Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Stem Cells ; 42(1): 29-41, 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-37933895

RESUMEN

Poor proliferative capacity of adult cardiomyocytes is the primary cause of heart failure after myocardial infarction (MI), thus exploring the molecules and mechanisms that promote the proliferation of adult cardiomyocytes is crucially useful for cardiac repair after MI. Here, we found that miR-130b-5p was highly expressed in mouse embryonic and neonatal hearts and able to promote cardiomyocyte proliferation both in vitro and in vivo. Mechanistic studies revealed that miR-130b-5p mainly promoted the cardiomyocyte proliferation through the MAPK-ERK signaling pathway, and the dual-specific phosphatase 6 (Dusp6), a negative regulator of the MAPK-ERK signaling, was the direct target of miR-130b-5p. Moreover, we found that overexpression of miR-130b-5p could promote the proliferation of cardiomyocytes and improve cardiac function in mice after MI. These studies thus revealed the critical role of miR-130b-5p and its targeted MAPK-ERK signaling in the cardiomyocyte proliferation of adult hearts and proved that miR-130b-5p could be a potential target for cardiac repair after MI.


Asunto(s)
MicroARNs , Infarto del Miocardio , Ratones , Animales , Miocitos Cardíacos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Transducción de Señal/genética , Proliferación Celular/genética , Apoptosis
2.
Aging Cell ; 22(4): e13794, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36797653

RESUMEN

Hippocampal neural stem cell (NSC) proliferation is known to decline with age, which is closely linked to learning and memory impairments. In the current study, we found that the expression level of miR-181a-5p was decreased in the hippocampal NSCs of aged mice and that exogenous overexpression of miR-181a-5p promoted NSC proliferation without affecting NSC differentiation into neurons and astrocytes. The mechanistic study revealed that phosphatase and tensin homolog (PTEN), a negative regulator of the AKT signaling pathway, was the target of miR-181a-5p and knockdown of PTEN could rescue the impairment of NSC proliferation caused by low miR-181a-5p levels. Moreover, overexpression of miR-181a-5p in the dentate gyrus enhanced the proliferation of NSCs and ameliorated learning and memory impairments in aged mice. Taken together, our findings indicated that miR-181a-5p played a functional role in NSC proliferation and aging-related, hippocampus-dependent learning and memory impairments.


Asunto(s)
MicroARNs , Células-Madre Neurales , Animales , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Transducción de Señal , Diferenciación Celular/genética , Células-Madre Neurales/metabolismo , Proliferación Celular/genética , Apoptosis
3.
Mol Ther Nucleic Acids ; 27: 1064-1077, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35228900

RESUMEN

Individuals with diffuse large B cell lymphoma (DLBCL) infected with hepatitis B virus (HBV) have worse chemotherapy efficacy and poorer outcomes. It is still unclear whether long noncoding RNAs (lncRNAs) serve as prognostic and therapeutic targets in the chemotherapy resistance of individuals with DLBCL and HBV infection. Here we found that the core component of HBV (HBX) directly upregulated the expression of lncNBAT1, which was closely associated with the chemotherapy outcomes of HBV-infected individuals with DLBCL. Upregulation of lncNBAT1 reduced the sensitivity of DLBCL cells to chemotherapeutic agents (methotrexate [MTX] or cytarabine [Ara-C]) that induced S phase arrest, whereas knockdown of lncNBAT1 significantly relieved the chemoresistance of HBX-expressing DLBCLs. Mechanistically, lncNBAT1 could interact with the signal transducer and activator of transcription 1 (STAT1) to prevent its enrichment at the promoter region of the functional target gene apolipoprotein B mRNA editing enzyme catalytic subunit 3A (APOBEC3A), inhibiting expression of APOBEC3A and inducing resistance to MTX in DLBCL cells. Furthermore, clinical data analysis showed that lncNBAT1 and APOBEC3A expression was closely related to the poor prognosis and short survival of individuals with DLBCL. Our findings suggest a potential prognostic marker and a candidate lncRNA target for treating HBV-infected individuals with DLBCL.

4.
EMBO Rep ; 23(2): e53015, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34927789

RESUMEN

Long noncoding RNAs (lncRNAs) are abundantly expressed in the nervous system, but their regulatory roles in neuronal differentiation are poorly understood. Using a human embryonic stem cell (hESC)-based 2D neural differentiation approach and a 3D cerebral organoid system, we show that SOX1-OT variant 1 (SOX1-OT V1), a SOX1 overlapping noncoding RNA, plays essential roles in both dorsal cortical neuron differentiation and ventral GABAergic neuron differentiation by facilitating SOX1 expression. SOX1-OT V1 physically interacts with HDAC10 through its 5' region, acts as a decoy to block HDAC10 binding to the SOX1 promoter, and thus maintains histone acetylation levels at the SOX1 promoter. SOX1 in turn activates ASCL1 expression and promotes neuronal differentiation. Taken together, we identify a SOX1-OT V1/HDAC10-SOX1-ASCL1 axis, which promotes neurogenesis, highlighting a role for lncRNAs in hESC neuronal differentiation.


Asunto(s)
Células Madre Embrionarias Humanas , Neuronas/citología , ARN Largo no Codificante , Factores de Transcripción SOXB1 , Diferenciación Celular/genética , Histona Desacetilasas/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Neuronas/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
6.
Nucleic Acids Res ; 49(4): 1935-1950, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33544864

RESUMEN

Long noncoding RNAs (lncRNAs) play a wide range of roles in the epigenetic regulation of crucial biological processes, but the functions of lncRNAs in cortical development are poorly understood. Using human embryonic stem cell (hESC)-based 2D neural differentiation approach and 3D cerebral organoid system, we identified that the lncRNA PAUPAR, which is adjacent to PAX6, plays essential roles in cortical differentiation by interacting with PAX6 to regulate the expression of a large number of neural genes. Mechanistic studies showed that PAUPAR confers PAX6 proper binding sites on the target neural genes by directly binding the genomic regions of these genes. Moreover, PAX6 recruits the histone methyltransferase NSD1 through its C-terminal PST enrichment domain, then regulate H3K36 methylation and the expression of target genes. Collectively, our data reveal that the PAUPAR/PAX6/NSD1 complex plays a critical role in the epigenetic regulation of hESC cortical differentiation and highlight the importance of PAUPAR as an intrinsic regulator of cortical differentiation.


Asunto(s)
Corteza Cerebral/metabolismo , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción PAX6/metabolismo , ARN Largo no Codificante/metabolismo , Sitios de Unión , Diferenciación Celular/genética , Células Cultivadas , Células Madre Embrionarias/citología , Eliminación de Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Metilación , Organoides , ARN Largo no Codificante/genética
7.
Neurotox Res ; 39(2): 444-455, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33025360

RESUMEN

Prenatal glucocorticoid (GC) overexposure impacts fetal hippocampal neural stem cells (NSCs) and increases the risk for relative cognitive and mood disorders in offspring. However, the precise underlying mechanisms remain elusive. Here, we treated mouse hippocampal NSCs with dexamethasone (DEX) in vitro and found that DEX inhibited cell proliferation and Sirt7 expression. In addition, prenatal mouse overexposure to DEX induced the suppression of Sirt7 in the hippocampus of offspring. Sirt7 knockdown significantly decreased the percentage of proliferating cells but did not further reduce the NSC proliferation rate in the presence of DEX, whereas Sirt7 overexpression rescued DEX-induced inhibition of hippocampal NSC proliferation. Moreover, DEX inhibited Sirt7 expression through the glucocorticoid receptor (GR), and p21 was found to mediate the functional effect of DEX-induced Sirt7 suppression. In conclusion, our data demonstrate for the first time the effect of DEX on the Sirt7-p21 pathway in hippocampal NSCs, identifying a new potential therapeutic target for prenatal GC overexposure-related neurodevelopmental disorders in offspring.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Dexametasona/toxicidad , Células-Madre Neurales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Sirtuinas/metabolismo
8.
FASEB J ; 34(10): 13257-13271, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32860269

RESUMEN

Fetal growth restriction (FGR) is a severe perinatal complication that can increase risk for mental illness. To investigate the mechanism by which FGR mice develop mental illness in adulthood, we established the FGR mouse model and the FGR mice did not display obvious depression-like behaviors, but after environmental stress exposure, FGR mice were more likely to exhibit depression-like behaviors than control mice. Moreover, FGR mice had significantly fewer dopaminergic neurons in the ventral tegmental area but no difference in serotoninergic neurons in the dorsal raphe. RNA-seq analysis showed that the downregulated genes in the midbrain of FGR mice were associated with many mental diseases and were especially involved in the regulation of NMDA-selective glutamate receptor (NMDAR) activity. Furthermore, the NMDAR antagonist memantine can relieve the stress-induced depression-like behaviors of FGR mice. In summary, our findings provide a theoretical basis for future research and treatment of FGR-related depression.


Asunto(s)
Depresión/patología , Neuronas Dopaminérgicas/patología , Retardo del Crecimiento Fetal/patología , Estrés Psicológico/patología , Área Tegmental Ventral/metabolismo , Animales , Depresión/tratamiento farmacológico , Depresión/metabolismo , Neuronas Dopaminérgicas/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/patología , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Retardo del Crecimiento Fetal/metabolismo , Masculino , Memantina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Estrés Psicológico/metabolismo , Área Tegmental Ventral/embriología , Área Tegmental Ventral/patología
9.
Cell Death Differ ; 27(2): 808-825, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31296962

RESUMEN

Aging-related cognitive ability impairments are one of the main threats to public health, and impaired hippocampal neurogenesis is a major cause of cognitive decline during aging. However, the regulation of adult neurogenesis in the hippocampus requires further study. Here, we investigated the role of microRNA-153 (miR-153), a highly conserved microRNA in mice and humans, in adult neurogenesis. During the passaging of neural stem cells (NSCs) in vitro, endogenous miR-153 expression was downregulated, with a decrease in neuronal differentiation ability. In addition, miR-153 overexpression increased the neurogenesis of NSCs. Further studies showed that miR-153 regulated neurogenesis by precisely targeting the Notch signaling pathway through inhibition of Jagged1 and Hey2 translation. In vivo analysis demonstrated that miR-153 expression was decreased in the hippocampi of aged mice with impaired cognitive ability, and that miR-153 overexpression in the hippocampus promoted neurogenesis and markedly increased the cognitive abilities of the aged mice. Overall, our findings revealed that miR-153 affected neurogenesis by regulating the Notch signaling pathway and elucidated the function of miR-153 in aging-related, hippocampus-dependent cognitive ability impairments, and neurodegenerative diseases.


Asunto(s)
Envejecimiento , Disfunción Cognitiva/metabolismo , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Receptores Notch/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Células-Madre Neurales/citología , Neurogénesis , Transducción de Señal
10.
Cancer Lett ; 419: 203-209, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29355660

RESUMEN

Glioma is one of most malignant primary tumors of the brain. However, due to a lack of effective means for diagnosing and treating glioma, the prognosis of glioma patients remains poor. Therefore, understanding the molecular mechanism of glioma progression is essential for effective treatment. Long non-coding RNAs (lncRNAs) are novel regulators of gene expression at the transcriptional, post-transcriptional and epigenetic levels. Recent evidence indicates that lncRNAs may play important roles in regulating the progression of glioma. In this article, we review the expression profile of lncRNAs in glioma and discuss the functions and known mechanisms of several representative lncRNAs in detail, as well as the prospects of lncRNAs as diagnostic and prognostic biomarkers and therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma/genética , ARN Largo no Codificante/genética , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Glioma/patología , Humanos , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA