Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nanomaterials (Basel) ; 14(13)2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38998688

RESUMEN

The impact of oral administration of mechanically fibrillated cellulose nanofibers (fib-CNF), a commonly used nanofiber, on toxicity and health remains unclear, despite reports of the safety and beneficial effects of chitin-based nanofibers. Thus, evaluating the oral toxicity of fib-CNF in accordance with OECD Test Guideline 407 (TG407) is essential. This study aimed to assess the safety of orally administered fib-CNF through an acute toxicity study in rats, following the OECD TG407 guidelines for 4 weeks. CNF "BiNFi-s" FMa-10005, derived from mechanically fibrillated pulp cellulose, was administered via gavage to male and female Crl:CD(SD) rats at doses of 50, 150, 500, and 1000 mg/kg/day for 28 days, with a control group receiving water for injection. The study evaluated the toxic effects of repeated administration, and the rats were monitored for an additional 14 days post-administration to assess recovery from any toxic effects. The results showed no mortality in either sex during the administration period, and no toxicological effects related to the test substance were observed in various assessments, including general condition and behavioral function observations, urinalysis, hematological examination, blood biochemical examination, necropsy findings, organ weights, and histopathological examination. Notably, only female rats treated with 1000 mg/kg/day of CNF exhibited a consistent reduction in body weight during the 14-day recovery period after the end of treatment. They also showed a slight decrease in pituitary and liver weights. However, hematological and blood biochemical tests did not reveal significant differences, suggesting a potential weight-suppressive effect of CNF ingestion.

2.
FEBS Open Bio ; 14(6): 906-921, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38604990

RESUMEN

The Ras homology (Rho) family of GTPases serves various functions, including promotion of cell migration, adhesion, and transcription, through activation of effector molecule targets. One such pair of effectors, the Rho-associated coiled-coil kinases (ROCK1 and ROCK2), induce reorganization of actin cytoskeleton and focal adhesion through substrate phosphorylation. Studies on ROCK knockout mice have confirmed that ROCK proteins are essential for embryonic development, but their physiological functions in adult mice remain unknown. In this study, we aimed to examine the roles of ROCK1 and ROCK2 proteins in normal adult mice. Tamoxifen (TAM)-inducible ROCK1 and ROCK2 single and double knockout mice (ROCK1flox/flox and/or ROCK2flox/flox;Ubc-CreERT2) were generated and administered a 5-day course of TAM. No deaths occurred in either of the single knockout strains, whereas all of the ROCK1/ROCK2 double conditional knockout mice (DcKO) had died by Day 11 following the TAM course. DcKO mice exhibited increased lung tissue vascular permeability, thickening of alveolar walls, and a decrease in percutaneous oxygen saturation compared with noninducible ROCK1/ROCK2 double-floxed control mice. On Day 3 post-TAM, there was a decrease in phalloidin staining in the lungs in DcKO mice. On Day 5 post-TAM, immunohistochemical analysis also revealed reduced staining for vascular endothelial (VE)-cadherin, ß-catenin, and p120-catenin at cell-cell contact sites in vascular endothelial cells in DcKO mice. Additionally, VE-cadherin/ß-catenin complexes were decreased in DcKO mice, indicating that ROCK proteins play a crucial role in maintaining lung function by regulating cell-cell adhesion.


Asunto(s)
Células Endoteliales , Ratones Noqueados , Quinasas Asociadas a rho , Animales , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/genética , Ratones , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Pulmón/metabolismo , Pulmón/patología , Cadherinas/metabolismo , Cadherinas/genética , beta Catenina/metabolismo , beta Catenina/genética , Masculino , Antígenos CD
3.
Toxics ; 12(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38393216

RESUMEN

Mechanically fibrillated cellulose nanofibers, known as fib-CNF (fiber length: 500 nm; diameter: 45 nm), are used in composites and as a natural thickener in foods. To evaluate their safety, we conducted a 28-day study in mice with inhalation exposure at 0.2 mg/body and oral administration of 400 mg/kg/day. Inhalation exposure to fib-CNF caused transient weight loss, changes in blood cell counts, and increased lung weights. These changes were attributed to adaptive responses. The oral administration of fib-CNF for 28 days resulted in no apparent toxic effects except for a slight decrease in platelet counts. The fib-CNF administration using the protocols studied appears to be safe in mice.

4.
Biochem Biophys Res Commun ; 643: 55-60, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586159

RESUMEN

The cell motility of smooth muscle cells (SMCs) is essential for vascular and internal organ development and tissue regeneration in response to damage. Cell migration requires dynamic changes in the actin-cytoskeleton via the p-21 activated kinase (Pak)-Cofilin signaling cascade, which is the central axis of the actin filaments. We previously identified that the Inka2 gene was preferentially expressed in the central nervous system (CNS) and revealed that Inka2 directly binds Pak4 to suppress its kinase activity, thereby regulating actin de-polymerization in dendritic spine formation of the forebrain neurons. However, its physiological significance outside the CNS remains unclear. Here we determined the Inka2 expression profile in various organs using in situ hybridization analysis and lacZ staining on Inka2flox/+ mice. Robust Inka2 expression was consistently detected in the SMCs of many peripheral organs, including the arteries, esophagus, stomach, intestine, and bladder. The scratch assay was used on primary cultured SMCs and revealed that Inka2-/- SMC exhibits accelerated cell migration ability without a change in the cell proliferation rate. Inka2-/- SMCs displayed Cofilin activation/phosphorylation, a downstream molecule of Pak4 signal cascade. These results suggest that Inka2 regulates SMC motility through modulating actin reorganization as the endogenous inhibitor of Pak4.


Asunto(s)
Actinas , Miocitos del Músculo Liso , Animales , Ratones , Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Movimiento Celular/fisiología , Células Cultivadas , Miocitos del Músculo Liso/metabolismo
5.
PLoS Genet ; 18(10): e1010438, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36301793

RESUMEN

The actin filament is a fundamental part of the cytoskeleton defining cell morphology and regulating various physiological processes, including filopodia formation and dendritic spinogenesis of neurons. Serine/threonine-protein kinase Pak4, an essential effector, links Rho GTPases to control actin polymerization. Previously, we identified the Inka2 gene, a novel mammalian protein exhibiting sequence similarity to Inka1, which serves as a possible inhibitor for Pak4. Although Inka2 is dominantly expressed in the nervous system and involved in focal-adhesion dynamics, its molecular role remains unclear. Here, we found that Inka2-iBox directly binds to Pak4 catalytic domain to suppress actin polymerization. Inka2 promoted actin depolymerization and inhibited the formation of cellular protrusion caused by Pak4 activation. We further generated the conditional knockout mice of the Inka2 gene. The beta-galactosidase reporter indicated the preferential Inka2 expression in the dorsal forebrain neurons. Cortical pyramidal neurons of Inka2-/- mice exhibited decreased density and aberrant morphology of dendritic spines with marked activation/phosphorylation of downstream molecules of Pak4 signal cascade, including LIMK and Cofilin. These results uncovered the unexpected function of endogenous Pak4 inhibitor in neurons. Unlike Inka1, Inka2 is a critical mediator for actin reorganization required for dendritic spine development.


Asunto(s)
Actinas , Proteínas Adaptadoras Transductoras de Señales , Neurogénesis , Quinasas p21 Activadas , Animales , Ratones , Actinas/genética , Actinas/metabolismo , Citoesqueleto/metabolismo , Fosforilación , Ratones Noqueados , Quinasas p21 Activadas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
6.
Biol Psychiatry ; 91(9): 821-831, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35219498

RESUMEN

BACKGROUND: IQSEC3, a gephyrin-binding GABAergic (gamma-aminobutyric acidergic) synapse-specific guanine nucleotide exchange factor, was recently reported to regulate activity-dependent GABAergic synapse maturation, but the underlying signaling mechanisms remain incompletely understood. METHODS: We generated mice with conditional knockout (cKO) of Iqsec3 to examine whether altered synaptic inhibition influences hippocampus-dependent fear memory formation. In addition, electrophysiological recordings, immunohistochemistry, and behavioral assays were used to address our question. RESULTS: We found that Iqsec3-cKO induces a specific reduction in GABAergic synapse density, GABAergic synaptic transmission, and maintenance of long-term potentiation in the hippocampal CA1 region. In addition, Iqsec3-cKO mice exhibited impaired fear memory formation. Strikingly, Iqsec3-cKO caused abnormally enhanced activation of ribosomal P70-S6K1-mediated signaling in the hippocampus but not in the cortex. Furthermore, inhibiting upregulated S6K1 signaling by expressing dominant-negative S6K1 in the hippocampal CA1 of Iqsec3-cKO mice completely rescued impaired fear learning and inhibitory synapse density but not deficits in long-term potentiation maintenance. Finally, upregulated S6K1 signaling was rescued by IQSEC3 wild-type, but not by an ARF-GEF (adenosine diphosphate ribosylation factor-guanine nucleotide exchange factor) inactive IQSEC3 mutant. CONCLUSIONS: Our results suggest that IQSEC3-mediated balanced synaptic inhibition in hippocampal CA1 is critical for the proper formation of hippocampus-dependent fear memory.


Asunto(s)
Miedo , Factores de Intercambio de Guanina Nucleótido , Hipocampo , Sinapsis , Animales , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sinapsis/metabolismo , Regulación hacia Arriba
7.
Biochem Biophys Res Commun ; 556: 192-198, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33845309

RESUMEN

Helicobacter pylori (H. pylori) infection mainly causes gastroduodenal diseases, including chronic gastritis, peptic ulcer disease and gastric cancer. In recent years, several studies have demonstrated that infection with H. pylori, especially strains harboring the virulence factor CagA (cytotoxin-associated gene A), contribute to the development of non-gastric systemic diseases, including hypercholesterolemia and atherosclerotic cardiovascular diseases. However, mechanisms underlying this association has not been defined. In this study, we carried out a large-scale genetic screen using Drosophila and identified a novel CagA target low-density lipoprotein receptor (LDLR), which aids in the clearance of circulating LDL. We showed that CagA physically interacted with LDLR via its carboxy-terminal region and inhibited LDLR-mediated LDL uptake into cells. Since deficiency of LDLR-mediated LDL uptake has been known to increase plasma LDL and accelerate atherosclerosis, our findings may provide a novel mechanism for the association between infection with CagA-positive H. pylori and hypercholesterolemia leading to atherosclerotic cardiovascular diseases.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidad , Lipoproteínas LDL/metabolismo , Receptores de LDL/metabolismo , Factores de Virulencia/metabolismo , Animales , Animales Modificados Genéticamente , Aterosclerosis/microbiología , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ojo/metabolismo , Femenino , Humanos , Hipercolesterolemia/microbiología , Lipoproteínas LDL/sangre , Masculino , Unión Proteica
8.
Cancer Res ; 76(9): 2612-25, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26941286

RESUMEN

The progression from precursor lesions of pancreatic cancer, including pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasm (IPMN), to invasive disease is characterized by stepwise accumulation of genetic alterations. However, it remains unclear whether additional alterations are required for the progression of high-grade neoplasms to invasive pancreatic carcinoma. We compared the genomic profiles of paired noninvasive and invasive carcinoma tissues collected from patients with IPMN. We demonstrate that the frequency of genomic copy-number aberrations significantly increased during the course of invasion, and the loss of 8p11.22-ter was more often associated with invasive tissues. Expression profiling in pancreatic cancer cell lines with and without 8p11.22-ter revealed that DUSP4, an MAPK phosphatase, was significantly downregulated in cells lacking 8p11.22-ter as well as in invasive carcinomas due to genomic loss. Restoration of DUSP4 expression in pancreatic cancer cells significantly suppressed invasiveness and anoikis resistance via ERK inactivation. Accordingly, we found that blockade of ERK signaling by MEK inhibition was effective in an orthotopic xenograft model and significantly extended survival. Collectively, our findings demonstrate a genetic mechanism by which pancreatic precursor lesions progress to invasive carcinomas and highlight DUSP4 as a novel invasion suppressor that can be therapeutically exploited through manipulation of ERK signaling. Cancer Res; 76(9); 2612-25. ©2016 AACR.


Asunto(s)
Adenocarcinoma in Situ/patología , Carcinoma Ductal Pancreático/patología , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Neoplasias Pancreáticas/patología , Adenocarcinoma in Situ/genética , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/mortalidad , Adenocarcinoma Mucinoso/patología , Adenocarcinoma Papilar/genética , Adenocarcinoma Papilar/mortalidad , Adenocarcinoma Papilar/patología , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/mortalidad , Hibridación Genómica Comparativa , Progresión de la Enfermedad , Fosfatasas de Especificidad Dual/genética , Xenoinjertos , Humanos , Estimación de Kaplan-Meier , Ratones , Microscopía Confocal , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidad , Transcriptoma
9.
J Pathol ; 239(1): 97-108, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26913567

RESUMEN

We have previously reported that Salvador homologue 1 (SAV1), a component of the Hippo pathway, is significantly down-regulated in high-grade clear cell renal cell carcinoma (ccRCC) due to 14q copy number loss, and that this down-regulation contributes to the proliferation and survival of renal tubular epithelial cells through activation of Yes-associated protein 1 (YAP1), a downstream target of the Hippo pathway. However, the impact of SAV1 loss on the proliferation and survival of kidney cells in vivo remained to be determined. To address this issue, we generated kidney-specific Sav1-knockout (Cdh16-Cre;Sav1(fl/fl) ) mice. Sav1 deficiency enhanced the proliferation of renal tubular epithelial cells in Cdh16-Cre;Sav1(fl/fl) mice, accompanied by nuclear localization of Yap1, suggesting suppression of the Hippo pathway. Sav1 deficiency in renal tubules also caused structural and cellular abnormalities of the epithelial cells, including significant enlargement of their nuclei. Furthermore, Cdh16-Cre;Sav1(fl/fl) mice developed both glomerular and tubular cysts. Although lining cells of the glomerular cysts showed no atypia, those of the tubular cysts showed variations in cell size and nuclear shape, which became more severe as the mice aged. In aged Cdh16-Cre;Sav1(fl/fl) mice, we observed focal disruption of proximal tubules and perivascular lymphocytic infiltration. In conclusion, Sav1 is required for the maintenance of growth, nuclear size and structure of renal tubules under physiological conditions, and its deficiency leads to the acquisition of enhanced proliferation of renal epithelial cells through suppression of Hippo signalling.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Proliferación Celular/fisiología , Túbulos Renales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma de Células Claras/etiología , Animales , Cadherinas/metabolismo , Células Madre Embrionarias/metabolismo , Células Epiteliales/metabolismo , Vía de Señalización Hippo , Neoplasias Renales/etiología , Ratones Transgénicos , Nefritis/etiología , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Proteínas Señalizadoras YAP
10.
Cancer Sci ; 107(4): 417-23, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26790128

RESUMEN

Previously, we reported that genomic loss of 14q occurs more frequently in high-grade than in low-grade clear cell renal cell carcinomas (ccRCCs), and has a significant impact on the levels of expression of genes located in this region, suggesting that such genes may be involved in the malignant transformation of ccRCCs. Here, we found that six of the genes located in the minimal common region of 14q loss were significantly downregulated in high-grade ccRCCs due to copy number loss. Using a dataset from The Cancer Genome Atlas Research Network, we found that downregulation of one of these six genes, WDR20, was significantly associated with poorer outcome in patients with ccRCC, suggesting that WDR20 downregulation may be involved in the malignant transformation of ccRCCs. In functional assays, exogenous WDR20 significantly inhibited the growth of RCC cell lines and induced apoptosis. Interestingly, the phosphorylation levels of ERK and protein kinase B/AKT, which reportedly contribute to the malignant phenotype of RCC cells, were clearly reduced by exogenous expression of WDR20. Thus, our data suggest that downregulation of WDR20 due to 14q loss may be involved in the malignant transformation of ccRCCs, in part through activation of the ERK and protein kinase B/AKT pathways.


Asunto(s)
Carcinoma de Células Renales/genética , Proteínas Portadoras/biosíntesis , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Apoptosis/genética , Carcinoma de Células Renales/patología , Proteínas Portadoras/genética , Cromosomas Humanos Par 14 , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/genética , Proteína Oncogénica v-akt/genética
11.
Cell Mol Life Sci ; 73(3): 523-33, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26507245

RESUMEN

Targeted genome modifications using techniques that alter the genomic information of interest have contributed to multiple studies in both basic and applied biology. Traditionally, in gene targeting, the target-site integration of a targeting vector by homologous recombination is used. However, this strategy has several technical problems. The first problem is the extremely low frequency of gene targeting, which makes obtaining recombinant clones an extremely labor intensive task. The second issue is the limited number of biomaterials to which gene targeting can be applied. Traditional gene targeting hardly occurs in most of the human adherent cell lines. However, a new approach using designer nucleases that can introduce site-specific double-strand breaks in genomic DNAs has increased the efficiency of gene targeting. This new method has also expanded the number of biomaterials to which gene targeting could be applied. Here, we summarize various strategies for target gene modification, including a comparison of traditional gene targeting with designer nucleases.


Asunto(s)
Marcación de Gen/métodos , Roturas del ADN de Doble Cadena , Desoxirribonucleasas/química , Desoxirribonucleasas/fisiología , Eucariontes/genética , Marcación de Gen/tendencias , Recombinación Homóloga , Modelos Genéticos , Ingeniería de Proteínas
12.
Mech Dev ; 135: 31-42, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25463925

RESUMEN

Methylation and de-methylation of histone lysine residues play pivotal roles in mammalian early development; these modifications influence chromatin architecture and regulate gene transcription. Fbxl11 (F-box and leucine-rich repeat 11)/Kdm2a is a histone demethylase that selectively removes mono- and di-methylation from histone H3K36. Previously, two other histone H3K36 demethylases (Jmjd5 or Fbxl10) were analyzed based on the phenotypes of the corresponding knockout (KO) mice; the results of those studies implicated H3K36 demethylases in cell proliferation, apoptosis, and senescence (Fukuda et al., 2011; Ishimura et al., 2012). To elucidate the physiological role of Fbxl11, we generated and examined Fbxl11 KO mice. Fbxl11 was expressed throughout the body during embryogenesis, and the Fbxl11 KO mice exhibited embryonic lethality at E10.5-12.5, accompanied with severe growth defects leading to reduced body size. Furthermore, knockout of Fbxl11 decreased cell proliferation and increased apoptosis. The lack of Fbxl11 resulted in downregulation of the Polycomb group protein (PcG) Ezh2, PcG mediated H2A ubiquitination and upregulation of the cyclin-dependent kinase inhibitor p21Cip1. Taken together, our findings suggest that Fbxl11 plays an essential role in embryonic development and homeostasis by regulating cell proliferation and survival.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Supervivencia Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/enzimología , Desarrollo Embrionario , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Histonas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Grupo Polycomb/metabolismo , Procesamiento Proteico-Postraduccional
13.
Cereb Cortex ; 23(8): 1824-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22705452

RESUMEN

Polypyrimidine tract-binding protein (PTB) is a well-characterized RNA-binding protein and known to be preferentially expressed in neural stem cells (NSCs) in the central nervous system; however, its role in NSCs in the developing brain remains unclear. To explore the role of PTB in embryonic NSCs in vivo, Nestin-Cre-mediated conditional Ptb knockout mice were generated for this study. In the mutant forebrain, despite the depletion of PTB protein, neither abnormal neurogenesis nor flagrant morphological abnormalities were observed at embryonic day 14.5 (E14.5). Nevertheless, by 10 weeks, nearly all mutant mice succumbed to hydrocephalus (HC), which was caused by a lack of the ependymal cell layer in the dorsal cortex. Upon further analysis, a gradual loss of adherens junctions (AJs) was observed in the ventricular zone (VZ) of the dorsal telencephalon in the mutant brains, beginning at E14.5. In the AJs-deficient VZ, impaired interkinetic nuclear migration and precocious differentiation of NSCs were observed after E14.5. These findings demonstrated that PTB depletion in the dorsal telencephalon is causally involved in the development of HC and that PTB is important for the maintenance of AJs in the NSCs of the dorsal telencephalon.


Asunto(s)
Uniones Adherentes/ultraestructura , Hidrocefalia/etiología , Proteína de Unión al Tracto de Polipirimidina/fisiología , Telencéfalo/embriología , Animales , Hidrocefalia/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/ultraestructura , Proteína de Unión al Tracto de Polipirimidina/genética , Telencéfalo/anomalías
14.
Dev Cell ; 22(4): 707-20, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22445366

RESUMEN

Regulation of self-renewal and differentiation of neural stem cells is still poorly understood. Here we investigate the role of a developmentally expressed protein, Botch, which blocks Notch, in neocortical development. Downregulation of Botch in vivo leads to cellular retention in the ventricular and subventricular zones, whereas overexpression of Botch drives neural stem cells into the intermediate zone and cortical plate. In vitro neurosphere and differentiation assays indicate that Botch regulates neurogenesis by promoting neuronal differentiation. Botch prevents cell surface presentation of Notch by inhibiting the S1 furin-like cleavage of Notch, maintaining Notch in the immature full-length form. Understanding the function of Botch expands our knowledge regarding both the regulation of Notch signaling and the complex signaling mediating neuronal development.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular , Embrión de Mamíferos/metabolismo , Células-Madre Neurales/citología , Neurogénesis/fisiología , Fármacos Neuroprotectores/metabolismo , Receptores Notch/antagonistas & inhibidores , Receptores Notch/metabolismo , Animales , Western Blotting , Proteínas Portadoras/genética , Membrana Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos/citología , Aparato de Golgi/metabolismo , Humanos , Inmunoprecipitación , Ratones , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/genética , Transducción de Señal , gamma-Glutamilciclotransferasa
15.
Cell Cycle ; 10(21): 3706-13, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22037210

RESUMEN

Polypyrimidine tract-binding protein (PTB/PTBP1/hnRNP I) is a member of the heterogeneous nuclear ribonucleoprotein family that binds specifically to pyrimidine-rich sequences of RNAs. Although PTB is a multifunctional protein involved in RNA processing and internal ribosome entry site (IRES)-dependent translation, the role of PTB in early mouse development is unclear. Ptb knockout mice exhibit embryonic lethality shortly after implantation and Ptb-/- embryonic stem (ES) cells have a severe proliferation defect that includes a prolonged G2/M phase. The present study shows that PTB promotes M phase progression by the direct repression of CDK11(p58) IRES activity in ES cells. The protein expression and IRES activity of CDK11(p58) in Ptb-/- ES cells is higher than that of wild-type ES cells, indicating that PTB is involved in the repression of CDK11(p58) expression through IRES-dependent translation in ES cells. Interestingly, CDK11(p58) IRES activity is activated by upstream of N-Ras (UNR) in 293T and NIH3T3 cells, whereas UNR is not present in the Cdk11 mRNA-protein complex in ES cells. In addition, PTB interacts directly with the IRES region of CDK11(p58) in ES cells. These results suggest that PTB regulates the precise expression of CDK11(p58) through direct interaction with CDK11(p58) IRES and promotes M phase progression in ES cells.


Asunto(s)
Ciclo Celular/fisiología , Ciclina D3/genética , Proteína de Unión al Tracto de Polipirimidina/fisiología , Animales , Ciclina D3/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Ratones , Biosíntesis de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , ARN Mensajero/metabolismo , ARN Mensajero/fisiología
16.
Mol Cell Neurosci ; 46(3): 614-24, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21220025

RESUMEN

Histone methylation is the important transcription regulatory system that affects mammalian development and cell differentiation. Alterations in epigenetic gene regulation are associated with disease. Fbxl10 (F-box and leucine-rich repeat protein 10) is a JmjC domain-containing histone demethylase. Although Fbxl10 has been implicated in cell cycle regulation, cell death, senescence, and tumorigenesis, these functions are controversial and its physiological function is unclear. To determine the in vivo function of Fbxl10, in this study, we generated a homozygous mutation in the mouse Fbxl10 gene. About half of Fbxl10-deficient mice exhibit failure of neural tube closure, resulting in exencephaly and die shortly after birth. Fbxl10 deficiency also causes retinal coloboma and a curled tail with low penetrances. Fbxl10 mRNA is specifically expressed in the cranial neural folds at E8.5 embryos, and apoptosis increased in the neuroepithelium and mesenchyme of Fbxl10-deficient E9.5 embryos, consistent with neural tube defects found in Fbxl10-deficient mice. Depletion of Fbxl10 induced the increased expression of p19ARF, an inducer of apoptosis, in E8.5 embryos and mouse embryonic fibroblast cells. In addition, the number of mitotic neural progenitor cells is significantly increased in the mutant E14.5 brain. Our findings suggest that the Fbxl10 gene makes important contributions to embryonic neural development by regulating cell proliferation and cell death in mice.


Asunto(s)
Muerte Celular/fisiología , Embrión de Mamíferos/anomalías , Proteínas F-Box/genética , Histona Demetilasas con Dominio de Jumonji/genética , Células-Madre Neurales/fisiología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/fisiopatología , Animales , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Histona Demetilasas con Dominio de Jumonji/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural , Células-Madre Neurales/citología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología
17.
J Cell Biol ; 189(1): 159-70, 2010 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-20351065

RESUMEN

Once they have differentiated, cells retain their individual character and repress genes that are specifically expressed in other cell lineages, but how alternative fate choice is restricted during and/or after differentiation remains unclear. In the mammalian central nervous system, neurons, astrocytes, and oligodendrocytes are generated throughout life from common tripotent neural progenitor cells (NPCs). Bone morphogenetic proteins (BMPs) are well-known astrocyte-inducing cytokines. We show here that the expression of a transcriptional repressor, RE1 silencer of transcription (REST)/neuron-restrictive silencer factor (NRSF), is up-regulated and sustained by BMP signal activation in the course of astrocytic differentiation of NPCs, and restricts neuronal differentiation. We further show that, in differentiated astrocytes, endogenous REST/NRSF associates with various neuronal genes and that disruption of its function resulted in their derepression, thereby explaining how ectopic neuronal gene expression is prevented in cells with astrocytic traits. Collectively, our results suggest that REST/NRSF functions as a molecular regulator of the nonneuronal phenotype in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Embrión de Mamíferos/metabolismo , Ratones , Neuronas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Regulación hacia Arriba
18.
FEBS J ; 276(22): 6658-68, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19843185

RESUMEN

Polypyrimidine tract-binding protein (PTB) is a widely expressed RNA-binding protein with multiple roles in RNA processing, including the splicing of alternative exons, mRNA stability, mRNA localization, and internal ribosome entry site-dependent translation. Although it has been reported that increased expression of PTB is correlated with cancer cell growth, the role of PTB in mammalian development is still unclear. Here, we report that a homozygous mutation in the mouse Ptb gene causes embryonic lethality shortly after implantation. We also established Ptb(-/-) embryonic stem (ES) cell lines and found that these mutant cells exhibited severe defects in cell proliferation without aberrant differentiation in vitro or in vivo. Furthermore, cell cycle analysis and a cell synchronization assay revealed that Ptb(-/-) ES cells have a prolonged G(2)/M phase. Thus, our data indicate that PTB is essential for early mouse development and ES cell proliferation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Blastocisto/citología , Blastocisto/metabolismo , Northern Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Reacción en Cadena de la Polimerasa
19.
J Neurosci Res ; 87(16): 3521-34, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19598246

RESUMEN

The Notch signaling pathway has a crucial role in the differentiation of hair cells and supporting cells by mediating "lateral inhibition" via the ligands Delta-like1 (Dll1) and Jagged2 (Jag2) and the effectors Hes1 and Hes5 during mammalian inner ear development. Recently, another Notch ligand, Jagged1 (Jag1)-dependent Notch activation, has been revealed to be important for the determination of the prosensory region in the earlier stage before cell differentiation. However, little is known about the effectors of the Notch pathway in this context. P27(Kip1), a cyclin-dependent kinase inhibitor, is also known to demarcate the prosensory region in the cochlear primordium, which consists of the sensory progenitors that have completed their terminal mitoses. Hes1 reportedly promotes precursor cell proliferation through the transcriptional down-regulation of p27(Kip1) in the thymus, liver, and brain. In this study, we observed Hes1 as a mediator between the Notch signaling pathway and the regulation of proliferation of sensory precursor cells by p27(Kip1) in the developing cochlea. We showed that Hes1, but not Hes5, was weakly expressed at the time of onset of p27(Kip1). The expression pattern of Hes1 prior to cell differentiation was similar to that of activated Notch1. P27(Kip1) was up-regulated and BrdU-positive S-phase cells were reduced in the developing cochlear epithelium of Hes1 null mice. These results suggest that the Notch-Hes1 pathway may contribute to the adequate proliferation of sensory precursor cells via the potential transcriptional down-regulation of p27(Kip1) expression and play a pivotal role in the correct prosensory determination.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cóclea/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Homeodominio/metabolismo , Neurogénesis/fisiología , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Ciclo Celular/genética , Ciclo Celular/fisiología , Cóclea/citología , Cóclea/crecimiento & desarrollo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/genética , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica/genética , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Proteínas de Homeodominio/genética , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Notch1/genética , Proteínas Serrate-Jagged , Transducción de Señal/genética , Factor de Transcripción HES-1
20.
Cell Stem Cell ; 2(5): 461-71, 2008 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-18462696

RESUMEN

The Notch signaling pathway plays important roles in cell-fate determination during embryonic development and adult life. In this study, we focus on the role of Notch signaling in governing cell-fate choices in human embryonic stem cells (hESCs). Using genetic and pharmacological approaches, we achieved both blockade and conditional activation of Notch signaling in several hESC lines. We report here that activation of Notch signaling is required for undifferentiated hESCs to form the progeny of all three embryonic germ layers, but not trophoblast cells. In addition, transient Notch signaling pathway activation enhanced generation of hematopoietic cells from committed hESCs. These new insights into the roles of Notch in hESC-fate determination may help to efficiently direct hESC differentiation into therapeutically relevant cell types.


Asunto(s)
Diferenciación Celular , Linaje de la Célula/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Estratos Germinativos/citología , Estratos Germinativos/fisiología , Receptores Notch/fisiología , Trofoblastos/citología , Trofoblastos/fisiología , Adulto , Células Cultivadas , Dipéptidos/administración & dosificación , Femenino , Vectores Genéticos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Humanos , Lentivirus , Embarazo , Receptores Notch/antagonistas & inhibidores , Transducción de Señal , Transformación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA