Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 170(5): 845-859.e19, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28823557

RESUMEN

The lateral ventricle subventricular zone (SVZ) is a frequent and consequential site of pediatric and adult glioma spread, but the cellular and molecular mechanisms mediating this are poorly understood. We demonstrate that neural precursor cell (NPC):glioma cell communication underpins this propensity of glioma to colonize the SVZ through secretion of chemoattractant signals toward which glioma cells home. Biochemical, proteomic, and functional analyses of SVZ NPC-secreted factors revealed the neurite outgrowth-promoting factor pleiotrophin, along with required binding partners SPARC/SPARCL1 and HSP90B, as key mediators of this chemoattractant effect. Pleiotrophin expression is strongly enriched in the SVZ, and pleiotrophin knock down starkly reduced glioma invasion of the SVZ in the murine brain. Pleiotrophin, in complex with the binding partners, activated glioma Rho/ROCK signaling, and ROCK inhibition decreased invasion toward SVZ NPC-secreted factors. These findings demonstrate a pathogenic role for NPC:glioma interactions and potential therapeutic targets to limit glioma invasion. PAPERCLIP.


Asunto(s)
Neoplasias Encefálicas/patología , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Glioma/patología , Ventrículos Laterales/patología , Invasividad Neoplásica/patología , Anciano , Animales , Neoplasias Encefálicas/metabolismo , Comunicación Celular , Niño , Sistemas de Liberación de Medicamentos , Femenino , Glioma/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Xenoinjertos , Humanos , Ventrículos Laterales/metabolismo , Masculino , Ratones , Trasplante de Neoplasias , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
2.
Respir Res ; 25(1): 326, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39210401

RESUMEN

OBJECTIVES: In this study, we investigated whether neural precursor cell-expressed developmentally down-regulated gene 4-like (NEDD4L) is the E3 enzyme of angiotensin-converting enzyme 2 (ACE2) and whether NEDD4L degrades ACE2 via ubiquitination, leading to the progression of pulmonary arterial hypertension (PAH). METHODS: Bioinformatic analyses were used to explore the E3 ligase that ubiquitinates ACE2. Cultured pulmonary arterial smooth muscle cells (PASMCs) and specimens from patients with PAH were used to investigate the crosstalk between NEDD4L and ACE2 and its ubiquitination in the context of PAH. RESULTS: The inhibition of ubiquitination attenuated hypoxia-induced proliferation of PASMCs. The levels of NEDD4L were increased, and those of ACE2 were decreased in lung tissues from patients with PAH and in PASMCs. NEDD4L, the E3 ligase of ACE2, inhibited the expression of ACE2 in PASMCs, possibly through ubiquitination-mediated degradation. PAH was associated with upregulation of NEDD4L expression and downregulation of ACE2 expression. CONCLUSIONS: NEDD4L, the E3 ubiquitination enzyme of ACE2, promotes the proliferation of PASMCs, ultimately leading to PAH.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Ubiquitina-Proteína Ligasas Nedd4 , Hipertensión Arterial Pulmonar , Ubiquitinación , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/biosíntesis , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/genética , Humanos , Células Cultivadas , Masculino , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Hipertensión Arterial Pulmonar/enzimología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Animales , Proliferación Celular/fisiología , Peptidil-Dipeptidasa A/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/biosíntesis , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/enzimología , Femenino , Ratas , Ratas Sprague-Dawley
3.
Calcif Tissue Int ; 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198270

RESUMEN

To investigate the potential mechanism of Morinda officinalis F. C. How polysaccharides (MOPs) in regulating osteoclast differentiation and apoptosis through miR-214-3p and its target protein. Ovariectomy was performed in 8-week female C57BL6 mice to establish the postmenopausal osteoporosis (PMOP) model. Mice were treated immediately with 500 mg/kg of MOPs (prevention group); others were treated 2 weeks after operation (treatment group). Left femur bone mineral density (BMD) was examined. RAW264.7 cells were administered with receptor activator of NF-κB ligand (RANKL) to establish the osteoclast (OC) model and treated with serum containing 1 or 2 g/kg of MOPs. Apoptosis-related indexes, miR-214-3p, and Expressed Developmentally Down-regulated 4-Like (NEDD4L) were detected by western blot, quantitative real-time-reverse transcription polymerase chain reaction (qRT-PCR), and flow cytometry. OC received a miR-214-3p inhibitor or NEDD4L small interfering RNA (siRNA). MOPs reversed the PMOP-induced changes in bones. Compared with the RANKL group, MOPs increased the apoptosis and related markers in OCs. MOPs decreased the femur miR-214-3p of PMOP mice (P < 0.001). Higher concentrations of MOPs reversed the upregulation of miR-214 mRNA in OCs (P < 0.001). miR-214-3p inhibitor increased the expression of Bax and CC3 (P < 0.01) and decreased the expression of Bcl-2 (P < 0.05). NEDD4L is targeted by miR-214. NEDD4L was upregulated in the RANKL + MOPs group (P < 0.01). miR-214-3p inhibitor increased the upregulation of NEDD4L induced by MOPs (P < 0.05). siRNA NEDD4L significantly reversed the inhibition of MOPs on osteoclast differentiation with miR-214-3p inhibitor (P < 0.01). MOPs effectively prevent PMOP by inhibiting osteoclastogenesis and inducing OC apoptosis through the miR-214-3p/NEDD4L pathway.

4.
Fish Shellfish Immunol ; 141: 109037, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37640120

RESUMEN

Mitophagy, the selective degradation of damaged mitochondria by autophagy, plays a crucial role in the survival of coelomocytes in Apostichopus japonicus following Vibrio splendidus infection by suppressing the generation of reactive oxygen species (ROS) and attenuating cell apoptosis. A recent study revealed that reducing the expression of the neural precursor cell-expressed developmentally downregulated gene 4 (NEDD4), an enzyme 3 (E3) ubiquitin ligase, significantly affects mitochondrial degradation. Prior to the present study, the functional role of NEDD4 in marine invertebrates was largely unexplored. Therefore, we investigated the role of NEDD4 in the activation of mitophagy, modulation of ROS levels, and induction of apoptosis in A. japonicus infected with V. splendidus. The results demonstrated that V. splendidus infection and lipopolysaccharide (LPS) challenge significantly increased the mRNA levels of NEDD4 in A. japonicus coelomocytes, which was consistent with changes in mitophagy under the same conditions. Knockdown of AjNEDD4 using specific small interfering RNAs (siRNAs) impaired mitophagy and caused accumulation of damaged mitochondria, as observed using transmission electron microscopy (TEM) and confocal microscopy. Furthermore, AjNEDD4 was localized to the mitochondria in both coelomocytes and HEK293T cells. Simultaneously, coelomocytes were treated with the inhibitor indole-3-carbinol (I3C) to confirm the regulatory role of AjNEDD4 in mitophagy. The accumulation of AjNEDD4 in the mitochondria and the level of mitophagy decreased. Subsequent investigations demonstrated that AjNEDD4 interacts directly with the microtubule-associated protein light chain 3 (LC3), a key regulator of autophagy and mitophagy, indicating its involvement in the mitophagy pathway. Moreover, AjNEDD4 interference hindered the interaction between AjNEDD4 and LC3, thereby impairing the engulfment and subsequent clearance of damaged mitochondria. Finally, AjNEDD4 interference led to a significant increase in intracellular ROS levels, followed by increased apoptosis. Collectively, these findings suggest that NEDD4 acts as a crucial regulator of mitophagy in A. japonicus and plays a vital role in maintaining cellular homeostasis following V. splendidus infection. NEDD4 suppresses ROS production and subsequent apoptosis by promoting mitophagy, thereby safeguarding the survival of A. japonicus under pathogenic conditions. Further investigation of the mechanisms underlying NEDD4-mediated mitophagy may provide valuable insights into the development of novel strategies for disease control in aquaculture farms.


Asunto(s)
Stichopus , Vibriosis , Vibrio , Humanos , Animales , Mitofagia/genética , Stichopus/genética , Especies Reactivas de Oxígeno/metabolismo , Células HEK293 , Vibrio/metabolismo , Vibriosis/veterinaria , Apoptosis
5.
Development ; 145(20)2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30266828

RESUMEN

As human pluripotent stem cells (hPSCs) exit pluripotency, they reportedly switch from glycolytic energy production to primarily mitochondrial metabolism. Here, we show that upon ectoderm differentiation to neural precursor cells (NPCs), hPSCs increase glycolytic rate, ultimately producing more carbon as lactate than is consumed as glucose. However, glucose, lactate and pyruvate utilization decrease to half their PSC levels by the NPC stage, establishing a more quiescent metabolic state. Furthermore, we characterize a metabolic exit event within the first 24 h of differentiation, plausibly necessary to transition hPSCs out of the pluripotent state. Contrary to current thinking, mitochondrial mass does not increase during NPC induction. Instead, mitochondrial DNA copies and mitochondrial activity decrease, suggesting that mitochondrial metabolism either requires suppression, or is not required, for nascent ectoderm differentiation. Our work, therefore, contrasts with the dogma that the hPSC state is primarily glycolytic, transitioning to an oxidative metabolism upon the loss of the pluripotent state. Instead, we show that heightened glycolytic metabolism is acquired, indicating that metabolic modulation of both glycolysis and mitochondrial metabolism occurs during exit from pluripotency in hPSCs.


Asunto(s)
Diferenciación Celular , Glucólisis , Mitocondrias/metabolismo , Neuronas/citología , Células Madre Pluripotentes/citología , Atmósfera , Carbono/farmacología , Línea Celular , Medios de Cultivo , Ectodermo/citología , Glucosa/farmacología , Glucólisis/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxígeno/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo
6.
Zhonghua Gan Zang Bing Za Zhi ; 29(2): 188-192, 2021 Feb 20.
Artículo en Zh | MEDLINE | ID: mdl-33685092

RESUMEN

Neural precursor cells-expressed developmentally down-regulated protein-8 (NEDD8) is one of the important members of the ubiquitin family, which plays an important role in maintaining cell stability, cell cycle regulation, signal transduction, transcription, and translation, DNA repair, and tumorigenesis through covalently bound substrates (also known as neddylation modification). In recent years, studies have found that the dysfunction of NEDD8 and its related enzymes is common in liver diseases, and is widely involved in the biological processes of hepatitis, liver fibrosis, proliferation, invasion, apoptosis and autophagy of liver cancer cells. This article focuses on the research progress of NEDD8 in liver diseases.


Asunto(s)
Células-Madre Neurales , Apoptosis , Humanos , Cirrosis Hepática , Transducción de Señal , Ubiquitina
7.
Am J Physiol Renal Physiol ; 318(1): F1-F13, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31657249

RESUMEN

Renal Na+ reabsorption, facilitated by the epithelial Na+ channel (ENaC), is subject to multiple forms of control to ensure optimal body blood volume and pressure through altering both the ENaC population and activity at the cell surface. Here, the focus is on regulating the number of ENaCs present in the apical membrane domain through pathways of ENaC synthesis and targeting to the apical membrane as well as ENaC removal, recycling, and degradation. Finally, the mechanisms by which ENaC trafficking pathways are regulated are summarized.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Riñón/metabolismo , Transporte de Proteínas/fisiología , Animales , Humanos , Sodio/metabolismo
8.
Exp Mol Pathol ; 115: 104452, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32413360

RESUMEN

BACKGROUND: Hypospadias, as a congenital disorder of the urethra, is the second most common birth abnormality of the male reproductive system. This study primarily investigates the effects of microRNA-494 (miR-494) on the transforming growth factor-ß1 (TGF-ß1)/Smads signaling pathway and on the development of hypospadias by binding to neural precursor cell expressed developmentally downregulated gene 4-like (Nedd4L). METHODS: We induced a mouse model of hypospadias through di-(2-ethylhexyl) phthalate treatment. The underlying regulatory mechanisms of miR-494 in this model were analyzed upon treatment of miR-494 mimic, miR-494 inhibitor, or small interfering RNA against Nedd4L in urethral epithelial cells isolated from mice with hypospadias. We then verified the binding site between miR-494 and Nedd4L and applied a gain- and loss-of-function approach to determine the effects of miR-494 on cell proliferation, cycle distribution, and apoptosis. RESULTS: Male mice with hypospadias exhibited significantly higher miR-494 expression and lower Nedd4L expression in urethral tissues than normal male mice. Nedd4L was verified as a target gene of miR-494. Treatment with miR-494 inhibitor suppressed the activation of the TGF-ß1/Smads signaling pathway, whereas down-regulation of miR-494 exerted protective effects on urethral epithelial cells by impeding cell proliferation and inducing cell apoptosis. CONCLUSIONS: The study indicates that downregulation of miR-494 inhibits the TGF-ß1/Smads signaling pathway and prevents the development of hypospadias through upregulating Nedd4L.


Asunto(s)
Regulación hacia Abajo/genética , Hipospadias/genética , Hipospadias/prevención & control , MicroARNs/genética , Ubiquitina-Proteína Ligasas Nedd4/genética , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/genética , Animales , Apoptosis/genética , Secuencia de Bases , Ciclo Celular/genética , Proliferación Celular/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Hipospadias/patología , Masculino , Ratones , MicroARNs/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Uretra/patología
9.
Am J Physiol Renal Physiol ; 317(6): F1513-F1525, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31566435

RESUMEN

The metabolic sensor AMP-activated protein kinase (AMPK) inhibits the epithelial Na+ channel (ENaC), a key regulator of salt reabsorption by the kidney and thus total body volume and blood pressure. Recent studies have suggested that AMPK promotes the association of p21-activated kinase-interacting exchange factor-ß1 ß1Pix, 14-3-3 proteins, and the ubiquitin ligase neural precursor cell expressed developmentally downregulated protein (Nedd)4-2 into a complex that inhibits ENaC by enhancing Nedd4-2 binding to ENaC and ENaC degradation. Functional ß1Pix is required for ENaC inhibition by AMPK and promotes Nedd4-2 phosphorylation and stability in mouse kidney cortical collecting duct cells. Here, we report that AMPK directly phosphorylates ß1Pix in vitro. Among several AMPK phosphorylation sites on ß1Pix detected by mass spectrometry, Ser71 was validated as functionally significant. Compared with wild-type ß1Pix, overexpression of a phosphorylation-deficient ß1Pix-S71A mutant attenuated ENaC inhibition and the AMPK-activated interaction of both ß1Pix and Nedd4-2 to 14-3-3 proteins in cortical collecting duct cells. Similarly, overexpression of a ß1Pix-Δ602-611 deletion tract mutant unable to bind 14-3-3 proteins decreased the interaction between Nedd4-2 and 14-3-3 proteins, suggesting that 14-3-3 binding to ß1Pix is critical for the formation of a ß1Pix/Nedd4-2/14-3-3 complex. With expression of a general peptide inhibitor of 14-3-3-target protein interactions (R18), binding of both ß1Pix and Nedd4-2 to 14-3-3 proteins was reduced, and AMPK-dependent ENaC inhibition was also attenuated. Altogether, our results demonstrate the importance of AMPK-mediated phosphorylation of ß1Pix at Ser71, which promotes 14-3-3 interactions with ß1Pix and Nedd4-2 to form a tripartite ENaC inhibitory complex, in the mechanism of ENaC regulation by AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Riñón/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Regulación Enzimológica de la Expresión Génica/genética , Células HEK293 , Humanos , Túbulos Renales Colectores/metabolismo , Ratones , Mutación/genética , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Fosforilación , Factores de Intercambio de Guanina Nucleótido Rho/genética
10.
Neurosurg Focus ; 46(3): E9, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30835678

RESUMEN

Spinal cord injury (SCI) has been associated with a dismal prognosis-recovery is not expected, and the most standard interventions have been temporizing measures that do little to mitigate the extent of damage. While advances in surgical and medical techniques have certainly improved this outlook, limitations in functional recovery continue to impede clinically significant improvements. These limitations are dependent on evolving immunological mechanisms that shape the cellular environment at the site of SCI. In this review, we examine these mechanisms, identify relevant cellular components, and discuss emerging treatments in stem cell grafts and adjuvant immunosuppressants that target these pathways. As the field advances, we expect that stem cell grafts and these adjuvant treatments will significantly shift therapeutic approaches to acute SCI with the potential for more promising outcomes.


Asunto(s)
Rechazo de Injerto/prevención & control , Enfermedad Injerto contra Huésped/prevención & control , Inmunosupresores/uso terapéutico , Células Madre Pluripotentes Inducidas/trasplante , Células Precursoras de Oligodendrocitos/trasplante , Traumatismos de la Médula Espinal/terapia , Adyuvantes Inmunológicos , Aloinjertos , Animales , Basiliximab/uso terapéutico , Células Cultivadas , Ensayos Clínicos como Asunto , Ciclosporina/uso terapéutico , Femenino , Supervivencia de Injerto/inmunología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/inmunología , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Masculino , Ratones , Ácido Micofenólico/uso terapéutico , Células Precursoras de Oligodendrocitos/inmunología , Ratas , Tacrolimus/uso terapéutico , Trasplante Autólogo
11.
Int J Mol Sci ; 20(11)2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31159418

RESUMEN

Human tonsil-derived mesenchymal stem cells (T-MSCs) are newly identified MSCs and present typical features of MSCs, including having the differentiation capacity into the three germ layers and excellent proliferation capacity. They are easily sourced and are useful for stem cell therapy in various disease states. We previously reported that T-MSCs could be differentiated into skeletal myocytes and Schwann-like cells; therefore, they are a promising candidate for cell therapies for neuromuscular disease. Motor neurons (MNs), which regulate spontaneous behavior, are affected by a wide range of MN diseases (MNDs) for which there are no effective remedies. We investigated the differentiation potential of MN-like cells derived from T-MSCs (T-MSC-MNCs) for application to therapy of MNDs. After the process of MN differentiation, the expression of MN-related markers, including Islet 1, HB9/HLXB9 (HB9), and choline acetyltransferase (ChAT), was increased when compared with undifferentiated T-MSCs. The secretion of acetylcholine to the conditioned medium was significantly increased after MN differentiation. We cocultured T-MSC-MNCs and human skeletal muscle cells, and confirmed the presence of the acetylcholine receptor clusters, which demonstrated the formation of neuromuscular junctions. The potential functional improvements afforded by these T-MSC-MNCs could be useful in the treatment of MNDs caused by genetic mutation, viral infection, or environmental problems.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Unión Neuromuscular/fisiología , Tonsila Palatina/citología , Acetilcolina/metabolismo , Biomarcadores , Células Cultivadas , Expresión Génica , Humanos , Inmunohistoquímica , Fibras Musculares Esqueléticas/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
12.
Glia ; 66(5): 987-998, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29380422

RESUMEN

Previously, we determined microRNA-31 (miR-31) is a noncoding tumor suppressive gene frequently deleted in glioblastoma (GBM); miR-31 suppresses tumor growth, in part, by limiting the activity of NF-κB. Herein, we expand our previous studies by characterizing the role of miR-31 during neural precursor cell (NPC) to astrocyte differentiation. We demonstrate that miR-31 expression and activity is suppressed in NPCs by stem cell factors such as Lin28, c-Myc, SOX2 and Oct4. However, during astrocytogenesis, miR-31 is induced by STAT3 and SMAD1/5/8, which mediate astrocyte differentiation. We determined miR-31 is required for terminal astrocyte differentiation, and that the loss of miR-31 impairs this process and/or prevents astrocyte maturation. We demonstrate that miR-31 promotes astrocyte development, in part, by reducing the levels of Lin28, a stem cell factor implicated in NPC renewal. These data suggest that miR-31 deletions may disrupt astrocyte development and/or homeostasis.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Immunoblotting , Hibridación in Situ , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Xenopus laevis
13.
Mol Ther ; 25(9): 2028-2037, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28705346

RESUMEN

Generation of functional dopamine (DA) neurons is an essential step for the development of effective cell therapy for Parkinson's disease (PD). The generation of DA neurons can be accomplished by overexpression of DA-inducible genes using virus- or DNA-based gene delivery methods. However, these gene delivery methods often cause chromosomal anomalies. In contrast, mRNA-based gene delivery avoids this problem and therefore is considered safe to use in the development of cell-based therapy. Thus, we used mRNA-based gene delivery method to generate safe DA neurons. In this study, we generated transformation-free DA neurons by transfection of mRNA encoding DA-inducible genes Nurr1 and FoxA2. The delivery of mRNA encoding dopaminergic fate inducing genes proved sufficient to induce naive rat forebrain precursor cells to differentiate into neurons exhibiting the biochemical, electrophysiological, and functional properties of DA neurons in vitro. Additionally, the generation efficiency of DA neurons was improved by the addition of small molecules, db-cAMP, and the adjustment of transfection timing. The successful generation of DA neurons using an mRNA-based method offers the possibility of developing clinical-grade cell sources for neuronal cell replacement treatment for PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , ARN Mensajero/síntesis química , ARN Mensajero/genética , Factores de Transcripción/genética , Animales , Línea Celular , Neuronas Dopaminérgicas/citología , Expresión Génica , Regulación de la Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/genética , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Ratas , Transfección , Tirosina 3-Monooxigenasa/genética
14.
J Cell Mol Med ; 21(8): 1605-1618, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28244646

RESUMEN

Human embryonic stem cells (hESCs) can self-renew and differentiate into all cell lineages. E2 is known to exhibit positive effects on embryo development. Although the importance of E2 in many physiological processes has been reported, to date few researchers have investigated the effects of E2 on hESCs differentiation. We studied the effects of E2 on dopamine (DA) neuron induction of hESCs and its related signalling pathways using the three-stage protocol. In our study, 0.1 µM E2 were applied to hESCs-derived human embryoid bodies (hEBs) and effects of E2 on neural cells differentiation were investigated. Protein and mRNA level assay indicated that E2 up-regulated the expression of insulin-like growth factors (IGF)-1, ectoderm, neural precursor cells (NPC) and DA neuron markers, respectively. The population of hESC-derived NPCs and DA neurons was increased to 92% and 93% to that of DMSO group, respectively. Furthermore, yield of DA neuron-secreted tyrosine hydroxylase (TH) and dopamine was also increased. E2-caused promotion was relieved in single inhibitor (ICI or JB1) group partly, and E2 effects were repressed more stronger in inhibitors combination (ICI plus JB1) group than in single inhibitor group at hEBs, hNPCs and hDA neurons stages. Owing to oestrogen receptors regulate multiple brain functions, when single or two inhibitors were used to treat neural differentiation stage, we found that oestrogen receptor (ER)ß but not ERα is strongly repressed at the hNPCs and hDA neurons stage. These findings, for the first time, demonstrate the molecular cascade and related cell biology events involved in E2-improved hNPC and hDA neuron differentiation through cross-talk between IGF-1 and ERß in vitro.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células-Madre Neurales/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oligopéptidos/farmacología , Transducción de Señal , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
Development ; 141(22): 4343-53, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25344075

RESUMEN

In the developing neocortex, neural precursor cells (NPCs) sequentially generate various neuronal subtypes in a defined order. Although the precise timing of the NPC fate switches is essential for determining the number of neurons of each subtype and for precisely generating the cortical layer structure, the molecular mechanisms underlying these switches are largely unknown. Here, we show that epigenetic regulation through Ring1B, an essential component of polycomb group (PcG) complex proteins, plays a key role in terminating NPC-mediated production of subcerebral projection neurons (SCPNs). The level of histone H3 residue K27 trimethylation at and Ring1B binding to the promoter of Fezf2, a fate determinant of SCPNs, increased in NPCs as Fezf2 expression decreased. Moreover, deletion of Ring1B in NPCs, but not in postmitotic neurons, prolonged the expression of Fezf2 and the generation of SCPNs that were positive for CTIP2. These results indicate that Ring1B mediates the timed termination of Fezf2 expression and thereby regulates the number of SCPNs.


Asunto(s)
Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neocórtex/embriología , Neurogénesis/fisiología , Neuronas/fisiología , Complejo Represivo Polycomb 1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Bromodesoxiuridina , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/genética , Inmunohistoquímica , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuronas/citología , ARN Interferente Pequeño/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/metabolismo
16.
Development ; 141(4): 761-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24496614

RESUMEN

Understanding how dopamine (DA) phenotypes are acquired in midbrain DA (mDA) neuron development is important for bioassays and cell replacement therapy for mDA neuron-associated disorders. Here, we demonstrate a feed-forward mechanism of mDA neuron development involving Nurr1 and Foxa2. Nurr1 acts as a transcription factor for DA phenotype gene expression. However, Nurr1-mediated DA gene expression was inactivated by forming a protein complex with CoREST, and then recruiting histone deacetylase 1 (Hdac1), an enzyme catalyzing histone deacetylation, to DA gene promoters. Co-expression of Nurr1 and Foxa2 was established in mDA neuron precursor cells by a positive cross-regulatory loop. In the presence of Foxa2, the Nurr1-CoREST interaction was diminished (by competitive formation of the Nurr1-Foxa2 activator complex), and CoREST-Hdac1 proteins were less enriched in DA gene promoters. Consequently, histone 3 acetylation (H3Ac), which is responsible for open chromatin structures, was strikingly increased at DA phenotype gene promoters. These data establish the interplay of Nurr1 and Foxa2 as the crucial determinant for DA phenotype acquisition during mDA neuron development.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/fisiología , Factor Nuclear 3-beta del Hepatocito/metabolismo , Mesencéfalo/citología , Neurogénesis/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Análisis de Varianza , Animales , Inmunoprecipitación de Cromatina , Proteínas Co-Represoras , Neuronas Dopaminérgicas/metabolismo , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Histona Desacetilasa 1/metabolismo , Inmunoprecipitación , Ratones , Análisis por Micromatrices , Proteínas del Tejido Nervioso/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/metabolismo , Retroviridae , Transducción Genética
17.
Cell Mol Neurobiol ; 37(5): 899-909, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27655030

RESUMEN

Overexpression of suppressor of cytokine signalling 2 (SOCS2) has been shown to promote hippocampal neurogenesis in vivo and promote neurite outgrowth of neurons in vitro. In the adult mouse brain, SOCS2 is most highly expressed in the hippocampal CA3 region and at lower levels in the dentate gyrus, an expression pattern that suggests a role in adult neurogenesis. Herein we examine generation of neuroblasts and their maturation into more mature neurons in SOCS2 null (SOCS2KO) mice. EdU was administered for 7 days to label proliferative neural precursor cells. The number of EdU-labelled doublecortin+ neuroblasts and NeuN+ mature neurons they generated was examined at day 8 and day 35, respectively. While no effect of SOCS2 deletion was observed in neuroblast generation, it reduced the numbers of EdU-labelled mature newborn neurons at 35 days. As SOCS2 regulates neurite outgrowth and dentate granule neurons project to the CA3 region, alterations in dendritic arborisation or spine formation may have correlated with the decreased numbers of EdU-labelled newborn neurons. SOCS2KO mice were crossed with Nes-CreERT2/mTmG mice, in which membrane eGFP is inducibly expressed in neural precursor cells and their progeny, and the dendrite and dendritic spine morphology of newborn neurons were examined at 35 days. SOCS2 deletion had no effect on total dendrite length, number of dendritic segments, number of branch points or total dendritic spine density but increased the number of mature "mushroom" spines. Our results suggest that endogenous SOCS2 regulates numbers of EdU-labelled mature newborn adult hippocampal neurons, possibly by mediating their survival and that this may be via a mechanism regulating dendritic spine maturation.


Asunto(s)
Envejecimiento/metabolismo , Diferenciación Celular , Espinas Dendríticas/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Neurogénesis , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Animales Recién Nacidos , Supervivencia Celular , Giro Dentado/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Proc Natl Acad Sci U S A ; 111(28): E2885-94, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982138

RESUMEN

Human oligodendrocyte progenitor cell (OPC) specification and differentiation occurs slowly and limits the potential for cell-based treatment of demyelinating disease. In this study, using FACS-based isolation and microarray analysis, we identified a set of transcription factors expressed by human primary CD140a(+)O4(+) OPCs relative to CD133(+)CD140a(-) neural stem/progenitor cells (NPCs). Among these, lentiviral overexpression of transcription factors ASCL1, SOX10, and NKX2.2 in NPCs was sufficient to induce Sox10 enhancer activity, OPC mRNA, and protein expression consistent with OPC fate; however, unlike ASCL1 and NKX2.2, only the transcriptome of SOX10-infected NPCs was induced to a human OPC gene expression signature. Furthermore, only SOX10 promoted oligodendrocyte commitment, and did so at quantitatively equivalent levels to native OPCs. In xenografts of shiverer/rag2 animals, SOX10 increased the rate of mature oligodendrocyte differentiation and axon ensheathment. Thus, SOX10 appears to be the principle and rate-limiting regulator of myelinogenic fate from human NPCs.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Elementos de Facilitación Genéticos , Xenoinjertos , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Lentivirus , Ratones , Células-Madre Neurales/citología , Proteínas Nucleares , Oligodendroglía/citología , Trasplante de Células Madre , Factores de Transcripción/genética , Transcriptoma , Transducción Genética
19.
Int J Mol Sci ; 18(8)2017 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-28825630

RESUMEN

Impairment of mucociliary clearance with reduced airway fluid secretion leads to chronically inflamed airways. Cystic fibrosis transmembrane conductance regulator (CFTR) is crucially involved in airway fluid secretion and dexamethasone (dexa) has previously been shown to elevate CFTR activity in airway epithelial cells. However, the pathway by which dexa increases CFTR activity is largely unknown. We aimed to determine whether the increase of CFTR activity by dexa is achieved by non-genomic signaling and hypothesized that the phosphoinositide 3-kinase (PI3K) pathway is involved in CFTR stimulation. Primary rat airway epithelial cells and human bronchial submucosal gland-derived Calu-3 cells were analyzed in Ussing chambers and kinase activation was determined by Western blots. Results demonstrated a critical involvement of PI3K and protein kinase B (AKT) signaling in the dexa-induced increase of CFTR activity, while serum and glucocorticoid dependent kinase 1 (SGK1) activity was not essential. We further demonstrated a reduced neural precursor cell expressed, developmentally downregulated 4-like (NEDD4L) ubiquitin E3 ligase activity induced by dexa, possibly responsible for the elevated CFTR activity. Finally, increases of CFTR activity by dexa were demonstrated within 30 min accompanied by rapid activation of AKT. In conclusion, dexa induces a rapid stimulation of CFTR activity which depends on PI3K/AKT signaling in airway epithelial cells. Glucocorticoids might thus represent, in addition to their immunomodulatory actions, a therapeutic strategy to rapidly increase airway fluid secretion.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Dexametasona/administración & dosificación , Transducción de Señal/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Ratas
20.
Biochim Biophys Acta ; 1852(7): 1298-310, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25817400

RESUMEN

Ethanol (EtOH) exposure during early postnatal life triggers obvious neurotoxic effects on the developing hippocampus and results in long-term effects on hippocampal neurogenesis. Resveratrol (RSV) has been demonstrated to exert potential neuroprotective effects by promoting hippocampal neurogenesis. However, the effects of RSV on the EtOH-mediated impairment of hippocampal neurogenesis remain undetermined. Thus, mice were pretreated with RSV and were later exposed to EtOH to evaluate its protective effects on EtOH-mediated toxicity during hippocampal development. The results indicated that a brief exposure of EtOH on postnatal day 7 resulted in a significant impairment in hippocampal neurogenesis and a depletion of hippocampal neural precursor cells (NPCs). This effect was attenuated by pretreatment with RSV. Furthermore, EtOH exposure resulted in a reduction in spine density on the granular neurons of the dentate gyrus (DG), and the spines exhibited a less mature morphological phenotype characterized by a higher proportion of stubby spines and a lower proportion of mushroom spines. However, RSV treatment effectively reversed these responses. We further confirmed that RSV treatment reversed the EtOH-induced down-regulation of hippocampal pERK and Hes1 protein levels, which may be related to the proliferation and maintenance of NPCs. Furthermore, EtOH exposure in the C17.2 NPCs also diminished cell proliferation and activated apoptosis, which could be reversed by pretreatment of RSV. Overall, our results suggest that RSV pretreatment protects against EtOH-induced defects in neurogenesis in postnatal mice and may thus play a critical role in preventing EtOH-mediated toxicity in the developing hippocampus.


Asunto(s)
Etanol/toxicidad , Hipocampo/efectos de los fármacos , Neurogénesis , Fármacos Neuroprotectores/farmacología , Estilbenos/farmacología , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Resveratrol , Factor de Transcripción HES-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA