Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37830916

RESUMEN

Dopamine system dysfunction is implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive and novel paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in Arc or Disc1 to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc-/- mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in Disc1+/- mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.


Asunto(s)
Antipsicóticos , Dopamina , Animales , Ratones , Dopamina/fisiología , Lóbulo Frontal , Cognición , Corteza Prefrontal/fisiología , Proteínas del Tejido Nervioso
2.
bioRxiv ; 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37745332

RESUMEN

Mutations in the GBA1 gene have been identified as a prevalent genetic risk factor for Parkinson's disease (PD). GBA1 mutations impair enzymatic activity, leading to lysosomal dysfunction and elevated levels of α-synuclein (α-syn). While most research has primarily focused on GBA1's role in promoting synucleinopathy, emerging evidence suggests that neuroinflammation may be a key pathogenic alteration caused by GBA1 deficiency. To examine the molecular mechanism underlying GBA1 deficiency-mediated neuroinflammation, we generated Gba1 E326K knock-in (KI) mice using the CRISPR/Cas9 technology, which is linked to an increased risk of PD and dementia with Lewy bodies (DLB). In the ventral midbrain and hippocampus of 24-month-old Gba1 E326K KI mice, we found a moderate decline in GBA1 enzymatic activity, a buildup of glucosylceramide, and an increase in microglia density. Furthermore, we observed increased levels of pro-inflammatory cytokines and formation of reactive astrocytes in primary microglia and astrocytes, respectively, cultured from Gba1 E326K KI mice following treatment with pathologic α-syn preformed fibrils (PFF). Additionally, the gut inoculation of α-syn PFF in Gba1 E326K KI mice significantly enhanced the accumulation of Lewy bodies in the dentate gyrus of the hippocampus, accompanied by aggravated neuroinflammation and exacerbated non-motor symptoms. This research significantly enhances our understanding of the Gba1 E326K mutation's involvement in neuroinflammation and the cell-to-cell transmission of pathogenic α-syn in the brain, thereby opening new therapeutic avenues.

3.
Immunity ; 56(9): 2105-2120.e13, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37527657

RESUMEN

Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.


Asunto(s)
Astrocitos , Fagocitosis , Estrés Psicológico , Animales , Niño , Humanos , Ratones , Astrocitos/metabolismo , Tirosina Quinasa c-Mer/genética , Hormonas/metabolismo , Sinapsis/metabolismo , Estrés Psicológico/metabolismo
4.
Stem Cell Reports ; 18(7): 1468-1481, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37390823

RESUMEN

Quiescence is a hallmark of adult neural stem cells (NSCs) in the mammalian brain, and establishment and maintenance of quiescence is essential for life-long continuous neurogenesis. How NSCs in the dentate gyrus (DG) of the hippocampus acquire their quiescence during early postnatal stages and continuously maintain quiescence in adulthood is poorly understood. Here, we show that Hopx-CreERT2-mediated conditional deletion of Nkcc1, which encodes a chloride importer, in mouse DG NSCs impairs both their quiescence acquisition at early postnatal stages and quiescence maintenance in adulthood. Furthermore, PV-CreERT2-mediated deletion of Nkcc1 in PV interneurons in the adult mouse brain leads to activation of quiescent DG NSCs, resulting in an expanded NSC pool. Consistently, pharmacological inhibition of NKCC1 promotes NSC proliferation in both early postnatal and adult mouse DG. Together, our study reveals both cell-autonomous and non-cell-autonomous roles of NKCC1 in regulating the acquisition and maintenance of NSC quiescence in the mammalian hippocampus.


Asunto(s)
Hipocampo , Células-Madre Neurales , Animales , Ratones , Neurogénesis/fisiología , División Celular , Giro Dentado , Mamíferos
5.
bioRxiv ; 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-36778456

RESUMEN

Dopamine system dysfunction is commonly implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive to such pharmacological treatments and novel research paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in the Arc or DISC1 genes to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc mutant mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in DISC1 mutant mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.

6.
Cell Stem Cell ; 30(3): 300-311.e11, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36764294

RESUMEN

Increasing evidence implicates the critical roles of various epitranscriptomic RNA modifications in different biological processes. Methyltransferase METTL8 installs 3-methylcytosine (m3C) modification of mitochondrial tRNAs in vitro; however, its role in intact biological systems is unknown. Here, we show that Mettl8 is localized in mitochondria and installs m3C specifically on mitochondrial tRNAThr/Ser(UCN) in mouse embryonic cortical neural stem cells. At molecular and cellular levels, Mettl8 deletion in cortical neural stem cells leads to reduced mitochondrial protein translation and attenuated respiration activity. At the functional level, conditional Mettl8 deletion in mice results in impaired embryonic cortical neural stem cell maintenance in vivo, which can be rescued by pharmacologically enhancing mitochondrial functions. Similarly, METTL8 promotes mitochondrial protein expression and neural stem cell maintenance in human forebrain cortical organoids. Together, our study reveals a conserved epitranscriptomic mechanism of Mettl8 and mitochondrial tRNA m3C modification in maintaining embryonic cortical neural stem cells in mice and humans.


Asunto(s)
Metiltransferasas , Mitocondrias , Ratones , Animales , Humanos , Mitocondrias/metabolismo , Metiltransferasas/genética , ARN de Transferencia/metabolismo , Neurogénesis , Proteínas Mitocondriales/metabolismo
7.
Neurosci Res ; 187: 3-13, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36170922

RESUMEN

Based on experience during our life, neuronal connectivity continuously changes through structural remodeling of synapses. Recent studies have shown that the complex interaction between astrocytes and synapses regulates structural synapse remodeling by inducing the formation and elimination of synapses, as well as their functional maturation. Defects in this astrocyte-mediated synapse remodeling cause problems in not only neuronal network activities but also animal behaviors. Moreover, in various neurological disorders, astrocytes have been shown to play central roles in the initiation and progression of synaptic pathophysiology through impaired interactions with synapses. In this review, we will discuss recent studies identifying the novel roles of astrocytes in neuronal circuit remodeling, focusing on synapse formation and elimination. We will also discuss the potential implication of defective astrocytic function in evoking various brain disorders.


Asunto(s)
Encefalopatías , Enfermedades del Sistema Nervioso , Animales , Astrocitos/fisiología , Sinapsis/fisiología , Neuronas/fisiología , Plasticidad Neuronal/fisiología
8.
Biol Psychiatry ; 92(10): 815-826, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34247782

RESUMEN

BACKGROUND: Gene dosage imbalance caused by copy number variations (CNVs) is a prominent contributor to brain disorders. In particular, 15q11.2 CNV duplications and deletions have been associated with autism spectrum disorder and schizophrenia, respectively. The mechanism underlying these diametric contributions remains unclear. METHODS: We established both loss-of-function and gain-of-function mouse models of Cyfip1, one of four genes within 15q11.2 CNVs. To assess the functional consequences of altered CYFIP1 levels, we performed systematic investigations on behavioral, electrophysiological, and biochemical phenotypes in both mouse models. In addition, we utilized RNA immunoprecipitation sequencing (RIP-seq) analysis to reveal molecular targets of CYFIP1 in vivo. RESULTS: Cyfip1 loss-of-function and gain-of function mouse models exhibited distinct and shared behavioral abnormalities related to autism spectrum disorder and schizophrenia. RIP-seq analysis identified messenger RNA targets of CYFIP1 in vivo, including postsynaptic NMDA receptor (NMDAR) complex components. In addition, these mouse models showed diametric changes in levels of postsynaptic NMDAR complex components at synapses because of dysregulated protein translation, resulting in bidirectional alteration of NMDAR-mediated signaling. Importantly, pharmacological balancing of NMDAR signaling in these mouse models with diametric Cyfip1 dosages rescues behavioral abnormalities. CONCLUSIONS: CYFIP1 regulates protein translation of NMDAR and associated complex components at synapses to maintain normal synaptic functions and behaviors. Our integrated analyses provide insight into how gene dosage imbalance caused by CNVs may contribute to divergent neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastornos Mentales , Ratones , Animales , Receptores de N-Metil-D-Aspartato/genética , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Variaciones en el Número de Copia de ADN , Ratones Endogámicos C57BL , N-Metilaspartato/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Modelos Animales de Enfermedad , ARN Mensajero , ARN
9.
Neurobiol Stress ; 15: 100373, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34401409

RESUMEN

Depression is accompanied by neuronal atrophy and decreased neuroplasticity. Leucine-rich glioma-inactivated protein 1 (LGI1), a metastasis suppressor, plays an important role in the development of CNS synapses. We found that LGI1 expression was reduced in the hippocampi of mice that underwent chronic unpredictable stress (CUS), and could be rescued by the antidepressant, fluoxetine. Recombinant soluble neuritin, an endogenous protein previously implicated in antidepressant-like behaviors, elevated hippocampal LGI1 expression in a manner dependent on histone deacetylase 5 (HDAC5) phosphorylation. Accordingly, Nrn1 flox/flox ;Pomc-cre (Nrn1 cOE) mice, which conditionally overexpress neuritin, displayed increases in hippocampal LGI1 level under CUS and exhibited resilience to CUS that were blocked by hippocampal depletion of LGI1. Interestingly, neuritin-mediated LGI1 expression was inhibited by HNMPA-(AM)3, an insulin receptor inhibitor, as was neuritin-mediated HDAC5 phosphorylation. We thus establish hippocampal LGI1 as an effector of neurite outgrowth and stress resilience, and suggest that HDAC5-LGI1 plays a critical role in ameliorating pathological depression.

11.
Nat Commun ; 12(1): 1398, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658519

RESUMEN

We previously identified a causal link between a rare patient mutation in DISC1 (disrupted-in-schizophrenia 1) and synaptic deficits in cortical neurons differentiated from isogenic patient-derived induced pluripotent stem cells (iPSCs). Here we find that transcripts related to phosphodiesterase 4 (PDE4) signaling are significantly elevated in human cortical neurons differentiated from iPSCs with the DISC1 mutation and that inhibition of PDE4 or activation of the cAMP signaling pathway functionally rescues synaptic deficits. We further generated a knock-in mouse line harboring the same patient mutation in the Disc1 gene. Heterozygous Disc1 mutant mice exhibit elevated levels of PDE4s and synaptic abnormalities in the brain, and social and cognitive behavioral deficits. Pharmacological inhibition of the PDE4 signaling pathway rescues these synaptic, social and cognitive behavioral abnormalities. Our study shows that patient-derived isogenic iPSC and humanized mouse disease models are integral and complementary for translational studies with a better understanding of underlying molecular mechanisms.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Inhibidores de Fosfodiesterasa 4/farmacología , Esquizofrenia/genética , Animales , Conducta Animal/efectos de los fármacos , Corteza Cerebral/fisiología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Ratones Mutantes , Mutación , Neuronas/efectos de los fármacos , Rolipram/farmacología , Esquizofrenia/patología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
12.
J Neurosci ; 40(10): 2015-2024, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31988061

RESUMEN

Neural stem cells (NSCs) persist throughout life in the subventricular zone (SVZ) neurogenic niche of the lateral ventricles as Type B1 cells in adult mice. Maintaining this population of NSCs depends on the balance between quiescence and self-renewing or self-depleting cell divisions. Interactions between B1 cells and the surrounding niche are important in regulating this balance, but the mechanisms governing these processes have not been fully elucidated. The cytoplasmic FMRP-interacting protein (Cyfip1) regulates apical-basal polarity in the embryonic brain. Loss of Cyfip1 during embryonic development in mice disrupts the embryonic niche and affects cortical neurogenesis. However, a direct role for Cyfip1 in the regulation of adult NSCs has not been established. Here, we demonstrate that Cyfip1 expression is preferentially localized to B1 cells in the adult mouse SVZ. Loss of Cyfip1 in the embryonic mouse brain results in altered adult SVZ architecture and expansion of the adult B1 cell population at the ventricular surface. Furthermore, acute deletion of Cyfip1 in adult NSCs results in a rapid change in adherens junction proteins as well as increased proliferation and number of B1 cells at the ventricular surface. Together, these data indicate that Cyfip1 plays a critical role in the formation and maintenance of the adult SVZ niche; furthermore, deletion of Cyfip1 unleashes the capacity of adult B1 cells for symmetric renewal to increase the adult NSC pool.SIGNIFICANCE STATEMENT Neural stem cells (NSCs) persist in the subventricular zone of the lateral ventricles in adult mammals, and the size of this population is determined by the balance between quiescence and self-depleting or renewing cell division. The mechanisms regulating these processes are not fully understood. This study establishes that the cytoplasmic FMRP interacting protein 1 (Cyfip1) regulates NSC fate decisions in the adult subventricular zone and adult NSCs that are quiescent or typically undergo self-depleting divisions retain the ability to self-renew. These results contribute to our understanding of how adult NSCs are regulated throughout life and has potential implications for human brain disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ventrículos Laterales/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Nicho de Células Madre/fisiología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Envejecimiento , Animales , Ventrículos Laterales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología
13.
Development ; 145(14)2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29950389

RESUMEN

Mammary glands develop through primary ductal elongation and side branching to maximize the spatial area. Although primary ducts are generated by bifurcation of terminal end buds, the mechanism through which side branching occurs is still largely unclear. Here, we show that inhibitor of DNA-binding 2 (ID2) drives side branch formation through the differentiation of K6+ bipotent progenitor cells (BPs) into CD61+ luminal progenitor cells (LPs). Id2-null mice had side-branching defects, along with developmental blockage of the differentiation of K6+ BPs into CD61+ LPs. Notably, CD61+ LPs were found in budding and side branches, but not in terminal end buds. Hormone reconstitution studies using ovariectomized MMTV-hemagglutinin-nuclear localized sequence-tagged Id2 transgenic mice revealed that ID2 is a key mediator of progesterone, which drives luminal lineage differentiation and side branching. Our results suggest that CD61 is a marker of side branches and that ID2 regulates side branch formation by inducing luminal lineage commitment from K6+ BPs to CD61+ LPs.


Asunto(s)
Tipificación del Cuerpo , Linaje de la Célula , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Núcleo Celular/metabolismo , Femenino , Eliminación de Gen , Imagenología Tridimensional , Integrina beta3/metabolismo , Ratones , Modelos Biológicos , Progesterona/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
14.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28965759

RESUMEN

N6-methyladenosine (m6A), installed by the Mettl3/Mettl14 methyltransferase complex, is the most prevalent internal mRNA modification. Whether m6A regulates mammalian brain development is unknown. Here, we show that m6A depletion by Mettl14 knockout in embryonic mouse brains prolongs the cell cycle of radial glia cells and extends cortical neurogenesis into postnatal stages. m6A depletion by Mettl3 knockdown also leads to a prolonged cell cycle and maintenance of radial glia cells. m6A sequencing of embryonic mouse cortex reveals enrichment of mRNAs related to transcription factors, neurogenesis, the cell cycle, and neuronal differentiation, and m6A tagging promotes their decay. Further analysis uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells. m6A signaling also regulates human cortical neurogenesis in forebrain organoids. Comparison of m6A-mRNA landscapes between mouse and human cortical neurogenesis reveals enrichment of human-specific m6A tagging of transcripts related to brain-disorder risk genes. Our study identifies an epitranscriptomic mechanism in heightened transcriptional coordination during mammalian cortical neurogenesis.


Asunto(s)
Neurogénesis , Prosencéfalo/embriología , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Animales , Ciclo Celular , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Organoides/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , Estabilidad del ARN
15.
Cell Stem Cell ; 21(3): 349-358.e6, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28826723

RESUMEN

Zika virus (ZIKV) directly infects neural progenitors and impairs their proliferation. How ZIKV interacts with the host molecular machinery to impact neurogenesis in vivo is not well understood. Here, by systematically introducing individual proteins encoded by ZIKV into the embryonic mouse cortex, we show that expression of ZIKV-NS2A, but not Dengue virus (DENV)-NS2A, leads to reduced proliferation and premature differentiation of radial glial cells and aberrant positioning of newborn neurons. Mechanistically, in vitro mapping of protein-interactomes and biochemical analysis suggest interactions between ZIKA-NS2A and multiple adherens junction complex (AJ) components. Functionally, ZIKV-NS2A, but not DENV-NS2A, destabilizes the AJ complex, resulting in impaired AJ formation and aberrant radial glial fiber scaffolding in the embryonic mouse cortex. Similarly, ZIKA-NS2A, but not DENV-NS2A, reduces radial glial cell proliferation and causes AJ deficits in human forebrain organoids. Together, our results reveal pathogenic mechanisms underlying ZIKV infection in the developing mammalian brain.


Asunto(s)
Uniones Adherentes/metabolismo , Corteza Cerebral/metabolismo , Mamíferos/metabolismo , Proteínas de la Membrana/metabolismo , Neurogénesis , Proteolisis , Proteínas no Estructurales Virales/metabolismo , Virus Zika/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Corteza Cerebral/embriología , Células HEK293 , Humanos , Ratones , Neuroglía/patología , Unión Proteica , Mapeo de Interacción de Proteínas , Infección por el Virus Zika/patología
16.
Mol Cell ; 61(1): 153-60, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26711009

RESUMEN

Lin28, a well-known RNA-binding protein, regulates diverse cellular properties. All physiological functions of Lin28A characterized so far have been attributed to its repression of let-7 miRNA biogenesis or modulation of mRNA translational efficiency. Here we show that Lin28A directly binds to a consensus DNA sequence in vitro and in mouse embryonic stem cells in vivo. ChIP-seq and RNA-seq reveal enrichment of Lin28A binding around transcription start sites and a positive correlation between its genomic occupancy and expression of many associated genes. Mechanistically, Lin28A recruits 5-methylcytosine-dioxygenase Tet1 to genomic binding sites to orchestrate 5-methylcytosine and 5-hydroxymethylcytosine dynamics. Either Lin28A or Tet1 knockdown leads to dysregulated DNA methylation and expression of common target genes. These results reveal a surprising role for Lin28A in transcriptional regulation via epigenetic DNA modifications and have implications for understanding mechanisms underlying versatile functions of Lin28A in mammalian systems.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular , Metilación de ADN , Proteínas de Unión al ADN/genética , Epigénesis Genética , Ratones , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteínas de Unión al ARN/genética , Transfección
17.
Nature ; 515(7527): 414-8, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25132547

RESUMEN

Dysregulated neurodevelopment with altered structural and functional connectivity is believed to underlie many neuropsychiatric disorders, and 'a disease of synapses' is the major hypothesis for the biological basis of schizophrenia. Although this hypothesis has gained indirect support from human post-mortem brain analyses and genetic studies, little is known about the pathophysiology of synapses in patient neurons and how susceptibility genes for mental disorders could lead to synaptic deficits in humans. Genetics of most psychiatric disorders are extremely complex due to multiple susceptibility variants with low penetrance and variable phenotypes. Rare, multiply affected, large families in which a single genetic locus is probably responsible for conferring susceptibility have proven invaluable for the study of complex disorders. Here we generated induced pluripotent stem (iPS) cells from four members of a family in which a frameshift mutation of disrupted in schizophrenia 1 (DISC1) co-segregated with major psychiatric disorders and we further produced different isogenic iPS cell lines via gene editing. We showed that mutant DISC1 causes synaptic vesicle release deficits in iPS-cell-derived forebrain neurons. Mutant DISC1 depletes wild-type DISC1 protein and, furthermore, dysregulates expression of many genes related to synapses and psychiatric disorders in human forebrain neurons. Our study reveals that a psychiatric disorder relevant mutation causes synapse deficits and transcriptional dysregulation in human neurons and our findings provide new insight into the molecular and synaptic etiopathology of psychiatric disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Trastornos Mentales/patología , Sinapsis/patología , Animales , Diferenciación Celular , Fibroblastos , Glutamina/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Linaje , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Unión Proteica , Sinapsis/metabolismo , Transcriptoma
18.
Cell Stem Cell ; 15(1): 79-91, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24996170

RESUMEN

Defects in brain development are believed to contribute toward the onset of neuropsychiatric disorders, but identifying specific underlying mechanisms has proven difficult. Here, we took a multifaceted approach to investigate why 15q11.2 copy number variants are prominent risk factors for schizophrenia and autism. First, we show that human iPSC-derived neural progenitors carrying 15q11.2 microdeletion exhibit deficits in adherens junctions and apical polarity. This results from haploinsufficiency of CYFIP1, a gene within 15q11.2 that encodes a subunit of the WAVE complex, which regulates cytoskeletal dynamics. In developing mouse cortex, deficiency in CYFIP1 and WAVE signaling similarly affects radial glial cells, leading to their ectopic localization outside of the ventricular zone. Finally, targeted human genetic association analyses revealed an epistatic interaction between CYFIP1 and WAVE signaling mediator ACTR2 and risk for schizophrenia. Our findings provide insight into how CYFIP1 regulates neural stem cell function and may contribute to the susceptibility of neuropsychiatric disorders.


Asunto(s)
Uniones Adherentes/genética , Trastorno Autístico/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Esquizofrenia/metabolismo , Proteína 2 Relacionada con la Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Uniones Adherentes/patología , Adulto , Animales , Trastorno Autístico/genética , Trastorno Autístico/patología , Línea Celular , Polaridad Celular/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 15/genética , Variaciones en el Número de Copia de ADN , Epistasis Genética , Estudios de Asociación Genética , Haploinsuficiencia , Humanos , Discapacidad Intelectual/genética , Masculino , Ratones , Ratones Endogámicos , Persona de Mediana Edad , Riesgo , Esquizofrenia/genética , Esquizofrenia/patología , Población Blanca , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
19.
Mol Brain ; 5: 40, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23111145

RESUMEN

BACKGROUND: Notch signaling is well recognized as a key regulator of the neuronal fate during embryonic development, but its function in the adult brain is still largely unknown. Mind bomb-1 (Mib1) is an essential positive regulator in the Notch pathway, acting non-autonomously in the signal-sending cells. Therefore, genetic ablation of Mib1 in mature neuron would give valuable insight to understand the cell-to-cell interaction between neurons via Notch signaling for their proper function. RESULTS: Here we show that the inactivation of Mib1 in mature neurons in forebrain results in impaired hippocampal dependent spatial memory and contextual fear memory. Consistently, hippocampal slices from Mib1-deficient mice show impaired late-phase, but not early-phase, long-term potentiation and long-term depression without change in basal synaptic transmission at SC-CA1 synapses. CONCLUSIONS: These data suggest that Mib1-mediated Notch signaling is essential for long-lasting synaptic plasticity and memory formation in the rodent hippocampus.


Asunto(s)
Memoria a Largo Plazo/fisiología , Plasticidad Neuronal/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Sinapsis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Envejecimiento/metabolismo , Animales , Hipocampo/anatomía & histología , Hipocampo/enzimología , Potenciación a Largo Plazo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fenotipo , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Receptores Notch/química
20.
Mol Cell Biol ; 31(23): 4775-88, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21947283

RESUMEN

RANKL plays an essential role in mammary gland development during pregnancy. However, the molecular mechanism by which RANK signaling leads to mammary gland development is largely unknown. We report here that RANKL stimulation induces phosphorylation of Id2 at serine 5, which leads to nuclear retention of Id2. In lactating Id2Tg; RANKL(-/-) mice, Id2 was not phosphorylated and was localized in the cytoplasm. In addition, in lactating Id2(S5A)Tg mice, Id2(S5A) (with serine 5 mutated to alanine) was exclusively localized in the cytoplasm of mammary epithelial cells (MECs), while endogenous Id2 was localized in the nucleus. Intriguingly, nuclear expression of Id2(S5A) rescued increased apoptosis and defective differentiation of MECs in RANKL(-/-) mice. Our results demonstrate that nuclear retention of Id2 due to RANK signaling plays a decisive role in the survival and differentiation of MECs during mammary gland development.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , Supervivencia Celular , Células Epiteliales/fisiología , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Proteína 2 Inhibidora de la Diferenciación/genética , Lactancia , Masculino , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Fosforilación , Embarazo , Transporte de Proteínas , Receptor Activador del Factor Nuclear kappa-B/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...