Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Psychiatry ; 95(2): 161-174, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-37704042

RESUMEN

BACKGROUND: 15q11.2 deletions and duplications have been linked to autism spectrum disorder, schizophrenia, and intellectual disability. Recent evidence suggests that dysfunctional CYFIP1 (cytoplasmic FMR1 interacting protein 1) contributes to the clinical phenotypes observed in individuals with 15q11.2 deletion/duplication syndrome. CYFIP1 plays crucial roles in neuronal development and brain connectivity, promoting actin polymerization and regulating local protein synthesis. However, information about the impact of single nucleotide variants in CYFIP1 on neurodevelopmental disorders is limited. METHODS: Here, we report a family with 2 probands exhibiting intellectual disability, autism spectrum disorder, spastic tetraparesis, and brain morphology defects and who carry biallelic missense point mutations in the CYFIP1 gene. We used skin fibroblasts from one of the probands, the parents, and typically developing individuals to investigate the effect of the variants on the functionality of CYFIP1. In addition, we generated Drosophila knockin mutants to address the effect of the variants in vivo and gain insight into the molecular mechanism that underlies the clinical phenotype. RESULTS: Our study revealed that the 2 missense variants are in protein domains responsible for maintaining the interaction within the wave regulatory complex. Molecular and cellular analyses in skin fibroblasts from one proband showed deficits in actin polymerization. The fly model for these mutations exhibited abnormal brain morphology and F-actin loss and recapitulated the core behavioral symptoms, such as deficits in social interaction and motor coordination. CONCLUSIONS: Our findings suggest that the 2 CYFIP1 variants contribute to the clinical phenotype in the probands that reflects deficits in actin-mediated brain development processes.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/genética , Actinas/genética , Actinas/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Polimerizacion , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
2.
Int J Mol Sci ; 24(6)2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36982451

RESUMEN

Cajal-Retzius cells (CRs) are a class of transient neurons in the mammalian cortex that play a critical role in cortical development. Neocortical CRs undergo almost complete elimination in the first two postnatal weeks in rodents and the persistence of CRs during postnatal life has been detected in pathological conditions related to epilepsy. However, it is unclear whether their persistence is a cause or consequence of these diseases. To decipher the molecular mechanisms involved in CR death, we investigated the contribution of the PI3K/AKT/mTOR pathway as it plays a critical role in cell survival. We first showed that this pathway is less active in CRs after birth before massive cell death. We also explored the spatio-temporal activation of both AKT and mTOR pathways and reveal area-specific differences along both the rostro-caudal and medio-lateral axes. Next, using genetic approaches to maintain an active pathway in CRs, we found that the removal of either PTEN or TSC1, two negative regulators of the pathway, lead to differential CR survivals, with a stronger effect in the Pten model. Persistent cells in this latter mutant are still active. They express more Reelin and their persistence is associated with an increase in the duration of kainate-induced seizures in females. Altogether, we show that the decrease in PI3K/AKT/mTOR activity in CRs primes these cells to death by possibly repressing a survival pathway, with the mTORC1 branch contributing less to the phenotype.


Asunto(s)
Ácido Kaínico , Proteínas Proto-Oncogénicas c-akt , Animales , Femenino , Ácido Kaínico/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Convulsiones/inducido químicamente , Mamíferos/metabolismo
3.
Biomedicines ; 10(12)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36551904

RESUMEN

Major progress has been made over the last decade in identifying novel genes involved in neurodevelopmental disorders, although the task of elucidating their corresponding molecular and pathophysiological mechanisms, which are an essential prerequisite for developing therapies, has fallen far behind. We selected 45 genes for intellectual disabilities to generate and characterize mouse models. Thirty-nine of them were based on the frequency of pathogenic variants in patients and literature reports, with several corresponding to de novo variants, and six other candidate genes. We used an extensive screen covering the development and adult stages, focusing specifically on behaviour and cognition to assess a wide range of functions and their pathologies, ranging from basic neurological reflexes to cognitive abilities. A heatmap of behaviour phenotypes was established, together with the results of selected mutants. Overall, three main classes of mutant lines were identified based on activity phenotypes, with which other motor or cognitive deficits were associated. These data showed the heterogeneity of phenotypes between mutation types, recapitulating several human features, and emphasizing the importance of such systematic approaches for both deciphering genetic etiological causes of ID and autism spectrum disorders, and for building appropriate therapeutic strategies.

4.
Cells ; 11(9)2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35563851

RESUMEN

Oligophrenin-1 (OPHN1) is a Rho-GTPase-activating protein (RhoGAP), whose mutations are associated with X-linked intellectual disability (XLID). OPHN1 is enriched at the synapse in both pre- and postsynaptic compartments, where it regulates the RhoA/ROCK/MLC2 signaling pathway, playing a critical role in cytoskeleton remodeling and vesicle recycling. Ophn1 knockout (KO) adult mice display some behavioral deficits in multiple tasks, reminiscent of some symptoms in the human pathology. We also previously reported a reduction in dendritic spine density in the adult hippocampus of KO mice. Yet the nature of the deficits occurring in these mice during postnatal development remains elusive. Here, we show that juvenile KO mice present normal basal synaptic transmission, but altered synaptic plasticity, with a selective impairment in long-term depression, but no change in long-term potentiation. This contrasts with the functional deficits that these mice display at the adult stage, as we found that both basal synaptic transmission and long-term potentiation are reduced at later stages, due to presynaptic alterations. In addition, the number of excitatory synapses in adult is increased, suggesting some unsuccessful compensation. Altogether, these results suggest that OPHN1 function at synapses is differentially affected during maturation of the brain, which provides some therapeutic opportunities for early intervention.


Asunto(s)
Proteínas del Citoesqueleto , Proteínas Activadoras de GTPasa , Hipocampo , Discapacidad Intelectual , Transmisión Sináptica , Animales , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Hipocampo/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
5.
Science ; 373(6550): 77-81, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34210880

RESUMEN

Brain postnatal development is characterized by critical periods of experience-dependent remodeling of neuronal circuits. Failure to end these periods results in neurodevelopmental disorders. The cellular processes defining critical-period timing remain unclear. Here, we show that in the mouse visual cortex, astrocytes control critical-period closure. We uncover the underlying pathway, which involves astrocytic regulation of the extracellular matrix, allowing interneuron maturation. Unconventional astrocyte connexin signaling hinders expression of extracellular matrix-degrading enzyme matrix metalloproteinase 9 (MMP9) through RhoA-guanosine triphosphatase activation. Thus, astrocytes not only influence the activity of single synapses but also are key elements in the experience-dependent wiring of brain circuits.


Asunto(s)
Astrocitos/fisiología , Período Crítico Psicológico , Plasticidad Neuronal , Corteza Visual/crecimiento & desarrollo , Animales , Astrocitos/metabolismo , Conexina 30/metabolismo , Activación Enzimática , GTP Fosfohidrolasas/metabolismo , Interneuronas/metabolismo , Interneuronas/fisiología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sinapsis/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
7.
Nucleic Acids Res ; 48(17): 9804-9821, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32816001

RESUMEN

CSTF2 encodes an RNA-binding protein that is essential for mRNA cleavage and polyadenylation (C/P). No disease-associated mutations have been described for this gene. Here, we report a mutation in the RNA recognition motif (RRM) of CSTF2 that changes an aspartic acid at position 50 to alanine (p.D50A), resulting in intellectual disability in male patients. In mice, this mutation was sufficient to alter polyadenylation sites in over 1300 genes critical for brain development. Using a reporter gene assay, we demonstrated that C/P efficiency of CSTF2D50A was lower than wild type. To account for this, we determined that p.D50A changed locations of amino acid side chains altering RNA binding sites in the RRM. The changes modified the electrostatic potential of the RRM leading to a greater affinity for RNA. These results highlight the significance of 3' end mRNA processing in expression of genes important for brain plasticity and neuronal development.


Asunto(s)
Factor de Estimulación del Desdoblamiento/genética , Discapacidad Intelectual/genética , Mutación Missense , Poliadenilación , Motivo de Reconocimiento de ARN , Regiones no Traducidas 3' , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Niño , Factor de Estimulación del Desdoblamiento/química , Factor de Estimulación del Desdoblamiento/metabolismo , Femenino , Células HeLa , Humanos , Discapacidad Intelectual/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Linaje , Unión Proteica
8.
Glia ; 68(9): 1729-1742, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32073702

RESUMEN

Astrocytes are involved in several aspects of neuronal development and properties which are altered in intellectual disability (ID). Oligophrenin-1 is a RhoGAP protein implicated in actin cytoskeleton regulation, and whose mutations are associated with X-linked ID. Oligophrenin-1 is expressed in neurons, where its functions have been widely reported at the synapse, as well as in glial cells. However, its roles in astrocytes are still largely unexplored. Using in vitro and in vivo models of oligophrenin1 disruption in astrocytes, we found that oligophrenin1 regulates at the molecular level the RhoA/ROCK/MLC2 pathway in astroglial cells. We also showed at the cellular level that oligophrenin1 modulates astrocyte morphology and migration both in vitro and in vivo, and is involved in glial scar formation. Altogether, these data suggest that oligophrenin1 deficiency alters not only neuronal but also astrocytic functions, which might contribute to the development of ID.


Asunto(s)
Astrocitos , Discapacidad Intelectual , Proteínas del Citoesqueleto/genética , Humanos , Discapacidad Intelectual/genética , Neuroglía , Neuronas
9.
J Neurosci ; 40(13): 2776-2788, 2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32098904

RESUMEN

Oligophrenin-1 (Ophn1) encodes a Rho GTPase activating protein whose mutations cause X-linked intellectual disability (XLID) in humans. Loss of function of Ophn1 leads to impairments in the maturation and function of excitatory and inhibitory synapses, causing deficits in synaptic structure, function and plasticity. Epilepsy is a frequent comorbidity in patients with Ophn1-dependent XLID, but the cellular bases of hyperexcitability are poorly understood. Here we report that male mice knock-out (KO) for Ophn1 display hippocampal epileptiform alterations, which are associated with changes in parvalbumin-, somatostatin- and neuropeptide Y-positive interneurons. Because loss of function of Ophn1 is related to enhanced activity of Rho-associated protein kinase (ROCK) and protein kinase A (PKA), we attempted to rescue Ophn1-dependent pathological phenotypes by treatment with the ROCK/PKA inhibitor fasudil. While acute administration of fasudil had no impact on seizure activity, seven weeks of treatment in adulthood were able to correct electrographic, neuroanatomical and synaptic alterations of Ophn1 deficient mice. These data demonstrate that hyperexcitability and the associated changes in GABAergic markers can be rescued at the adult stage in Ophn1-dependent XLID through ROCK/PKA inhibition.SIGNIFICANCE STATEMENT In this study we demonstrate enhanced seizure propensity and impairments in hippocampal GABAergic circuitry in Ophn1 mouse model of X-linked intellectual disability (XLID). Importantly, the enhanced susceptibility to seizures, accompanied by an alteration of GABAergic markers were rescued by Rho-associated protein kinase (ROCK)/protein kinase A (PKA) inhibitor fasudil, a drug already tested on humans. Because seizures can significantly impact the quality of life of XLID patients, the present data suggest a potential therapeutic pathway to correct alterations in GABAergic networks and dampen pathological hyperexcitability in adults with XLID.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Neuronas GABAérgicas/efectos de los fármacos , Proteínas Activadoras de GTPasa/genética , Hipocampo/efectos de los fármacos , Discapacidad Intelectual/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Convulsiones/fisiopatología , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Neuronas GABAérgicas/fisiología , Hipocampo/fisiopatología , Discapacidad Intelectual/genética , Ratones , Ratones Noqueados , Convulsiones/genética
10.
Trends Neurosci ; 42(8): 518-527, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31300246

RESUMEN

Neurodevelopmental disorders, including those involving intellectual disability, are characterized by abnormalities in formation and functions of synaptic circuits. Traditionally, research on synaptogenesis and synaptic transmission in health and disease focused on neurons, however, a growing number of studies have highlighted the role of astrocytes in this context. Tight structural and functional interactions of astrocytes and synapses indeed play important roles in brain functions, and the repertoire of astroglial regulations of synaptic circuits is large and complex. Recently, genetic studies of intellectual disabilities have underscored potential contributions of astrocytes in the pathophysiology of these disorders. Here we review how alterations of astrocyte functions in disease may interfere with neuronal excitability and the balance of excitatory and inhibitory transmission during development, and contribute to intellectual disabilities.


Asunto(s)
Astrocitos/patología , Astrocitos/fisiología , Encéfalo/fisiopatología , Discapacidad Intelectual/fisiopatología , Animales , Humanos
11.
Cell Rep ; 27(3): 835-846.e5, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995480

RESUMEN

Interleukin-38 (IL-38) is a cytokine of the IL-1 family with a role in chronic inflammation. However, its main cellular targets and receptors remain obscure. IL-38 is highly expressed in the skin and downregulated in psoriasis patients. We report an investigation in cellular targets of IL-38 during the progression of imiquimod-induced psoriasis. In this model, IL-38 knockout (IL-38 KO) mice show delayed disease resolution with exacerbated IL-17-mediated inflammation, which is reversed by the administration of mature IL-38 or γδ T cell-receptor-blocking antibodies. Mechanistically, X-linked IL-1 receptor accessory protein-like 1 (IL1RAPL1) is upregulated upon γδ T cell activation to feedforward-amplify IL-17 production and is required for IL-38 to suppress γδ T cell IL-17 production. Accordingly, psoriatic IL1RAPL1 KO mice show reduced inflammation and IL-17 production by γδ T cells. Our findings indicate a role for IL-38 in the regulation of γδ T cell activation through IL1RAPL1, with consequences for auto-inflammatory disease.


Asunto(s)
Interleucina-17/metabolismo , Interleucina-1/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Piel/patología , Linfocitos T/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Diferenciación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Imiquimod/toxicidad , Inflamación/prevención & control , Interleucina-1/genética , Interleucina-1/farmacología , Proteína Accesoria del Receptor de Interleucina-1/deficiencia , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/inducido químicamente , Psoriasis/patología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Regeneración/efectos de los fármacos , Piel/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología
12.
Gene ; 679: 305-313, 2018 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-30217758

RESUMEN

Histone lysine methylation influences processes such as gene expression and DNA repair. Thirty Jumonji C (JmjC) domain-containing proteins have been identified and phylogenetically clustered into seven subfamilies. Most JmjC domain-containing proteins have been shown to possess histone demethylase activity toward specific histone methylation marks. One of these subfamilies, the KDM5 family, is characterized by five conserved domains and includes four members. Interestingly, de novo loss-of-function and missense variants in KDM5B were identified in patients with intellectual disability (ID) and autism spectrum disorder (ASD) but also in unaffected individuals. Here, we report two novel de novo splice variants in the KDM5B gene in three patients with ID and ASD. The c.808 + 1G > A variant was identified in a boy with mild ID and autism traits and is associated with a significant reduced KDM5B mRNA expression without alteration of its H3K4me3 pattern. In contrast, the c.576 + 2T > C variant was found in twins with global delay in developmental milestones, poor language and ASD. This variant causes the production of an abnormal transcript which may produce an altered protein with the loss of the ARID1B domain with an increase in global gene H3K4me3. Our data reinforces the recent observation that the KDM5B haploinsufficiency is not a mechanism involved in intellectual disability and that KDM5B disorder associated with LOF variants is a recessive disorder. However, some variants may also cause gain of function, and need to be interpreted with caution, and functional studies should be performed to identify the molecular consequences of these different rare variants.


Asunto(s)
Empalme Alternativo , Trastorno del Espectro Autista/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Adolescente , Trastorno del Espectro Autista/metabolismo , Línea Celular , Niño , Discapacidades del Desarrollo/metabolismo , Regulación hacia Abajo , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Humanos , Discapacidad Intelectual/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Proteínas Nucleares/metabolismo , Linaje , Proteínas Represoras/metabolismo
13.
Neuromolecular Med ; 20(3): 409-417, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30014449

RESUMEN

A growing number of histone modifiers are involved in human neurodevelopmental disorders, suggesting that proper regulation of chromatin state is essential for the development of the central nervous system. Among them, heterozygous de novo variants in KMT2A, a gene coding for histone methyltransferase, have been associated with Wiedemann-Steiner syndrome (WSS), a rare developmental disorder mainly characterized by intellectual disability (ID) and hypertrichosis. As KMT2A is known to regulate the expression of multiple target genes through methylation of lysine 4 of histone 3 (H3K4me), we sought to investigate the transcriptomic consequences of KMT2A variants involved in WSS. Using fibroblasts from four WSS patients harboring loss-of-function KMT2A variants, we performed RNA sequencing and identified a number of genes for which transcription was altered in KMT2A-mutated cells compared to the control ones. Strikingly, analysis of the pathways and biological functions significantly deregulated between patients with WSS and healthy individuals revealed a number of processes predicted to be altered that are relevant for hypertrichosis and intellectual disability, the cardinal signs of this disease.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Hipertricosis/genética , Hipertricosis/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Enfermedades Raras/genética , Enfermedades Raras/metabolismo , Adolescente , Adulto , Niño , Femenino , Fibroblastos , Regulación de la Expresión Génica , Humanos , Mutación con Pérdida de Función , Lisina/metabolismo , Masculino , Metilación , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Síndrome , Transcripción Genética , Adulto Joven
14.
Eur J Hum Genet ; 26(1): 107-116, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29203834

RESUMEN

Variants in KMT2A, encoding the histone methyltransferase KMT2A, are a growing cause of intellectual disability (ID). Up to now, the majority of KMT2A variants are non-sense and frameshift variants causing a typical form of Wiedemann-Steiner syndrome. We studied KMT2A gene in a cohort of 200 patients with unexplained syndromic and non-syndromic ID and identified four novel variants, one splice and three missense variants, possibly deleterious. We used primary cells from the patients and molecular approaches to determine the deleterious effects of those variants on KMT2A expression and function. For the putative splice variant c.11322-1G>A, we showed that it led to only one nucleotide deletion and loss of the C-terminal part of the protein. For two studied KMT2A missense variants, c.3460C>T (p.(Arg1154Trp)) and c.8558T>G (p.(Met2853Arg)), located at the cysteine-rich CXXC domain and the transactivation domain of the protein, respectively, we found altered KMT2A target genes expression in patient's fibroblasts compared to controls. Furthermore, we found a disturbed subcellular distribution of KMT2A for the c.3460C>T mutant. Taken together, our results demonstrated the deleterious impact of the splice variant and of the missense variants located at two different functional domains and suggested reduction of KMT2A function as the disease-causing mechanism.


Asunto(s)
Anomalías Múltiples/genética , N-Metiltransferasa de Histona-Lisina/genética , Hipertricosis/genética , Discapacidad Intelectual/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Anomalías Múltiples/patología , Animales , Células COS , Células Cultivadas , Preescolar , Chlorocebus aethiops , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hipertricosis/patología , Lactante , Discapacidad Intelectual/patología , Mutación con Pérdida de Función , Masculino , Mutación Missense , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Empalme del ARN , Síndrome
15.
J Neurosci ; 37(46): 11114-11126, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29030432

RESUMEN

Classical and systems genetics have identified wide networks of genes associated with cognitive and neurodevelopmental diseases. In parallel to deciphering the role of each of these genes in neuronal or synaptic function, evaluating the response of neuronal and molecular networks to gene loss of function could reveal some pathophysiological mechanisms potentially accessible to nongenetic therapies. Loss of function of the Rho-GAP oligophrenin-1 is associated with cognitive impairments in both human and mouse. Upregulation of both PKA and ROCK has been reported in Ophn1-/y mice, but it remains unclear whether kinase hyperactivity contributes to the behavioral phenotypes. In this study, we thoroughly characterized a prominent perseveration phenotype displayed by Ophn1-deficient mice using a Y-maze spatial working memory (SWM) test. We report that Ophn1 deficiency in the mouse generated severe cognitive impairments, characterized by both a high occurrence of perseverative behaviors and a lack of deliberation during the SWM test. In vivo and in vitro pharmacological experiments suggest that PKA dysregulation in the mPFC underlies cognitive dysfunction in Ophn1-deficient mice, as assessed using a delayed spatial alternation task results. Functionally, mPFC neuronal networks appeared to be affected in a PKA-dependent manner, whereas hippocampal-PFC projections involved in SWM were not affected in Ophn1-/y mice. Thus, we propose that discrete gene mutations in intellectual disability might generate "secondary" pathophysiological mechanisms, which are prone to become pharmacological targets for curative strategies in adult patients.SIGNIFICANCE STATEMENT Here we report that Ophn1 deficiency generates severe impairments in performance at spatial working memory tests, characterized by a high occurrence of perseverative behaviors and a lack of decision making. This cognitive deficit is consecutive to PKA deregulation in the mPFC that prevents Ophn1 KO mice to exploit a correctly acquired rule. Functionally, mPFC neuronal networks appear to be affected in a PKA-dependent manner, whereas behaviorally important hippocampal projections were preserved by the mutation. Thus, we propose that discrete gene mutations in intellectual disability can generate "secondary" pathophysiological mechanisms prone to become pharmacological targets for curative strategies in adults.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/deficiencia , Proteínas Activadoras de GTPasa/deficiencia , Trastornos de la Memoria/metabolismo , Memoria a Corto Plazo/fisiología , Proteínas Nucleares/deficiencia , Corteza Prefrontal/metabolismo , Animales , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Técnicas de Cultivo de Órganos , Corteza Prefrontal/fisiopatología , Distribución Aleatoria
16.
J Neurosci ; 37(28): 6606-6627, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28576939

RESUMEN

Mutations and deletions of the interleukin-1 receptor accessory protein like 1 (IL1RAPL1) gene, located on the X chromosome, are associated with intellectual disability (ID) and autism spectrum disorder (ASD). IL1RAPL1 protein is located at the postsynaptic compartment of excitatory synapses and plays a role in synapse formation and stabilization. Here, using primary neuronal cultures and Il1rapl1-KO mice, we characterized the role of IL1RAPL1 in regulating dendrite morphology. In Il1rapl1-KO mice we identified an increased number of dendrite branching points in CA1 and CA2 hippocampal neurons associated to hippocampal cognitive impairment. Similarly, induced pluripotent stem cell-derived neurons from a patient carrying a null mutation of the IL1RAPL1 gene had more dendrites. In hippocampal neurons, the overexpression of full-length IL1RAPL1 and mutants lacking part of C-terminal domains leads to simplified neuronal arborization. This effect is abolished when we overexpressed mutants lacking part of N-terminal domains, indicating that the IL1RAPL1 extracellular domain is required for regulating dendrite development. We also demonstrate that PTPδ interaction is not required for this activity, while IL1RAPL1 mediates the activity of IL-1ß on dendrite morphology. Our data reveal a novel specific function for IL1RAPL1 in regulating dendrite morphology that can help clarify how changes in IL1RAPL1-regulated pathways can lead to cognitive disorders in humans.SIGNIFICANCE STATEMENT Abnormalities in the architecture of dendrites have been observed in a variety of neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Here we show that the X-linked intellectual disability protein interleukin-1 receptor accessory protein like 1 (IL1RAPL1) regulates dendrite morphology of mice hippocampal neurons and induced pluripotent stem cell-derived neurons from a patient carrying a null mutation of IL1RAPL1 gene. We also found that the extracellular domain of IL1RAPL1 is required for this effect, independently of the interaction with PTPδ, but IL1RAPL1 mediates the activity of IL-1ß on dendrite morphology. Our data reveal a novel specific function for IL1RAPL1 in regulating dendrite morphology that can help clarify how changes in IL1RAPL1-regulated pathways can lead to cognitive disorders in humans.


Asunto(s)
Dendritas/metabolismo , Dendritas/patología , Genes Ligados a X/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Proteína Accesoria del Receptor de Interleucina-1/genética , Animales , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Femenino , Hipocampo/patología , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley
17.
Hum Mutat ; 38(8): 932-941, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28585349

RESUMEN

De novo mutations are a frequent cause of disorders related to brain development. We report the results from the screening of two patients diagnosed with intellectual disability (ID) using exome sequencing to identify new causative de novo mutations. Exome sequencing was conducted in two patient-parent trios to identify de novo variants. In silico and expression studies were also performed to evaluate the functional consequences of these variants. The two patients presented developmental delay with minor facial dysmorphy. One of them presented pharmacoresistant myoclonic epilepsy. We identified two de novo splice variants (c.175+2T>G; c.367+2T>C) in the CSNK2B gene encoding the ß subunit of the Caseine kinase 2 (CK2). CK2 is a ubiquitously expressed kinase that is present in high levels in brain and it appears to be constitutively active. The mRNA transcripts were abnormal and significantly reduced in affected fibroblasts and most likely produced truncated proteins. Taking into account that mutations in CSNK2A1, encoding the α subunit of CK2, were previously identified in patients with neurodevelopmental disorders and dysmorphic features, our study confirmed that the protein kinase CK2 plays a major role in brain, and showed that CSNK2, encoding the ß subunit, is a novel ID gene. This study adds knowledge to the increasingly growing list of causative and candidate genes in ID and epilepsy, and highlights CSNK2B as a new gene for neurodevelopmental disorders.


Asunto(s)
Quinasa de la Caseína II/genética , Discapacidades del Desarrollo/genética , Epilepsias Mioclónicas/genética , Discapacidad Intelectual/genética , Quinasa de la Caseína II/metabolismo , Preescolar , Hibridación Genómica Comparativa , Exoma/genética , Femenino , Humanos , Lactante , Masculino , Mutación/genética , Trastornos del Neurodesarrollo/genética , Secuenciación del Exoma/métodos
18.
Eur J Hum Genet ; 25(5): 560-564, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28145425

RESUMEN

p.(Arg320His) mutation in the KCNC1 gene in human 11p15.1 has recently been identified in patients with progressive myoclonus epilepsies, a group of rare inherited disorders manifesting with action myoclonus, myoclonic epilepsy, and ataxia. This KCNC1 variant causes a dominant-negative effect. Here we describe three patients from the same family with intellectual disability and dysmorphic features. The three affected individuals carry a c.1015C>T (p.(Arg339*)) nonsense variant in KCNC1 gene. As previously observed in the mutant mouse carrying a disrupted KCNC1 gene, these findings reveal that individuals with a KCNC1 loss-of-function variant can present intellectual disability without seizure and epilepsy.


Asunto(s)
Codón sin Sentido , Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Convulsiones/genética , Canales de Potasio Shaw/genética , Adulto , Niño , Anomalías Craneofaciales/diagnóstico , Femenino , Humanos , Discapacidad Intelectual/diagnóstico , Masculino , Linaje , Convulsiones/diagnóstico , Síndrome
19.
Neurobiol Dis ; 100: 75-86, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28088401

RESUMEN

Oligophrenin-1 (OPHN1) is a Rho GTPase activating protein whose mutations cause X-linked intellectual disability (XLID). How loss of function of Ophn1 affects neuronal development is only partly understood. Here we have exploited adult hippocampal neurogenesis to dissect the steps of neuronal differentiation that are affected by Ophn1 deletion. We found that mice lacking Ophn1 display a reduction in the number of newborn neurons in the dentate gyrus. A significant fraction of the Ophn1-deficient newly generated neurons failed to extend an axon towards CA3, and showed an altered density of dendritic protrusions. Since Ophn1-deficient mice display overactivation of Rho-associated protein kinase (ROCK) and protein kinase A (PKA) signaling, we administered a clinically approved ROCK/PKA inhibitor (fasudil) to correct the neurogenesis defects. While administration of fasudil was not effective in rescuing axon formation, the same treatment completely restored spine density to control levels, and enhanced the long-term survival of adult-born neurons in mice lacking Ophn1. These results identify specific neurodevelopmental steps that are impacted by Ophn1 deletion, and indicate that they may be at least partially corrected by pharmacological treatment.


Asunto(s)
Hipocampo/metabolismo , Discapacidad Intelectual/fisiopatología , Neurogénesis/fisiología , Neuronas/metabolismo , Animales , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo
20.
Hum Mol Genet ; 25(23): 5198-5211, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27742778

RESUMEN

Among the X-linked genes associated with intellectual disability, Oligophrenin-1 (OPHN1) encodes for a Rho GTPase-activating protein, a key regulator of several developmental processes, such as dendrite and spine formation and synaptic activity. Inhibitory interneurons play a key role in the development and function of neuronal circuits. Whether a mutation of OPHN1 can affect morphology and synaptic properties of inhibitory interneurons remains poorly understood. To address these open questions, we studied in a well-established mouse model of X-linked intellectual disability, i.e. a line of mice carrying a null mutation of OPHN1, the development and function of adult generated inhibitory interneurons in the olfactory bulb. Combining quantitative morphological analysis and electrophysiological recordings we found that the adult generated inhibitory interneurons were dramatically reduced in number and exhibited a higher proportion of filopodia-like spines, with the consequences on their synaptic function, in OPHN1 ko mice. Furthermore, we found that olfactory behaviour was perturbed in OPHN1 ko mice. Chronic treatment with a Rho kinase inhibitor rescued most of the defects of the newly generated neurons. Altogether, our data indicated that OPHN1 plays a key role in regulating the number, morphology and function of adult-born inhibitory interneurons and contributed to identify potential therapeutic targets.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas Activadoras de GTPasa/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Discapacidad Intelectual/genética , Proteínas Nucleares/genética , Animales , Dendritas/efectos de los fármacos , Dendritas/genética , Dendritas/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/patología , Interneuronas/efectos de los fármacos , Interneuronas/patología , Ratones Noqueados , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/patología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...