Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33833053

RESUMEN

Copy number variation (CNV) at the 16p11.2 locus is associated with neuropsychiatric disorders, such as autism spectrum disorder and schizophrenia. CNVs of the 16p gene can manifest in opposing head sizes. Carriers of 16p11.2 deletion tend to have macrocephaly (or brain enlargement), while those with 16p11.2 duplication frequently have microcephaly. Increases in both gray and white matter volume have been observed in brain imaging studies in 16p11.2 deletion carriers with macrocephaly. Here, we use human induced pluripotent stem cells (hiPSCs) derived from controls and subjects with 16p11.2 deletion and 16p11.2 duplication to understand the underlying mechanisms regulating brain overgrowth. To model both gray and white matter, we differentiated patient-derived iPSCs into neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). In both NPCs and OPCs, we show that CD47 (a "don't eat me" signal) is overexpressed in the 16p11.2 deletion carriers contributing to reduced phagocytosis both in vitro and in vivo. Furthermore, 16p11.2 deletion NPCs and OPCs up-regulate cell surface expression of calreticulin (a prophagocytic "eat me" signal) and its binding sites, indicating that these cells should be phagocytosed but fail to be eliminated due to elevations in CD47. Treatment of 16p11.2 deletion NPCs and OPCs with an anti-CD47 antibody to block CD47 restores phagocytosis to control levels. While the CD47 pathway is commonly implicated in cancer progression, we document a role for CD47 in psychiatric disorders associated with brain overgrowth.


Asunto(s)
Trastorno Autístico/metabolismo , Encéfalo/metabolismo , Antígeno CD47/metabolismo , Trastornos de los Cromosomas/metabolismo , Discapacidad Intelectual/metabolismo , Adolescente , Adulto , Animales , Trastorno Autístico/patología , Encéfalo/patología , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/genética , Calreticulina/genética , Calreticulina/metabolismo , Línea Celular , Células Cultivadas , Niño , Preescolar , Deleción Cromosómica , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 16/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Discapacidad Intelectual/patología , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo
2.
Mol Psychiatry ; 26(6): 2605-2615, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32047265

RESUMEN

Genome-wide association studies (GWAS) have successfully identified 145 loci implicated in schizophrenia (SCZ). However, the underlying mechanisms remain largely unknown. Here, we analyze 1497 RNA-seq data in combination with their genotype data and identify SNPs that are associated with expression throughout the genome by dissecting expression features to genes (eGene) and exon-exon junctions (eJunction). Then, we colocalize eGene and eJunction with SCZ GWAS using SMR and fine mapping. Multiple ChIP-seq data and DNA methylation data generated from brain were used for identifying the causal variants. Finally, we used a hypothesis-free (no SCZ risk loci considered) enrichment analysis to determine implicated pathways. We identified 171 genes and eight splicing junctions located within four genes (SNX19, ARL6IP4, APOPT1, and CYP2D6) that potentially contribute to SCZ susceptibility. Among the genes, CYP2D6 is significantly associated with SCZ SNPs in eGene and eJunction. In-depth examination of the CYP2D6 region revealed that a nonsynonymous single nucleotide variant rs16947 is strongly associated with a higher abundance of CYP2D6 exon 3 skipping junctions. While we found rs133377 and other functional SNPs in high linkage disequilibrium with rs16947 (r2 = 0.9539), histone acetylation analysis showed they are located within active transcription start sites. Furthermore, our data-driven enrichment analysis showed that CYP2D6 is significantly involved in drug metabolism of codeine, tamoxifen, and citalopram. Our study facilitates an understanding of the genetic architecture of SCZ and provides new drug targets.


Asunto(s)
Esquizofrenia , Citocromo P-450 CYP2D6/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/genética
3.
Stem Cells ; 37(9): 1151-1157, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31135093

RESUMEN

Understanding the molecular properties of the cell cycle of human pluripotent stem cells (hPSCs) is critical for effectively promoting differentiation. Here, we use the Fluorescence Ubiquitin Cell Cycle Indicator system adapted into hPSCs and perform RNA sequencing on cell cycle sorted hPSCs primed and unprimed for differentiation. Gene expression patterns of signaling factors and developmental regulators change in a cell cycle-specific manner in cells primed for differentiation without altering genes associated with pluripotency. Furthermore, we identify an important role for PI3K signaling in regulating the early transitory states of hPSCs toward differentiation. Stem Cells 2019;37:1151-1157.


Asunto(s)
Ciclo Celular/genética , Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Células Madre Pluripotentes/metabolismo , Células Cultivadas , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre Pluripotentes/citología , Análisis de Secuencia de ARN/métodos , Transducción de Señal
4.
Nat Methods ; 10(6): 553-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23584186

RESUMEN

We describe a method to help overcome restrictions on the differentiation propensities of human pluripotent stem cells. Culturing pluripotent stem cells in dimethylsulfoxide (DMSO) activates the retinoblastoma protein, increases the proportion of cells in the early G1 phase of the cell cycle and, in more than 25 embryonic and induced pluripotent stem cell lines, improves directed differentiation into multiple lineages. DMSO treatment also improves differentiation into terminal cell types in several cell lines.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Cultivadas , Dimetilsulfóxido/farmacología , Células Madre Embrionarias/citología , Fase G1/efectos de los fármacos , Humanos , Proteína de Retinoblastoma/metabolismo
5.
Mol Psychiatry ; 19(12): 1275-1283, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24514565

RESUMEN

Stress can exert long-lasting changes on the brain that contribute to vulnerability to mental illness, yet mechanisms underlying this long-term vulnerability are not well understood. We hypothesized that stress may alter the production of oligodendrocytes in the adult brain, providing a cellular and structural basis for stress-related disorders. We found that immobilization stress decreased neurogenesis and increased oligodendrogenesis in the dentate gyrus (DG) of the adult rat hippocampus and that injections of the rat glucocorticoid stress hormone corticosterone (cort) were sufficient to replicate this effect. The DG contains a unique population of multipotent neural stem cells (NSCs) that give rise to adult newborn neurons, but oligodendrogenic potential has not been demonstrated in vivo. We used a nestin-CreER/YFP transgenic mouse line for lineage tracing and found that cort induces oligodendrogenesis from nestin-expressing NSCs in vivo. Using hippocampal NSCs cultured in vitro, we further showed that exposure to cort induced a pro-oligodendrogenic transcriptional program and resulted in an increase in oligodendrogenesis and decrease in neurogenesis, which was prevented by genetic blockade of glucocorticoid receptor (GR). Together, these results suggest a novel model in which stress may alter hippocampal function by promoting oligodendrogenesis, thereby altering the cellular composition and white matter structure.


Asunto(s)
Diferenciación Celular/fisiología , Corticosterona/metabolismo , Glucocorticoides/metabolismo , Hipocampo/fisiología , Oligodendroglía/fisiología , Estrés Psicológico/fisiopatología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Corticosterona/administración & dosificación , Modelos Animales de Enfermedad , Glucocorticoides/administración & dosificación , Hipocampo/efectos de los fármacos , Masculino , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oligodendroglía/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Restricción Física
6.
Methods Mol Biol ; 2683: 89-101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37300769

RESUMEN

Oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes (OLs) can be generated using human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs). By manipulating culture conditions, pluripotent cell types are serially guided through intermediary cell types, developing first into neural progenitor cells (NPCs) then OPCs before maturing as CNS-specific OLs. This procedure is conducted under adherent, feeder-free conditions to derive mature OLs in as few as 28 days.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Cultivadas , Diferenciación Celular , Oligodendroglía , Células Madre Embrionarias
7.
Sci Transl Med ; 13(622): eabg2919, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34851695

RESUMEN

Lysosome dysfunction is a shared feature of rare lysosomal storage diseases and common age-related neurodegenerative diseases. Microglia, the brain-resident macrophages, are particularly vulnerable to lysosome dysfunction because of the phagocytic stress of clearing dying neurons, myelin, and debris. CD22 is a negative regulator of microglial homeostasis in the aging mouse brain, and soluble CD22 (sCD22) is increased in the cerebrospinal fluid of patients with Niemann-Pick type C disease (NPC). However, the role of CD22 in the human brain remains unknown. In contrast to previous findings in mice, here, we show that CD22 is expressed by oligodendrocytes in the human brain and binds to sialic acid­dependent ligands on microglia. Using unbiased genetic and proteomic screens, we identify insulin-like growth factor 2 receptor (IGF2R) as the binding partner of sCD22 on human myeloid cells. Targeted truncation of IGF2R revealed that sCD22 docks near critical mannose 6-phosphate­binding domains, where it disrupts lysosomal protein trafficking. Interfering with the sCD22-IGF2R interaction using CD22 blocking antibodies ameliorated lysosome dysfunction in human NPC1 mutant induced pluripotent stem cell­derived microglia-like cells without harming oligodendrocytes in vitro. These findings reinforce the differences between mouse and human microglia and provide a candidate microglia-directed immunotherapeutic to treat NPC.


Asunto(s)
Microglía , Enfermedad de Niemann-Pick Tipo C , Animales , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Ratones , Microglía/metabolismo , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Proteómica , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/uso terapéutico
8.
J Vis Exp ; (165)2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33226027

RESUMEN

In Alzheimer's disease (AD) and other neurodegenerative disorders, oligodendroglial failure is a common early pathological feature, but how it contributes to disease development and progression, particularly in the gray matter of the brain, remains largely unknown. The dysfunction of oligodendrocyte lineage cells is hallmarked by deficiencies in myelination and impaired self-renewal of oligodendrocyte precursor cells (OPCs). These two defects are caused at least in part by the disruption of interactions between neuron and oligodendrocytes along the buildup of pathology. OPCs give rise to myelinating oligodendrocytes during CNS development. In the mature brain cortex, OPCs are the major proliferative cells (comprising ~5% of total brain cells) and control new myelin formation in a neural activity-dependent manner. Such neuron-to-oligodendrocyte communications are significantly understudied, especially in the context of neurodegenerative conditions such as AD, due to the lack of appropriate tools. In recent years, our group and others have made significant progress to improve currently available protocols to generate functional neurons and oligodendrocytes individually from human pluripotent stem cells. In this manuscript, we describe our optimized procedures, including the establishment of a co-culture system to model the neuron-oligodendrocyte connections. Our illustrative results suggest an unexpected contribution from OPCs/oligodendrocytes to the brain amyloidosis and synapse integrity and highlight the utility of this methodology for AD research. This reductionist approach is a powerful tool to dissect the specific hetero-cellular interactions out of the inherent complexity inside the brain. The protocols we describe here are expected to facilitate future studies on oligodendroglial defects in the pathogenesis of neurodegeneration.


Asunto(s)
Comunicación Celular , Técnicas de Cultivo de Célula/métodos , Neuronas/citología , Oligodendroglía/citología , Células Madre Pluripotentes/citología , Diferenciación Celular , Linaje de la Célula , Técnicas de Cocultivo , Dimetilsulfóxido/farmacología , Células HEK293 , Humanos , Vaina de Mielina/fisiología
9.
J Vis Exp ; (149)2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31380840

RESUMEN

Despite the growing use of pluripotent stem cells (PSCs), challenges in efficiently differentiating embryonic and induced pluripotent stem cells (ESCs and iPSCs) across various lineages remain. Numerous differentiation protocols have been developed, yet variability across cell lines and low rates of differentiation impart challenges in successfully implementing these protocols. Described here is an easy and inexpensive means to enhance the differentiation capacity of PSCs. It has been previously shown that treatment of stem cells with a low concentration of dimethyl sulfoxide (DMSO) significantly increases the propensity of a variety of PSCs to differentiate to different cell types following directed differentiation. This technique has now been shown to be effective across different species (e.g., mouse, primate, and human) into multiple lineages, ranging from neurons and cortical spheroids to smooth muscle cells and hepatocytes. The DMSO pretreatment improves PSC differentiation by regulating the cell cycle and priming stem cells to be more responsive to differentiation signals. Provided here is the detailed methodology for using this simple tool as a reproducible and widely applicable means to more efficiently differentiate PSCs to any lineage of choice.


Asunto(s)
Dimetilsulfóxido/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular , Humanos , Células Madre Pluripotentes/citología
10.
Brain Res ; 1232: 30-47, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18691562

RESUMEN

We examined the effects of aging on visuo-spatial attention. Participants performed a bi-field visual selective attention task consisting of infrequent target and task-irrelevant novel stimuli randomly embedded among repeated standards in either attended or unattended visual fields. Blood oxygenation level dependent (BOLD) responses to the different classes of stimuli were measured using functional magnetic resonance imaging. The older group had slower reaction times to targets, and committed more false alarms but had comparable detection accuracy to young controls. Attended target and novel stimuli activated comparable widely distributed attention networks, including anterior and posterior association cortex, in both groups. The older group had reduced spatial extent of activation in several regions, including prefrontal, basal ganglia, and visual processing areas. In particular, the anterior cingulate and superior frontal gyrus showed more restricted activation in older compared with young adults across all attentional conditions and stimulus categories. The spatial extent of activations correlated with task performance in both age groups, but the regional pattern of association between hemodynamic responses and behavior differed between the groups. Whereas the young subjects relied on posterior regions, the older subjects engaged frontal areas. The results indicate that aging alters the functioning of neural networks subserving visual attention, and that these changes are related to cognitive performance.


Asunto(s)
Envejecimiento/psicología , Corteza Prefrontal/fisiología , Desempeño Psicomotor/fisiología , Adulto , Anciano , Atención/fisiología , Conducta/fisiología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Circulación Cerebrovascular/fisiología , Electroencefalografía , Potenciales Evocados , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/fisiología , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Tiempo de Reacción/fisiología , Encuestas y Cuestionarios , Adulto Joven
11.
PLoS One ; 13(12): e0208110, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30540809

RESUMEN

The propensity for differentiation varies substantially across human pluripotent stem cell (hPSC) lines, greatly restricting the use of hPSCs for cell replacement therapy or disease modeling. Here, we investigate the underlying mechanisms and demonstrate that activation of the retinoblastoma (Rb) pathway in a transient manner is important for differentiation. In prior work, we demonstrated that pre-treating hPSCs with dimethylsulfoxide (DMSO) before directed differentiation enhanced differentiation potential across all three germ layers. Here, we show that exposure to DMSO improves the efficiency of hPSC differentiation through Rb and by repressing downstream E2F-target genes. While transient inactivation of the Rb family members (including Rb, p107, and p130) suppresses DMSO's capacity to enhance differentiation across all germ layers, transient expression of a constitutively active (non-phosphorylatable) form of Rb increases the differentiation efficiency similar to DMSO. Inhibition of downstream targets of Rb, such as E2F signaling, also promotes differentiation of hPSCs. More generally, we demonstrate that the duration of Rb activation plays an important role in regulating differentiation capacity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Aminopiridinas/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Línea Celular , Factores de Transcripción E2F/antagonistas & inhibidores , Factores de Transcripción E2F/metabolismo , Técnicas de Silenciamiento del Gen , Estratos Germinativos/citología , Estratos Germinativos/efectos de los fármacos , Estratos Germinativos/fisiología , Humanos , Hidroxiquinolinas/farmacología , Células Madre Pluripotentes/fisiología , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Transducción de Señal/genética , Factores de Tiempo
12.
J Cell Biol ; 210(7): 1257-68, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26416968

RESUMEN

Driving human pluripotent stem cells (hPSCs) into specific lineages is an inefficient and challenging process. We show that a potent Src inhibitor, PP1, regulates expression of genes involved in the G1 to S phase transition of the cell cycle, activates proteins in the retinoblastoma family, and subsequently increases the differentiation propensities of hPSCs into all three germ layers. We further demonstrate that genetic suppression of Src regulates the activity of the retinoblastoma protein and enhances the differentiation potential of hPSCs across all germ layers. These positive effects extend beyond the initial germ layer specification and enable efficient differentiation at subsequent stages of differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Línea Celular , Fase G1/efectos de los fármacos , Humanos , Células Madre Pluripotentes/citología , Proteínas Proto-Oncogénicas pp60(c-src) , Proteína de Retinoblastoma/metabolismo , Fase S/efectos de los fármacos
13.
Nat Biotechnol ; 33(11): 1182-92, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501952

RESUMEN

Research on human pluripotent stem cells has been hampered by the lack of a standardized, quantitative, scalable assay of pluripotency. We previously described an assay called ScoreCard that used gene expression signatures to quantify differentiation efficiency. Here we report an improved version of the assay based on qPCR that enables faster, more quantitative assessment of functional pluripotency. We provide an in-depth characterization of the revised signature panel (commercially available as the TaqMan hPSC Scorecard Assay) through embryoid body and directed differentiation experiments as well as a detailed comparison to the teratoma assay. We further show that the improved ScoreCard enables a wider range of applications, such as screening of small molecules, genetic perturbations and assessment of culture conditions. Our approach can be extended beyond stem cell applications to characterize and assess the utility of other cell types and lineages.


Asunto(s)
Diferenciación Celular/genética , Biología Computacional/métodos , Cuerpos Embrioides/fisiología , Células Madre Pluripotentes/fisiología , Reacción en Cadena de la Polimerasa/métodos , Animales , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Humanos , Ratones , Neoplasias Experimentales/patología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Teratoma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA