Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Psychiatry ; 13: 924956, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36405918

RESUMEN

16p13.11 copy number variants (CNVs) have been associated with autism, schizophrenia, psychosis, intellectual disability, and epilepsy. The majority of 16p13.11 deletions or duplications occur within three well-defined intervals, and despite growing knowledge of the functions of individual genes within these intervals, the molecular mechanisms that underlie commonly observed clinical phenotypes remain largely unknown. Patient-derived, induced pluripotent stem cells (iPSCs) provide a platform for investigating the morphological, electrophysiological, and gene-expression changes that result from 16p13.11 CNVs in human-derived neurons. Patient derived iPSCs with varying sizes of 16p13.11 deletions and familial controls were differentiated into cortical neurons for phenotypic analysis. High-content imaging and morphological analysis of patient-derived neurons demonstrated an increase in neurite branching in patients compared with controls. Whole-transcriptome sequencing revealed expression level changes in neuron development and synaptic-related gene families, suggesting a defect in synapse formation. Subsequent quantification of synapse number demonstrated increased numbers of synapses on neurons derived from early-onset patients compared to controls. The identification of common phenotypes among neurons derived from patients with overlapping 16p13.11 deletions will further assist in ascertaining common pathways and targets that could be utilized for screening drug candidates. These studies can help to improve future treatment options and clinical outcomes for 16p13.11 deletion patients.

2.
Mol Ther ; 30(7): 2416-2428, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35585789

RESUMEN

We are in an emerging era of gene-based therapeutics with significant promise for rare genetic disorders. The potential is particularly significant for genetic central nervous system disorders that have begun to achieve Food and Drug Administration approval for select patient populations. This review summarizes the discussions and presentations of the National Institute of Mental Health-sponsored workshop "Gene-Based Therapeutics for Rare Genetic Neurodevelopmental Psychiatric Disorders," which was held in January 2021. Here, we distill the points raised regarding various precision medicine approaches related to neurodevelopmental and psychiatric disorders that may be amenable to gene-based therapies.


Asunto(s)
Trastornos Mentales , Medicina de Precisión , Humanos , Trastornos Mentales/genética , Trastornos Mentales/psicología , Trastornos Mentales/terapia , Enfermedades Raras , Estados Unidos , United States Food and Drug Administration
3.
Arterioscler Thromb Vasc Biol ; 42(1): e27-e43, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34670408

RESUMEN

OBJECTIVE: Capillary malformation (CM) occurs sporadically and is associated with Sturge-Weber syndrome. The somatic mosaic mutation in GNAQ (c.548G>A, p.R183Q) is enriched in endothelial cells (ECs) in skin CM and Sturge-Weber syndrome brain CM. Our goal was to investigate how the mutant Gαq (G-protein αq subunit) alters EC signaling and disrupts capillary morphogenesis. Approach and Results: We used lentiviral constructs to express p.R183Q or wild-type GNAQ in normal human endothelial colony forming cells (EC-R183Q and EC-WT, respectively). EC-R183Q constitutively activated PLC (phospholipase C) ß3, a downstream effector of Gαq. Activated PLCß3 was also detected in human CM tissue sections. Bulk RNA sequencing analyses of mutant versus wild-type EC indicated constitutive activation of PKC (protein kinase C), NF-κB (nuclear factor kappa B) and calcineurin signaling in EC-R183Q. Increased expression of downstream targets in these pathways, ANGPT2 (angiopoietin-2) and DSCR (Down syndrome critical region protein) 1.4 were confirmed by quantitative PCR and immunostaining of human CM tissue sections. The Gαq inhibitor YM-254890 as well as siRNA targeted to PLCß3 reduced mRNA expression levels of these targets in EC-R183Q while the pan-PKC inhibitor AEB071 reduced ANGPT2 but not DSCR1.4. EC-R183Q formed enlarged blood vessels in mice, reminiscent of those found in human CM. shRNA knockdown of ANGPT2 in EC-R183Q normalized the enlarged vessels to sizes comparable those formed by EC-WT. CONCLUSIONS: Gαq-R183Q, when expressed in ECs, establishes constitutively active PLCß3 signaling that leads to increased ANGPT2 and a proangiogenic, proinflammatory phenotype. EC-R183Q are sufficient to form enlarged CM-like vessels in mice, and suppression of ANGPT2 prevents the enlargement. Our study provides the first evidence that endothelial Gαq-R183Q is causative for CM and identifies ANGPT2 as a contributor to CM vascular phenotype.


Asunto(s)
Angiopoyetina 2/metabolismo , Capilares/metabolismo , Células Progenitoras Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Neovascularización Patológica , Síndrome de Sturge-Weber/metabolismo , Adolescente , Adulto , Anciano , Angiopoyetina 2/genética , Animales , Capilares/anomalías , Células Cultivadas , Niño , Preescolar , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Lactante , Recién Nacido , Masculino , Ratones Desnudos , Mutación , Fenotipo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Síndrome de Sturge-Weber/genética , Síndrome de Sturge-Weber/patología , Regulación hacia Arriba
4.
Cell Stem Cell ; 28(9): 1507-1515, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34478628

RESUMEN

Over the course of the last decade, the biopharmaceutical industry has slowly adopted human inducible pluripotent stem cell (hiPSC) technology to enable the development of humanized model systems to test new therapeutic molecules and drug modalities. The adoption of hiPSC-based models by the industry has increased appreciably in the past 3-5 years. This increase has paralleled the explosion in availability of high-quality human genetic data to mine for new drug targets and the emergence of human-specific therapeutic modalities.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Descubrimiento de Drogas , Humanos , Modelos Biológicos , Miocitos Cardíacos
5.
Cell Rep ; 31(12): 107780, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579942

RESUMEN

Tuberous sclerosis complex (TSC) is a neurogenetic disorder that leads to elevated mechanistic targeting of rapamycin complex 1 (mTORC1) activity. Cilia can be affected by mTORC1 signaling, and ciliary deficits are associated with neurodevelopmental disorders. Here, we examine whether neuronal cilia are affected in TSC. We show that cortical tubers from TSC patients and mutant mouse brains have fewer cilia. Using high-content image-based assays, we demonstrate that mTORC1 activity inversely correlates with ciliation in TSC1/2-deficient neurons. To investigate the mechanistic relationship between mTORC1 and cilia, we perform a phenotypic screen for mTORC1 inhibitors with TSC1/2-deficient neurons. We identify inhibitors of the heat shock protein 90 (Hsp90) that suppress mTORC1 through regulation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling. Pharmacological inhibition of Hsp90 rescues ciliation through downregulation of Hsp27. Our study uncovers the heat-shock machinery as a druggable signaling node to restore mTORC1 activity and cilia due to loss of TSC1/2, and it provides broadly applicable platforms for studying TSC-related neuronal dysfunction.


Asunto(s)
Cilios/metabolismo , Respuesta al Choque Térmico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neuronas/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Envejecimiento/metabolismo , Animales , Benzoquinonas/farmacología , Encéfalo/patología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Lactamas Macrocíclicas/farmacología , Ratones Noqueados , Neuronas/efectos de los fármacos , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Sirolimus/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
6.
Mol Biol Cell ; 30(22): 2741-2743, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31609671

RESUMEN

Discovery and development of new medicines requires the talent and passion of both academic and industrial scientists. Identifying the optimal set of circumstances for direct collaboration between academic and industry teams requires a mutual understanding of what each partner brings to the relationship, and an appreciation of the specialized capabilities and scope of work to be undertaken by each group. We provide our perspective on the who, what, where, why, and how for establishing therapeutic and translational research collaborations between academic and industry scientists.


Asunto(s)
Desarrollo de Medicamentos/métodos , Desarrollo de Medicamentos/tendencias , Academias e Institutos/tendencias , Conducta Cooperativa , Industria Farmacéutica/tendencias , Humanos , Investigación Biomédica Traslacional/tendencias
7.
Cell Rep ; 28(12): 3224-3237.e5, 2019 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-31533043

RESUMEN

Dysregulated axonal trafficking of mitochondria is linked to neurodegenerative disorders. We report a high-content screen for small-molecule regulators of the axonal transport of mitochondria. Six compounds enhanced mitochondrial transport in the sub-micromolar range, acting via three cellular targets: F-actin, Tripeptidyl peptidase 1 (TPP1), or Aurora Kinase B (AurKB). Pharmacological inhibition or small hairpin RNA (shRNA) knockdown of each target promotes mitochondrial axonal transport in rat hippocampal neurons and induced pluripotent stem cell (iPSC)-derived human cortical neurons and enhances mitochondrial transport in iPSC-derived motor neurons from an amyotrophic lateral sclerosis (ALS) patient bearing one copy of SOD1A4V mutation. Our work identifies druggable regulators of axonal transport of mitochondria, provides broadly applicable methods for similar image-based screens, and suggests that restoration of proper axonal trafficking of mitochondria can be achieved in human ALS neurons.


Asunto(s)
Aminopeptidasas/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Aurora Quinasa B/metabolismo , Axones/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Hipocampo/metabolismo , Mitocondrias/metabolismo , Serina Proteasas/metabolismo , Aminopeptidasas/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Aurora Quinasa B/genética , Axones/patología , Transporte Biológico Activo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Femenino , Células HEK293 , Hipocampo/patología , Humanos , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/patología , Ratas , Ratas Sprague-Dawley , Serina Proteasas/genética , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Tripeptidil Peptidasa 1
8.
Transl Psychiatry ; 9(1): 151, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31123247

RESUMEN

Psychiatric disorders such as schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD) arise from complex interactions between genetic and environmental factors. Common genetic variants associated with multiple psychiatric disorders suggest that shared genetic architecture could contribute to divergent clinical syndromes. To evaluate shared transcriptional alterations across connected brain regions, Affymetrix microarrays were used to profile postmortem dorsolateral prefrontal cortex (DLPFC), hippocampus, and associative striatum from 19 well-matched tetrads of subjects with SCZ, BD, MDD, or unaffected controls. SCZ subjects showed a substantial burden of differentially expressed genes across all examined brain regions with the greatest effects in hippocampus, whereas BD and MDD showed less robust alterations. Pathway analysis of transcriptional profiles compared across diagnoses demonstrated commonly enriched pathways between all three disorders in hippocampus, significant overlap between SCZ and BD in DLPFC, but no significant overlap of enriched pathways between disorders in striatum. SCZ samples showed increased expression of transcripts associated with inflammation across all brain regions examined, which was not evident in BD or MDD, or in rat brain following chronic dosing with antipsychotic drugs. Several markers of inflammation were confirmed by RT-PCR in hippocampus, including S100A8/9, IL-6, MAFF, APOLD1, IFITM3, and BAG3. A cytokine ELISA panel showed significant increases in IL-2 and IL-12p70 protein content in hippocampal tissue collected from same SCZ subjects when compared to matched control subjects. These data suggest an overlapping subset of dysregulated pathways across psychiatric disorders; however, a widespread increase in inflammation appears to be a specific feature of the SCZ brain and is not likely to be attributable to chronic antipsychotic drug treatment.


Asunto(s)
Trastorno Bipolar , Cuerpo Estriado , Trastorno Depresivo Mayor , Perfilación de la Expresión Génica , Hipocampo , Inflamación , Corteza Prefrontal , Esquizofrenia , Animales , Autopsia , Trastorno Bipolar/genética , Trastorno Bipolar/inmunología , Trastorno Bipolar/metabolismo , Cuerpo Estriado/inmunología , Cuerpo Estriado/metabolismo , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/inmunología , Trastorno Depresivo Mayor/metabolismo , Hipocampo/inmunología , Hipocampo/metabolismo , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Corteza Prefrontal/inmunología , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/genética , Esquizofrenia/inmunología , Esquizofrenia/metabolismo
9.
Neuron ; 100(4): 783-797, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30465765

RESUMEN

From the beginning, induced pluripotent stem cell (iPSC) technology was touted as a path to improve our understanding of disease biology and enable drug discovery. Advances in iPSC culture, genome engineering, and differentiation protocols have rapidly expanded the use of iPSC-derived disease models from the specialized work of stem cell biology into the mainstream toolkit of cellular neuroscience. Here we provide guidance for using iPSC-derived neurons for disease modeling with a focus on enabling screening platforms amenable to therapeutic drug discovery. We also highlight the potential for incorporating three-dimensional systems that may create more translational in vitro models.


Asunto(s)
Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas/fisiología , Enfermedades Neurodegenerativas/patología , Neuronas/fisiología , Investigación Biomédica Traslacional/métodos , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Descubrimiento de Drogas/tendencias , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/genética , Neuronas/efectos de los fármacos , Investigación Biomédica Traslacional/tendencias
10.
Mol Genet Metab Rep ; 16: 23-29, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29922587

RESUMEN

Complex phenotypes may represent novel syndromes that are the composite interaction of several genetic and environmental factors. We describe an 9-year old male with high functioning autism spectrum disorder and Muckle-Wells syndrome who at age 5  years of age manifested perseverations that interfered with his functioning at home and at school. After age 6, he developed intermittent episodes of fatigue and somnolence lasting from hours to weeks that evolved over the course of months to more chronic hypersomnia. Whole exome sequencing showed three mutations in genes potentially involved in his clinical phenotype. The patient has a predicted pathogenic de novo heterozygous p.Ala681Thr mutation in the ATP1A3 gene (chr19:42480621C>T, GRCh37/hg19). Mutations in this gene are known to cause Alternating Hemiplegia of Childhood, Rapid Onset Dystonia Parkinsonism, and CAPOS syndrome, sometimes accompanied by autistic features. The patient also has compound heterozygosity for p.Arg490Lys/p.Val200Met mutations in the NLRP3 gene (chr1:247588214G>A and chr1:247587343G>A, respectively). NLRP3 mutations are associated in an autosomal dominant manner with clinically overlapping auto-inflammatory conditions including Muckle-Wells syndrome. The p.Arg490Lys is a known pathogenic mutation inherited from the patient's father. The p.Val200Met mutation, inherited from his mother, is a variant of unknown significance (VUS). Whether the de novoATP1A3mutation is responsible for or plays a role in the patient's episodes of fatigue and somnolence remains to be determined. The unprecedented combination of two NLRP3 mutations may be responsible for other aspects of his complex phenotype.

12.
J Med Chem ; 61(3): 1001-1018, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29293004

RESUMEN

Computational modeling was used to direct the synthesis of analogs of previously reported phosphodiesterase 2A (PDE2A) inhibitor 1 with an imidazotriazine core to yield compounds of significantly enhanced potency. The analog PF-05180999 (30) was subsequently identified as a preclinical candidate targeting cognitive impairment associated with schizophrenia. Compound 30 demonstrated potent binding to PDE2A in brain tissue, dose responsive mouse brain cGMP increases, and reversal of N-methyl-d-aspartate (NMDA) antagonist-induced (MK-801, ketamine) effects in electrophysiology and working memory models in rats. Preclinical pharmacokinetics revealed unbound brain/unbound plasma levels approaching unity and good oral bioavailability resulting in an average concentration at steady state (Cav,ss) predicted human dose of 30 mg once daily (q.d.). Modeling of a modified release formulation suggested that 25 mg twice daily (b.i.d.) could maintain plasma levels of 30 at or above targeted efficacious plasma levels for 24 h, which became part of the human clinical plan.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Animales , Disponibilidad Biológica , Encéfalo/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Humanos , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/farmacocinética , Imidazoles/farmacología , Concentración 50 Inhibidora , Memoria a Corto Plazo/efectos de los fármacos , Simulación del Acoplamiento Molecular , Conformación Proteica
13.
J Med Chem ; 60(13): 5673-5698, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28574706

RESUMEN

Phosphodiesterase 2A (PDE2A) inhibitors have been reported to demonstrate in vivo activity in preclinical models of cognition. To more fully explore the biology of PDE2A inhibition, we sought to identify potent PDE2A inhibitors with improved brain penetration as compared to current literature compounds. Applying estimated human dose calculations while simultaneously leveraging synthetically enabled chemistry and structure-based drug design has resulted in a highly potent, selective, brain penetrant compound 71 (PF-05085727) that effects in vivo biochemical changes commensurate with PDE2A inhibition along with behavioral and electrophysiological reversal of the effects of NMDA antagonists in rodents. This data supports the ability of PDE2A inhibitors to potentiate NMDA signaling and their further development for clinical cognition indications.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Perros , Haplorrinos , Humanos , Ratones , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/farmacocinética , Ratas
14.
Dialogues Clin Neurosci ; 19(4): 335-343, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29398929

RESUMEN

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by social deficits and repetitive/restrictive interests. ASD is associated with multiple comorbidities, including intellectual disability, anxiety, and epilepsy. Evidence that ASD is highly heritable has spurred major efforts to unravel its genetics, revealing possible contributions from hundreds of genes through rare and common variation and through copy-number changes. In this perspective, we provide an overview of the current state of ASD genetics and of how genetic research has spurred the development of in vivo and in vitro models using animals and patient cells to evaluate the impact of genetic mutations on cellular function leading to disease. Efforts to translate these findings into successful therapies have yet to bear fruit. We discuss how the valuable insight into the disorder provided by these new models can be used to better understand ASD and develop future clinical trials.


El trastorno del espectro autista (TEA) es un complejo trastorno del neurodesarrollo caracterizado por déficits sociales e intereses repetitivos/restrictivos. El TEA se asocia con múltiples comorbilidades, incluyendo discapacidad intelectual, ansiedad y epilepsia. La evidencia de la alta heredabilidad del TEA ha estimulado los mayores esfuerzos para descifrar su genética, revelando posibles contribuciones de cientos de genes a través de variaciones raras y comunes, y de la variabilidad en el número de copias. Desde esta perspectiva se entrega una panorámica del estado actual de la genética del TEA y de cómo la investigación genética ha estimulado el desarrollo de modelos in vivo e in vitro que emplean células de animales y de pacientes para evaluar el impacto de las mutaciones genéticas en la función celular que lleva a la enfermedad. Aún no han dado frutos los esfuerzos realizados en la traducción de estos hallazgos en terapias exitosas. Se discute cómo se puede emplear la valiosa información acerca del trastorno, proporcionada por estos nuevos modelos, para una mejor comprensión del TEA y para desarrollar futuros ensayos clínicos.


Le trouble du spectre de l'autisme (TSA) représente un trouble complexe neurodéveloppemental caractérisé par des déficits sociaux et des intérêts répétitifs et restreints. Les comorbidités associées au TSA sont multiples, comme la déficience intellectuelle, l'anxiété et l'épilepsie. La transmissibilité élevée démontrée du TSA a encouragé des efforts importants pour décoder sa génétique, des centaines de gènes étant potentiellement impliqués par des variations rares et courantes et par la variabilité du nombre de copies. Dans cette perspective, nous présentons un aperçu de l'état actuel de la génétique du TSA et de la façon dont la recherche génétique a stimulé le développement de modèles in vivo et in vitro utilisant des cellules humaines et animales pour évaluer l'incidence de mutations génétiques sur les fonctions cellulaires responsables de la maladie. Les efforts pour traduire ces résultats en traitements efficaces n'ont pas encore porté leurs fruits. Nous expliquons comment les informations précieuses apportées par ces nouveaux modèles peuvent être utilisées pour mieux comprendre le TSA et développer de futurs essais cliniques.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/terapia , Terapia Genética/métodos , Mutación , Investigación Biomédica Traslacional , Animales , Modelos Animales de Enfermedad , Humanos
15.
J Neurosci ; 36(45): 11402-11410, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27911742

RESUMEN

Autism spectrum disorder (ASD) is a constellation of neurodevelopmental presentations with high heritability and both phenotypic and genetic heterogeneity. To date, mutations in hundreds of genes have been associated to varying degrees with increased ASD risk. A better understanding of the functions of these genes and whether they fit together in functional groups or impact similar neuronal circuits is needed to develop rational treatment strategies. We will review current areas of emphasis in ASD research, starting from human genetics and exploring how mouse models of human mutations have helped identify specific molecular pathways (protein synthesis and degradation, chromatin remodeling, intracellular signaling), which are linked to alterations in circuit function and cognitive/social behavior. We will conclude by discussing how we can leverage the findings on molecular and cellular alterations found in ASD to develop therapies for neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/terapia , Encéfalo/metabolismo , Terapia Genética/métodos , Proteínas del Tejido Nervioso/genética , Trastorno del Espectro Autista/diagnóstico , Medicina Basada en la Evidencia , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas/métodos , Humanos , Terapia Molecular Dirigida/métodos , Proteínas del Tejido Nervioso/metabolismo , Resultado del Tratamiento
16.
Cold Spring Harb Mol Case Stud ; 2(5): a001008, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27626066

RESUMEN

We describe a child with onset of command auditory hallucinations and behavioral regression at 6 yr of age in the context of longer standing selective mutism, aggression, and mild motor delays. His genetic evaluation included chromosomal microarray analysis and whole-exome sequencing. Sequencing revealed a previously unreported heterozygous de novo mutation c.385G>A in ATP1A3, predicted to result in a p.V129M amino acid change. This gene codes for a neuron-specific isoform of the catalytic α-subunit of the ATP-dependent transmembrane sodium-potassium pump. Heterozygous mutations in this gene have been reported as causing both sporadic and inherited forms of alternating hemiplegia of childhood and rapid-onset dystonia parkinsonism. We discuss the literature on phenotypes associated with known variants in ATP1A3, examine past functional studies of the role of ATP1A3 in neuronal function, and describe a novel clinical presentation associated with mutation of this gene.

17.
Biol Reprod ; 94(5): 110, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27009040

RESUMEN

The meiotic cell cycle of mammalian oocytes in preovulatory follicles is held in prophase arrest by diffusion of cGMP from the surrounding granulosa cells into the oocyte. Luteinizing hormone (LH) then releases meiotic arrest by lowering cGMP in the granulosa cells. The LH-induced reduction of cGMP is caused in part by a decrease in guanylyl cyclase activity, but the observation that the cGMP phosphodiesterase PDE5 is phosphorylated during LH signaling suggests that an increase in PDE5 activity could also contribute. To investigate this idea, we measured cGMP-hydrolytic activity in rat ovarian follicles. Basal activity was due primarily to PDE1A and PDE5, and LH increased PDE5 activity. The increase in PDE5 activity was accompanied by phosphorylation of PDE5 at serine 92, a protein kinase A/G consensus site. Both the phosphorylation and the increase in activity were promoted by elevating cAMP and opposed by inhibiting protein kinase A, supporting the hypothesis that LH activates PDE5 by stimulating its phosphorylation by protein kinase A. Inhibition of PDE5 activity partially suppressed LH-induced meiotic resumption as indicated by nuclear envelope breakdown, but inhibition of both PDE5 and PDE1 activities was needed to completely inhibit this response. These results show that activities of both PDE5 and PDE1 contribute to the LH-induced resumption of meiosis in rat oocytes, and that phosphorylation and activation of PDE5 is a regulatory mechanism.


Asunto(s)
GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hormona Luteinizante/farmacología , Meiosis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
18.
Am J Med Genet A ; 170A(5): 1165-73, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26887912

RESUMEN

Copy number variability at 16p13.11 has been associated with intellectual disability, autism, schizophrenia, epilepsy, and attention-deficit hyperactivity disorder. Adolescent/adult- onset psychosis has been reported in a subset of these cases. Here, we report on two children with CNVs in 16p13.11 that developed psychosis before the age of 7. The genotype and neuropsychiatric abnormalities of these patients highlight several overlapping genes that have possible mechanistic relevance to pathways previously implicated in Autism Spectrum Disorders, including the mTOR signaling and the ubiquitin-proteasome cascades. A careful screening of the 16p13.11 region is warranted in patients with childhood onset psychosis.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos Par 16/genética , Discapacidades del Desarrollo/genética , Trastornos Psicóticos/genética , Esquizofrenia/genética , Trastorno Autístico/fisiopatología , Niño , Preescolar , Deleción Cromosómica , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN/genética , Discapacidades del Desarrollo/fisiopatología , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Trastornos Psicóticos/fisiopatología , Esquizofrenia/fisiopatología , Transducción de Señal
19.
Sci Transl Med ; 8(320): 320ps1, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26738793

RESUMEN

The absence of mouse pharmacokinetic reference data hinders translation. An analysis of recent literature highlights a systematic lack of discussion regarding rationale for the selection of dosing paradigms in preclinical studies, and in particular for neuroscience studies in which the lack of brain penetration can limit target-organ exposure. We propose solutions to improve study design.


Asunto(s)
Evaluación Preclínica de Medicamentos , Estadística como Asunto , Investigación Biomédica Traslacional , Animales , Bases de Datos como Asunto , Humanos
20.
Neurobiol Dis ; 77: 220-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25796564

RESUMEN

Brain-derived neurotrophic factor (BDNF) signaling is integral to a range of neural functions, including synaptic plasticity and exhibits activity-dependent regulation of expression. As altered BDNF signaling has been implicated in multiple psychiatric diseases, here we report a quantitative reverse transcription polymerase chain reaction (RT-PCR) analysis of mRNAs encoding TrkB, total BDNF, and the four most abundant BDNF transcripts (I, IIc, IV, and VI) in postmortem tissue from matched tetrads of subjects with schizophrenia, bipolar disorder, or major depressive disorder (MDD) and healthy comparison subjects. In all three regions examined, dorsolateral prefrontal cortex (DLPFC), associative striatum and hippocampus, total BDNF mRNA levels did not differ in any disease state. In DLPFC, BDNF IIc was significantly lower in schizophrenia relative to healthy comparison subjects. In hippocampus, BDNF I, IIc, and VI were lower in subjects with both schizophrenia and bipolar disorder relative to comparison subjects. In striatum, TrkB mRNA was lower in bipolar disorder and MDD, while BDNF IIc was elevated in MDD, relative to comparison subjects. These data highlight potential alterations in BDNF signaling in the corticohippocampal circuit in schizophrenia, and within the striatum in mood disorders. Novel therapies aimed at improving BDNF-TrkB signaling may therefore have potential to impact on a range of psychiatric disorders.


Asunto(s)
Trastorno Bipolar/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/metabolismo , Trastorno Depresivo Mayor/patología , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Esquizofrenia/patología , Adulto , Factor Neurotrófico Derivado del Encéfalo/genética , Cuerpo Estriado/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor trkB
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...