regions.


Asunto(s)
Corea/genética , Cromosomas Humanos Par 16 , Epilepsia Benigna Neonatal/genética , Dosificación de Gen , Estudios de Casos y Controles , Femenino , Humanos , Hibridación in Situ , Lactante , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Síndrome
16.
Hum Mol Genet ; 19(24): 4848-60, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20858596

RESUMEN

It is a challenge to identify the molecular networks contributing to the neural basis of human speech. Mutations in transcription factor FOXP2 cause difficulties mastering fluent speech (developmental verbal dyspraxia, DVD), whereas mutations of sushi-repeat protein SRPX2 lead to epilepsy of the rolandic (sylvian) speech areas, with DVD or with bilateral perisylvian polymicrogyria. Pathophysiological mechanisms driven by SRPX2 involve modified interaction with the plasminogen activator receptor (uPAR). Independent chromatin-immunoprecipitation microarray screening has identified the uPAR gene promoter as a potential target site bound by FOXP2. Here, we directly tested for the existence of a transcriptional regulatory network between human FOXP2 and the SRPX2/uPAR complex. In silico searches followed by gel retardation assays identified specific efficient FOXP2-binding sites in each of the promoter regions of SRPX2 and uPAR. In FOXP2-transfected cells, significant decreases were observed in the amounts of both SRPX2 (43.6%) and uPAR (38.6%) native transcripts. Luciferase reporter assays demonstrated that FOXP2 expression yielded a marked inhibition of SRPX2 (80.2%) and uPAR (77.5%) promoter activity. A mutant FOXP2 that causes DVD (p.R553H) failed to bind to SRPX2 and uPAR target sites and showed impaired down-regulation of SRPX2 and uPAR promoter activity. In a patient with polymicrogyria of the left rolandic operculum, a novel FOXP2 mutation (p.M406T) was found in the leucine-zipper (dimerization) domain. p.M406T partially impaired the FOXP2 regulation of SRPX2 promoter activity, whereas that of the uPAR promoter remained unchanged. Together with recently described FOXP2-CNTNAP2 and SRPX2/uPAR links, the FOXP2-SRPX2/uPAR network provides exciting insights into molecular pathways underlying speech-related disorders.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Redes Reguladoras de Genes , Proteínas del Tejido Nervioso/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Trastornos del Habla/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso/genética , Ensayo de Cambio de Movilidad Electroforética , Femenino , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/metabolismo , Masculino , Malformaciones del Desarrollo Cortical/complicaciones , Malformaciones del Desarrollo Cortical/genética , Proteínas de la Membrana , Datos de Secuencia Molecular , Mutación Missense/genética , Proteínas de Neoplasias , Proteínas del Tejido Nervioso/genética , Linaje , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Trastornos del Habla/complicaciones
17.
Hum Mol Genet ; 15(7): 1195-207, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16497722

RESUMEN

The rolandic and sylvian fissures divide the human cerebral hemispheres and the adjacent areas participate in speech processing. The relationship of rolandic (sylvian) seizure disorders with speech and cognitive impairments is well known, albeit poorly understood. We have identified the Xq22 gene SRPX2 as being responsible for rolandic seizures (RSs) associated with oral and speech dyspraxia and mental retardation (MR). SRPX2 is a secreted sushi-repeat containing protein expressed in neurons of the human adult brain, including the rolandic area. The disease-causing mutation (N327S) resulted in gain-of-glycosylation of the secreted mutant protein. A second mutation (Y72S) was identified within the first sushi domain of SRPX2 in a male with RSs and bilateral perisylvian polymicrogyria and his female relatives with mild MR or unaffected carrier status. In cultured cells, both mutations were associated with altered patterns of intracellular processing, suggesting protein misfolding. In the murine brain, Srpx2 protein expression appeared in neurons at birth. The involvement of SRPX2 in these disorders suggests an important role for SRPX2 in the perisylvian region critical for language and cognitive development.


Asunto(s)
Corteza Cerebral/metabolismo , Cognición , Trastornos del Lenguaje/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Adulto , Secuencia de Aminoácidos , Animales , Apraxias/genética , Apraxias/metabolismo , Secuencia de Bases , Células CHO , Niño , Preescolar , Cricetinae , Epilepsia Rolándica/genética , Epilepsia Rolándica/metabolismo , Femenino , Fibroblastos/metabolismo , Ligamiento Genético , Pruebas Genéticas , Glicosilación , Humanos , Inmunohistoquímica , Discapacidad Intelectual/metabolismo , Trastornos del Lenguaje/metabolismo , Trastornos del Lenguaje/fisiopatología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias , Proteínas del Tejido Nervioso/metabolismo , Transfección
18.
Arterioscler Thromb Vasc Biol ; 25(2): 359-64, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15569825

RESUMEN

OBJECTIVE: Bile salt-dependent lipase (BSDL), a lipolytic enzyme secreted in the duodenum by pancreatic acinar cells, has been detected in the serum of all patients and in atheromatous plaque, suggesting its potential implication in vascular pathophysiology. METHODS AND RESULTS: In vitro pancreatic BSDL evokes human umbilical vein endothelial cell (HUVEC) proliferation and chemotactic migration. BSDL at mitogen concentration is capable to heal wounded HUVEC monolayer and to promote capillary network formation. HUVEC proliferation depends on the displacement of basic fibroblast growth factor and vascular endothelial growth factor from the extracellular matrix and the activation of extracellular signal-regulated kinases (ERK1/2), p38 mitogen-activated protein kinase, and focal adhesion kinase signaling pathways. CONCLUSIONS: For the first time to our knowledge, it is suggested that circulating BSDL could be involved in pathophysiological angiogenesis. We delineate the in vitro effects of pancreatic BSDL on endothelial cells, and we show that BSDL promotes proliferation, migration, capillary network formation, and wound-healing of HUVECs via the displacement of bFGF and VEGF from the ECM, suggesting that BSDL could be involved in angiogenesis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Esterol Esterasa/farmacología , Animales , División Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Colágeno , Replicación del ADN/efectos de los fármacos , Combinación de Medicamentos , Endotelio Vascular/citología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Laminina , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macaca fascicularis , Neovascularización Fisiológica/fisiología , Páncreas/enzimología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteoglicanos , Venas Umbilicales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
19.
Mol Biol Cell ; 14(7): 2861-75, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12857870

RESUMEN

We have recently shown that the pancreatic bile salt-dependent lipase (BSDL) can be taken up by intestinal cells and transported to the blood circulation. This mechanism likely involves (specific) receptor(s) able to bind BSDL and located at the apical intestinal cell membrane. In this study, using Int407 human intestinal cells cultured to form a tight epithelium, we attempted to characterize (the) BSDL receptor(s). We found that an apical 50-kDa protein was able to bind BSDL. Further, we have demonstrated that Int407 cells expressed the lectin-like oxidized-LDL receptor (LOX-1), the upregulation of which by oxidized-LDL potentiates the transcytosis of BSDL, whereas carrageenan and to a lesser extent polyinosinic acid and fucoidan decrease the enzyme transcytosis. The mAb JTX92, which blocks the LOX-1 receptor function, also impaired the BSDL transcytosis. To confirm these results, the cDNA encoding the human intestinal receptor LOX-1 has been cloned, inserted into vectors, and transfected into Int407 cells. Overexpression of LOX-1 by these cells leads to a substantial increase in the BSDL transcytosis. Globally, these data support the view that LOX-1 could be an intestinal receptor for BSDL, which is implicated in the transcytosis of this enzyme throughout Int407 cells.


Asunto(s)
Endocitosis/fisiología , Receptores de LDL/metabolismo , Esterol Esterasa/metabolismo , Línea Celular , Clonación Molecular , Endocitosis/efectos de los fármacos , Citometría de Flujo , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citología , Lipoproteínas LDL/metabolismo , Poli I/farmacología , Polisacáridos/farmacología , Unión Proteica , Receptores de LDL Oxidadas , Receptores Depuradores de Clase E , Transferrina/metabolismo
20.
Circulation ; 108(1): 86-91, 2003 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-12821548

RESUMEN

BACKGROUND: Because bile salt-dependent lipase (BSDL), an enzyme secreted by the pancreatic acinar cells and associated with LDL in circulating blood, also locates with smooth muscle cells (SMCs) in atherosclerotic lesions, we aimed to investigate its effects on SMCs. METHODS AND RESULTS: Immunohistochemical experiments allowed us to detect an expression of BSDL in atherosclerotic lesions from hypercholesterolemic monkeys and from human arteries. BSDL was found to be associated with SMCs but not with macrophages. BSDL was significantly mitogenic for cultured SMCs. This effect was inhibited by heparin and anti-BSDL antibodies, whereas heat-denaturated and diisopropylfluorophosphate-treated BSDL were inefficient. The mitogenic effect of BSDL was associated with an activation of the extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway, which was inhibited by heparin, and involved several mechanisms, among them diacylglycerol and oleic acid production as well as a rapid basic fibroblast growth factor release. CONCLUSIONS: Circulating BSDL is associated with SMCs within the intimal arteria and may trigger SMC proliferation, which could contribute to the development of atherosclerotic lesions.


Asunto(s)
Arteriosclerosis/enzimología , Músculo Liso Vascular/efectos de los fármacos , Páncreas/enzimología , Esterol Esterasa/farmacología , Animales , Anticuerpos/farmacología , Aorta/enzimología , Aorta/patología , Arteriosclerosis/complicaciones , Arteriosclerosis/patología , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Hipercolesterolemia/complicaciones , Hipercolesterolemia/enzimología , Inmunohistoquímica , Isoflurofato/farmacología , Macaca fascicularis , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , ARN Mensajero/metabolismo , Conejos , Sistemas de Mensajero Secundario/efectos de los fármacos , Esterol Esterasa/antagonistas & inhibidores , Esterol Esterasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...