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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(5): 945-955.e10, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525759

RESUMEN

Gene-editing technologies have made it feasible to create nonhuman primate models for human genetic disorders. Here, we report detailed genotypes and phenotypes of TALEN-edited MECP2 mutant cynomolgus monkeys serving as a model for a neurodevelopmental disorder, Rett syndrome (RTT), which is caused by loss-of-function mutations in the human MECP2 gene. Male mutant monkeys were embryonic lethal, reiterating that RTT is a disease of females. Through a battery of behavioral analyses, including primate-unique eye-tracking tests, in combination with brain imaging via MRI, we found a series of physiological, behavioral, and structural abnormalities resembling clinical manifestations of RTT. Moreover, blood transcriptome profiling revealed that mutant monkeys resembled RTT patients in immune gene dysregulation. Taken together, the stark similarity in phenotype and/or endophenotype between monkeys and patients suggested that gene-edited RTT founder monkeys would be of value for disease mechanistic studies as well as development of potential therapeutic interventions for RTT.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Animales , Encéfalo/fisiología , Cromosomas Humanos X , Ritmo Circadiano , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Edición Génica , Humanos , Macaca fascicularis , Imagen por Resonancia Magnética , Masculino , Mutación , Dolor , Síndrome de Rett/fisiopatología , Sueño , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Transcriptoma
2.
Cell ; 161(5): 1175-1186, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-26000486

RESUMEN

The scarcity of tissue-specific stem cells and the complexity of their surrounding environment have made molecular characterization of these cells particularly challenging. Through single-cell transcriptome and weighted gene co-expression network analysis (WGCNA), we uncovered molecular properties of CD133(+)/GFAP(-) ependymal (E) cells in the adult mouse forebrain neurogenic zone. Surprisingly, prominent hub genes of the gene network unique to ependymal CD133(+)/GFAP(-) quiescent cells were enriched for immune-responsive genes, as well as genes encoding receptors for angiogenic factors. Administration of vascular endothelial growth factor (VEGF) activated CD133(+) ependymal neural stem cells (NSCs), lining not only the lateral but also the fourth ventricles and, together with basic fibroblast growth factor (bFGF), elicited subsequent neural lineage differentiation and migration. This study revealed the existence of dormant ependymal NSCs throughout the ventricular surface of the CNS, as well as signals abundant after injury for their activation.


Asunto(s)
Epéndimo/citología , Células-Madre Neurales/metabolismo , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Diferenciación Celular , Movimiento Celular , Epéndimo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Glicoproteínas/metabolismo , Ratones , Células-Madre Neurales/citología , Péptidos/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
J Cell Physiol ; 238(12): 2827-2840, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37801347

RESUMEN

Growth differentiation factor 11 (GDF11) is a putative systemic rejuvenation factor. In this study, we characterized the mechanism by which GDF11 reversed aging of mesenchymal stem cells (MSCs). In culture, aged MSCs proliferate slower and are positive for senescence markers senescence-associated ß-galactosidase and P16ink4a . They have shortened telomeres, decreased GDF11 expression, and reduced osteogenic potential. GDF11 can block MSC aging in vitro and reverse age-dependent bone loss in vivo. The antiaging effect of GDF11 is via activation of the Smad2/3-PI3K-AKT-mTOR pathway. Unexpectedly, GDF11 also upregulated a DNA demethylase Tet2, which served as a key mediator for GDF11 to autoregulate itself via demethylation of the GDF11 promoter. Mutation of Tet2 facilitates MSC aging by blocking GDF11 expression. Mutagenesis of Tet2-regulated CpG sites also blocks GDF11 expression, leading to MSC aging. Together, a novel mutual regulatory relationship between GDF11 and an epigenetic factor Tet2 unveiled their antiaging roles.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas , Senescencia Celular/genética , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Humanos
4.
Proc Natl Acad Sci U S A ; 117(32): 19578-19589, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32727894

RESUMEN

The CreER/LoxP system is widely accepted to track neural lineages and study gene functions upon tamoxifen (TAM) administration. We have observed that prenatal TAM treatment caused high rates of delayed delivery and fetal mortality. This substance could produce undesired results, leading to data misinterpretation. Here, we report that administration of TAM during early stages of cortical neurogenesis promoted precocious neural differentiation, while it inhibited neural progenitor cell (NPC) proliferation. The TAM-induced inhibition of NPC proliferation led to deficits in cortical neurogenesis, dendritic morphogenesis, synaptic formation, and cortical patterning in neonatal and postnatal offspring. Mechanistically, by employing single-cell RNA-sequencing (scRNA-seq) analysis combined with in vivo and in vitro assays, we show TAM could exert these drastic effects mainly through dysregulating the Wnt-Dmrta2 signaling pathway. In adult mice, administration of TAM significantly attenuated NPC proliferation in both the subventricular zone and the dentate gyrus. This study revealed the cellular and molecular mechanisms for the adverse effects of TAM on corticogenesis, suggesting that care must be taken when using the TAM-induced CreER/LoxP system for neural lineage tracing and genetic manipulation studies in both embryonic and adult brains.


Asunto(s)
Encéfalo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/patología , Tamoxifeno/efectos adversos , Animales , Encéfalo/embriología , Encéfalo/patología , Diferenciación Celular , Proliferación Celular , Giro Dentado/efectos de los fármacos , Giro Dentado/patología , Femenino , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/patología , Ratones , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
5.
Cereb Cortex ; 31(12): 5396-5410, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34117744

RESUMEN

To explore the brain structural basis underlying the behavioral abnormalities associated with Rett syndrome (RTT), we carried out detailed longitudinal noninvasive magnetic resonance imaging analyses of RTT monkey models created by gene-editing, from weaning, through adolescence, till sexual maturation. Here, we report abnormal developmental dynamics of brain white matter (WM) microstructures and network topological organizations via diffusion tensor imaging. Specifically, disrupted WM microstructural integrity was observed at 9 months, but recovered thereafter, whereas WM network topological properties showed persistent abnormal dynamics from 9 to 37 months. Changes in the WM microstructure and WM network topology were correlated well with RTT-associated behavioral abnormalities including sleep latency, environmental exploration, and conflict encounters. Deleterious and protracted early WM myelination process likely lead to abnormal synaptic pruning, resulting in poor functional segregations. Together, this study provides initial evidence for changes in WM microstructure and network topological organization, which may underlie the neuro-patho-etilogy of RTT.


Asunto(s)
Sustancia Blanca , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen de Difusión Tensora/métodos , Haplorrinos , Fenotipo , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología
6.
Mol Cell ; 55(3): 482-94, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25018020

RESUMEN

Histone H3K4 demethylase LSD1 plays an important role in stem cell biology, especially in the maintenance of the silencing of differentiation genes. However, how the function of LSD1 is regulated and the differentiation genes are derepressed are not understood. Here, we report that elimination of LSD1 promotes embryonic stem cell (ESC) differentiation toward neural lineage. We showed that the destabilization of LSD1 occurs posttranscriptionally via the ubiquitin-proteasome pathway by an E3 ubiquitin ligase, Jade-2. We demonstrated that Jade-2 is a major LSD1 negative regulator during neurogenesis in vitro and in vivo in both mouse developing cerebral cortices and zebra fish embryos. Apparently, Jade-2-mediated degradation of LSD1 acts as an antibraking system and serves as a quick adaptive mechanism for re-establishing epigenetic landscape without more laborious transcriptional regulations. As a potential anticancer strategy, Jade-2-mediated LSD1 degradation could potentially be used in neuroblastoma cells to induce differentiation toward postmitotic neurons.


Asunto(s)
Células Madre Embrionarias/metabolismo , Histona Demetilasas/metabolismo , Neuroblastoma/metabolismo , Neurogénesis , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Histona Demetilasas/genética , Humanos , Ratones , Neuroblastoma/fisiopatología , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Ubiquitina-Proteína Ligasas/genética , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(24): E5595-E5604, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29844162

RESUMEN

Spinal cord injury (SCI) often leads to permanent loss of motor, sensory, and autonomic functions. We have previously shown that neurotrophin3 (NT3)-loaded chitosan biodegradable material allowed for prolonged slow release of NT3 for 14 weeks under physiological conditions. Here we report that NT3-loaded chitosan, when inserted into a 1-cm gap of hemisectioned and excised adult rhesus monkey thoracic spinal cord, elicited robust axonal regeneration. Labeling of cortical motor neurons indicated motor axons in the corticospinal tract not only entered the injury site within the biomaterial but also grew across the 1-cm-long lesion area and into the distal spinal cord. Through a combination of magnetic resonance diffusion tensor imaging, functional MRI, electrophysiology, and kinematics-based quantitative walking behavioral analyses, we demonstrated that NT3-chitosan enabled robust neural regeneration accompanied by motor and sensory functional recovery. Given that monkeys and humans share similar genetics and physiology, our method is likely translatable to human SCI repair.


Asunto(s)
Quitosano/farmacología , Regeneración Nerviosa/efectos de los fármacos , Neurotrofina 3/farmacología , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Axones/efectos de los fármacos , Imagen de Difusión Tensora/métodos , Femenino , Haplorrinos , Neuronas Motoras/efectos de los fármacos , Tractos Piramidales/efectos de los fármacos , Médula Espinal/efectos de los fármacos
8.
Genes Dev ; 25(7): 679-84, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21460036

RESUMEN

Recent studies have demonstrated that the Ten-eleven translocation (Tet) family proteins can enzymatically convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). While 5mC has been studied extensively, little is known about the distribution and function of 5hmC. Here we present a genome-wide profile of 5hmC in mouse embryonic stem (ES) cells. A combined analysis of global 5hmC distribution and gene expression profile in wild-type and Tet1-depleted ES cells suggests that 5hmC is enriched at both gene bodies of actively transcribed genes and extended promoter regions of Polycomb-repressed developmental regulators. Thus, our study reveals the first genome-wide 5hmC distribution in pluripotent stem cells, and supports its dual function in regulating gene expression.


Asunto(s)
Citosina/análogos & derivados , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , 5-Metilcitosina/análogos & derivados , Animales , Línea Celular , Cromatina/metabolismo , Citosina/química , Citosina/metabolismo , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo
9.
Nature ; 473(7347): 389-93, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21451524

RESUMEN

Epigenetic modification of the mammalian genome by DNA methylation (5-methylcytosine) has a profound impact on chromatin structure, gene expression and maintenance of cellular identity. The recent demonstration that members of the Ten-eleven translocation (Tet) family of proteins can convert 5-methylcytosine to 5-hydroxymethylcytosine raised the possibility that Tet proteins are capable of establishing a distinct epigenetic state. We have recently demonstrated that Tet1 is specifically expressed in murine embryonic stem (ES) cells and is required for ES cell maintenance. Using chromatin immunoprecipitation coupled with high-throughput DNA sequencing, here we show in mouse ES cells that Tet1 is preferentially bound to CpG-rich sequences at promoters of both transcriptionally active and Polycomb-repressed genes. Despite an increase in levels of DNA methylation at many Tet1-binding sites, Tet1 depletion does not lead to downregulation of all the Tet1 targets. Interestingly, although Tet1-mediated promoter hypomethylation is required for maintaining the expression of a group of transcriptionally active genes, it is also involved in repression of Polycomb-targeted developmental regulators. Tet1 contributes to silencing of this group of genes by facilitating recruitment of PRC2 to CpG-rich gene promoters. Thus, our study not only establishes a role for Tet1 in modulating DNA methylation levels at CpG-rich promoters, but also reveals a dual function of Tet1 in promoting transcription of pluripotency factors as well as participating in the repression of Polycomb-targeted developmental regulators.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Silenciador del Gen , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , 5-Metilcitosina/análogos & derivados , Animales , Línea Celular , Cromatina/metabolismo , Islas de CpG/genética , Citosina/análogos & derivados , Citosina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Ratones , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas/genética , Proteínas Represoras/metabolismo
10.
Front Immunol ; 14: 1212330, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37614232

RESUMEN

Background: Systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS) are common systemic autoimmune diseases that share a wide range of clinical manifestations and serological features. This study investigates genes, signaling pathways, and transcription factors (TFs) shared between SLE and pSS. Methods: Gene expression profiles of SLE and pSS were obtained from the Gene Expression Omnibus (GEO). Weighted gene co-expression network analysis (WGCNA) and differentially expressed gene (DEG) analysis were conducted to identify shared genes related to SLE and pSS. Overlapping genes were then subject to Gene Ontology (GO) and protein-protein interaction (PPI) network analyses. Cytoscape plugins cytoHubba and iRegulon were subsequently used to screen shared hub genes and predict TFs. In addition, gene set variation analysis (GSVA) and CIBERSORTx were used to calculate the correlations between hub genes and immune cells as well as related pathways. To confirm these results, hub genes and TFs were verified in microarray and single-cell RNA sequencing (scRNA-seq) datasets. Results: Following WGCNA and limma analysis, 152 shared genes were identified. These genes were involved in interferon (IFN) response and cytokine-mediated signaling pathway. Moreover, we screened six shared genes, namely IFI44L, ISG15, IFIT1, USP18, RSAD2 and ITGB2, out of which three genes, namely IFI44L, ISG15 and ITGB2 were found to be highly expressed in both microarray and scRNA-seq datasets. IFN response and ITGB2 signaling pathway were identified as potentially relevant pathways. In addition, STAT1 and IRF7 were identified as common TFs in both diseases. Conclusion: This study revealed IFI44L, ISG15 and ITGB2 as the shared genes and identified STAT1 and IRF7 as the common TFs of SLE and pSS. Notably, the IFN response and ITGB2 signaling pathway played vital roles in both diseases. Our study revealed common pathogenetic characteristics of SLE and pSS. The particular roles of these pivotal genes and mutually overlapping pathways may provide a basis for further mechanistic research.


Asunto(s)
Lupus Eritematoso Sistémico , Síndrome de Sjögren , Humanos , Análisis de Expresión Génica de una Sola Célula , Síndrome de Sjögren/genética , Genes Sobrepuestos , Lupus Eritematoso Sistémico/genética , Antígenos CD18 , Biología Computacional , Ubiquitina Tiolesterasa
11.
ACS Chem Neurosci ; 14(17): 3249-3264, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37583253

RESUMEN

The brain-derived neurotrophic factor (BDNF)/TrkB pathway plays a crucial role in neural plasticity and neuronal survival but is often deficient in neurodegenerative diseases like Alzheimer's disease (AD). CF3CN acts as a specific TrkB agonist that displays therapeutic effects in the AD mouse model, but its brain/plasma ratio (B/P ratio) distribution is not satisfactory. To increase its brain exposure, we synthesized several derivatives and employed nanoparticle (NP) formulation to optimize the most potent #2 derivative's in vivo PK profiles. We generated stable #2-loaded zein/lactoferrin composite NPs (#2/zein/LF) using the antisolvent co-precipitation method. In vivo PK studies revealed that nanoencapsulation improved #2's oral bioavailability by approximately 2-fold and significantly enhanced its plasma Cmax and t1/2, but the brain profiles were comparable. Pharmacodynamics showed that #2/zein/LF activates TrkB signaling that phosphorylates asparagine endopeptidase (AEP) T322 and decreases its enzymatic activity, resulting in reduced AEP-cleaved amyloid precursor protein and Tau fragments in the brains of AD mice, correlating with its PK profiles. After 3 months of treatment in 3xTg mice, #2/zein/LF decreased AD pathologies and alleviated cognitive dysfunction. Hence, zein/LF composite nanoencapsulation is a promising drug delivery method for improving the PK profiles of a potential preclinical candidate for treating neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Zeína , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Zeína/metabolismo , Zeína/farmacología , Zeína/uso terapéutico , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Receptor trkB/metabolismo
13.
Biosci Rep ; 43(1)2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36541246

RESUMEN

Hypoxic-ischemic brain injury contributes to major neurodevelopmental disorders and is one of the leading causes of seizures, which substantially results in neurodevelopmental impairments with long-lasting outcomes and is one of the main causes of death in neonates. We aimed to investigate the correlation between miRNA-210 and SCN1B, a voltage-gated sodium channel gene, in brain tissue of fetal rats with hypoxic-ischemic brain injury. We found that after 10 min of hypoxia-ischemia, all reperfusion groups showed different degrees of damage. The degree of the injury increased in all the groups after 30 min of hypoxia-ischemia. Those changes include changes in the pericellular lumen, capillaries in the cortex, erythrocytes, enlarged pericellular lumen, the enlarged pericapillary lumen in the cortex, edema around glial cells, enlarged gap to form multiple necrotic foci, deformation of neurons, and loss of cell structure. The expression levels of HIF-1α, miRNA-210, and HIF-1α mRNA were higher in the hypoxic-ischemic groups than that in the control groups, among which the expression levels in the severe group were higher than that in mild group. SCN1B is down-regulated in both the mild and severe groups, and the lowest level was found at 30 min after hypoxia in both groups. MiRNA-210 plays a role in the development of hypoxic-ischemic encephalopathy (HIE) by regulating the expression changes of SCN1B. The brain tissue of fetal rats in the hypoxic-ischemic animal model showed pathological changes of brain injury.


Asunto(s)
Lesiones Encefálicas , Hipoxia-Isquemia Encefálica , MicroARNs , Animales , Ratas , Hipoxia-Isquemia Encefálica/genética , Encéfalo/patología , Neuronas/metabolismo , Lesiones Encefálicas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
14.
ACS Nano ; 17(21): 21690-21707, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37862095

RESUMEN

Abnormal mechanical loading often leads to the progressive degradation of cartilage and causes osteoarthritis (OA). Although multiple mechanoresponsive strategies based on biomaterials have been designed to restore healthy cartilage microenvironments, methods to remotely control the on-demand mechanical forces for cartilage repair pose significant challenges. Here, a magneto-mechanically controlled mesenchymal stem cell (MSC) platform, based on the integration of intercellular mechanical communication and intracellular mechanosignaling processes, is developed for OA treatment. MSCs loaded with antioxidative melanin@Fe3O4 magnetic nanoparticles (Magcells) rapidly assemble into highly ordered cell clusters with enhanced cell-cell communication under a time-varying magnetic field, which enables long-term retention and differentiation of Magcells in the articular cavity. Subsequently, via mimicking the gait cycle, chondrogenesis can be further enhanced by the dynamic activation of mechanical signaling processes in Magcells. This sophisticated magneto-mechanical actuation strategy provides a paradigm for developing mechano-therapeutics to repair cartilage in OA treatment.


Asunto(s)
Cartílago Articular , Células Madre Mesenquimatosas , Osteoartritis , Humanos , Condrogénesis , Condrocitos/metabolismo , Osteoartritis/terapia , Diferenciación Celular
15.
Cell Rep ; 42(11): 113368, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37917581

RESUMEN

Ischemic brain injury is a severe medical condition with high incidences in elderly people without effective treatment for the resulting neural damages. Using a unilateral mouse stroke model, we analyze single-cell transcriptomes of ipsilateral and contralateral cortical penumbra regions to objectively reveal molecular events with single-cell resolution at 4 h and 1, 3, and 7 days post-injury. Here, we report that neurons are among the first cells that sense the lack of blood supplies by elevated expression of CCAAT/enhancer-binding protein ß (C/EBPß). To our surprise, the canonical inflammatory cytokine gene targets for C/EBPß, including interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNF-α), are subsequently induced also in neuronal cells. Neuronal-specific silencing of C/EBPß or IL-1ß and TNF-α substantially alleviates downstream inflammatory injury responses and is profoundly neural protective. Taken together, our findings reveal a neuronal inflammatory mechanism underlying early pathological triggers of ischemic brain injury.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular , Humanos , Ratones , Animales , Anciano , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Modelos Animales de Enfermedad , Lesiones Encefálicas/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo
16.
J Mol Neurosci ; 73(11-12): 921-931, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37864623

RESUMEN

We aimed to investigate the mechanism underlying the roles of miRNA-377, Cystathionine-ß-synthase (CBS), and hydrogen sulfide (H2S) in the development of hypoxic-ischemic encephalopathy (HIE). We investigated the relationship between CBS, H2S, and miR-377 in both humans with HIE and animals with hypoxic-ischemic insult. An animal model of fetal rats with hypoxic-ischemic brain injury was established, and the fetal rats were randomly assigned to control and hypoxic-ischemic groups for 15 min (mild) and 30 min (moderate) groups. Human samples were collected from children diagnosed with HIE. Healthy or non-neurological disease children were selected as the control group. Hematoxylin-eosin (HE) staining, quantitative real-time polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and western blot were used to conduct this study. Hypoxia-ischemia induced pathological alterations in brain tissue changes were more severe in groups with severe hypoxic insult. miRNA-377 expression levels were upregulated in brain tissue and serum of fetal rats and human samples with HIE compared to controls. Conversely, CBS and H2S expression levels were significantly decreased in both human and animal samples compared to controls. Our findings suggest that CBS is a target gene of miR-377 which may contribute to the development of HIE by regulating CBS/H2S. H2S has a protective effect against hypoxic damage in brain tissue. The study provides new insights into the potential mechanisms underlying the protective role of H2S in hypoxic brain damage and may contribute to the development of novel therapies for HIE.


Asunto(s)
Sulfuro de Hidrógeno , Hipoxia-Isquemia Encefálica , MicroARNs , Niño , Humanos , Ratas , Animales , MicroARNs/genética , MicroARNs/metabolismo , Hipoxia-Isquemia Encefálica/genética , Cistationina , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Ratas Sprague-Dawley , Sulfuro de Hidrógeno/metabolismo
17.
Cell Rep Med ; 4(11): 101286, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37951217

RESUMEN

Internal tandem duplication mutations of the FMS-like tyrosine kinase-3 (FLT3-ITDs) occur in 25%-30% of patients with acute myeloid leukemia (AML) and are associated with dismal prognosis. Although FLT3 inhibitors have demonstrated initial clinical efficacy, the overall outcome of patients with FLT3-ITD AML remains poor, highlighting the urgency to develop more effective treatment strategies. In this study, we reveal that FLT3 inhibitors reduced protein stability of the anti-cancer protein p53, resulting in drug resistance. Blocking p53 degradation with proteasome inhibitors restores intracellular p53 protein levels and, in combination with FLT3-ITD inhibitors, shows superior therapeutic effects against FLT3-ITD AML in cells, mouse models, and patients. These data suggest that this combinatorial therapeutic approach may represent a promising strategy to target FLT3-ITD AML.


Asunto(s)
Leucemia Mieloide Aguda , Proteína p53 Supresora de Tumor , Animales , Ratones , Humanos , Proteína p53 Supresora de Tumor/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutación , Pronóstico , Resultado del Tratamiento , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Tirosina Quinasa 3 Similar a fms/uso terapéutico
18.
Proc Natl Acad Sci U S A ; 106(12): 4882-7, 2009 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-19225110

RESUMEN

Mutations of MECP2 (Methyl-CpG Binding Protein 2) cause Rett syndrome. As a chromatin-associated multifunctional protein, how MeCP2 integrates external signals and regulates neuronal function remain unclear. Although neuronal activity-induced phosphorylation of MeCP2 at serine 421 (S421) has been reported, the full spectrum of MeCP2 phosphorylation together with the in vivo function of such modifications are yet to be revealed. Here, we report the identification of several MeCP2 phosphorylation sites in normal and epileptic brains from multiple species. We demonstrate that serine 80 (S80) phosphorylation of MeCP2 is critical as its mutation into alanine (S80A) in transgenic knock-in mice leads to locomotor deficits. S80A mutation attenuates MeCP2 chromatin association at several gene promoters in resting neurons and leads to transcription changes of a small number of genes. Calcium influx in neurons causes dephosphorylation at S80, potentially contributing to its dissociation from the chromatin. We postulate that phosphorylation of MeCP2 modulates its dynamic function in neurons transiting between resting and active states within neural circuits that underlie behaviors.


Asunto(s)
Cromatina/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/metabolismo , Fosfoserina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Anticuerpos Fosfo-Específicos/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Proteína 2 de Unión a Metil-CpG/química , Ratones , Datos de Secuencia Molecular , Actividad Motora , Mutación/genética , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Ratas
19.
Signal Transduct Target Ther ; 7(1): 184, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35710784

RESUMEN

Spinal cord injury (SCI) is a severe damage usually leading to limb dysesthesia, motor dysfunction, and other physiological disability. We have previously shown that NT3-chitosan could trigger an acute SCI repairment in rats and non-human primates. Due to the negative effect of inhibitory molecules in glial scar on axonal regeneration, however, the role of NT3-chitosan in the treatment of chronic SCI remains unclear. Compared with the fresh wound of acute SCI, how to handle the lesion core and glial scars is a major issue related to chronic-SCI repair. Here we report, in a chronic complete SCI rat model, establishment of magnetic resonance-diffusion tensor imaging (MR-DTI) methods to monitor spatial and temporal changes of the lesion area, which matched well with anatomical analyses. Clearance of the lesion core via suction of cystic tissues and trimming of solid scar tissues before introducing NT3-chitosan using either a rigid tubular scaffold or a soft gel form led to robust neural regeneration, which interconnected the severed ascending and descending axons and accompanied with electrophysiological and motor functional recovery. In contrast, cystic tissue extraction without scar trimming followed by NT3-chitosan injection, resulted in little, if any regeneration. Taken together, after lesion core clearance, NT3-chitosan can be used to enable chronic-SCI repair and MR-DTI-based mapping of lesion area and monitoring of ongoing regeneration can potentially be implemented in clinical studies for subacute/chronic-SCI repair.


Asunto(s)
Quitosano , Traumatismos de la Médula Espinal , Animales , Cicatriz/patología , Imagen de Difusión Tensora , Regeneración Nerviosa , Ratas , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia
20.
Pharmaceutics ; 14(3)2022 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-35335971

RESUMEN

Neural repair within the central nervous system (CNS) has been extremely challenging due to limited abilities of adult CNS neurons to regenerate, particularly in a highly inflammatory injury environment that is also filled with myelin debris. Spinal cord injury (SCI) is a serious medical condition that often leads to paralysis and currently has no effective treatment. Here we report the construction of a novel biocompatible and biodegradable material, Bio-C, through coating of acid-desalted-collagen (ADC) tube with pre-modified hyaluronic acid, which, after implantation, can elicit quite robust neural regeneration and functional recovery after complete spinal-cord transection with a 2 mm-spinal-cord-segment removal in mice. We combined morphological, electrophysiological, and objective transcriptomic analyses, in addition to behavioral analyses, to demonstrate neural tissue regeneration and functional recovery through the establishment of Bio-C-induced anti-inflammatory, neurogenic, and neurotrophic microenvironment. Through this study, we unveiled the underlying logic for CNS neural repair.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA