Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 79(1): 84-98.e9, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32526163

RESUMEN

Rett syndrome (RTT), mainly caused by mutations in methyl-CpG binding protein 2 (MeCP2), is one of the most prevalent intellectual disorders without effective therapies. Here, we used 2D and 3D human brain cultures to investigate MeCP2 function. We found that MeCP2 mutations cause severe abnormalities in human interneurons (INs). Surprisingly, treatment with a BET inhibitor, JQ1, rescued the molecular and functional phenotypes of MeCP2 mutant INs. We uncovered that abnormal increases in chromatin binding of BRD4 and enhancer-promoter interactions underlie the abnormal transcription in MeCP2 mutant INs, which were recovered to normal levels by JQ1. We revealed cell-type-specific transcriptome impairment in MeCP2 mutant region-specific human brain organoids that were rescued by JQ1. Finally, JQ1 ameliorated RTT-like phenotypes in mice. These data demonstrate that BRD4 dysregulation is a critical driver for RTT etiology and suggest that targeting BRD4 could be a potential therapeutic opportunity for RTT.


Asunto(s)
Azepinas/farmacología , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Interneuronas/patología , Proteína 2 de Unión a Metil-CpG/fisiología , Síndrome de Rett/patología , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos , Triazoles/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Femenino , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Factores de Transcripción/genética
2.
Cell Mol Life Sci ; 81(1): 142, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38485770

RESUMEN

Thioredoxin interacting protein (Txnip) is a stress-responsive factor regulating Trx1 for redox balance and involved in diverse cellular processes including proliferation, differentiation, apoptosis, inflammation, and metabolism. However, the biological role of Txnip function in stem cell pluripotency has yet to be investigated. Here, we reveal the novel functions of mouse Txnip in cellular reprogramming and differentiation onset by involving in glucose-mediated histone acetylation and the regulation of Oct4, which is a fundamental component of the molecular circuitry underlying pluripotency. During reprogramming or PSC differentiation process, cellular metabolic and chromatin remodeling occur in order to change its cellular fate. Txnip knockout promotes induced pluripotency but hinders initial differentiation by activating pluripotency factors and promoting glycolysis. This alteration affects the intracellular levels of acetyl-coA, a final product of enhanced glycolysis, resulting in sustained histone acetylation on active PSC gene regions. Moreover, Txnip directly interacts with Oct4, thereby repressing its activity and consequently deregulating Oct4 target gene transcriptions. Our work suggests that control of Txnip expression is crucial for cell fate transitions by modulating the entry and exit of pluripotency.


Asunto(s)
Reprogramación Celular , Histonas , Animales , Ratones , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/genética , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
3.
Int J Mol Sci ; 22(9)2021 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922276

RESUMEN

Sialidosis, caused by a genetic deficiency of the lysosomal sialidase gene (NEU1), is a systemic disease involving various tissues and organs, including the nervous system. Understanding the neurological dysfunction and pathology associated with sialidosis remains a challenge, partially due to the lack of a human model system. In this study, we have generated two types of induced pluripotent stem cells (iPSCs) with sialidosis-specific NEU1G227R and NEU1V275A/R347Q mutations (sialidosis-iPSCs), and further differentiated them into neural precursor cells (iNPCs). Characterization of NEU1G227R- and NEU1V275A/R347Q- mutated iNPCs derived from sialidosis-iPSCs (sialidosis-iNPCs) validated that sialidosis-iNPCs faithfully recapitulate key disease-specific phenotypes, including reduced NEU1 activity and impaired lysosomal and autophagic function. In particular, these cells showed defective differentiation into oligodendrocytes and astrocytes, while their neuronal differentiation was not notably affected. Importantly, we found that the phenotypic defects of sialidosis-iNPCs, such as impaired differentiation capacity, could be effectively rescued by the induction of autophagy with rapamycin. Our results demonstrate the first use of a sialidosis-iNPC model with NEU1G227R- and NEU1V275A/R347Q- mutation(s) to study the neurological defects of sialidosis, particularly those related to a defective autophagy-lysosome pathway, and may help accelerate the development of new drugs and therapeutics to combat sialidosis and other LSDs.


Asunto(s)
Astrocitos/patología , Células Madre Pluripotentes Inducidas/patología , Mucolipidosis/patología , Células-Madre Neurales/patología , Neuraminidasa/metabolismo , Oligodendroglía/patología , Teratoma/patología , Astrocitos/metabolismo , Autofagia , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas , Mucolipidosis/genética , Mucolipidosis/metabolismo , Mutación , Células-Madre Neurales/metabolismo , Neuraminidasa/genética , Oligodendroglía/metabolismo , Fenotipo , Teratoma/genética , Teratoma/metabolismo
4.
Angiogenesis ; 22(2): 281-293, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30471052

RESUMEN

Testis-specific protein, Y-encoded like (TSPYL) family proteins (TSPYL1-6), which are members of the nucleosome assembly protein superfamily, have been determined to be involved in the regulation of various cellular functions. However, the potential role of TSPYL family proteins in endothelial cells (ECs) has not been determined. Here, we demonstrated that the expression of TSPYL5 is highly enriched in human ECs such as human umbilical vein endothelial cells (HUVECs) and human pluripotent stem cell-differentiated ECs (hPSC-ECs). Importantly, TSPYL5 overexpression was shown to promote EC proliferation and functions, such as migration and tube formation, by downregulating p53 expression. Adriamycin-induced senescence was markedly blocked by TSPYL5 overexpression. In addition, the TSPYL5 depletion-mediated loss of EC functions was blocked by p53 inhibition. Significantly, TSPYL5 overexpression promoted angiogenesis in Matrigel plug and wound repair in a mouse skin wound healing model in vivo. Our results suggest that TSPYL5, a novel angiogenic regulator, plays a key role in maintaining endothelial integrity and function. These findings extend the understanding of TSPYL5-dependent mechanisms underlying the regulation of p53-related functions in ECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/fisiología , Neovascularización Fisiológica/genética , Proteínas Nucleares/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Proteína p53 Supresora de Tumor/metabolismo
5.
Biochem Biophys Res Commun ; 520(2): 406-412, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31607477

RESUMEN

Selenium (Se) plays a vital role in reactive oxygen species (ROS) homeostasis and redox regulation in intracellular signaling via selenocysteine (Sec), known as the 21st proteinogenic amino acid, but its specific biological functions in development and disease remain undiscovered. In this study, we explored the role of selenophosphate synthetase 1 (SEPHS1) in the pluripotency maintenance and reprogramming. We found that high level of SEPHS1 is retained in undifferentiated embryonic stem cells (ESCs), which is decreased during their differentiation. SEPHS1 knockdown significantly reduced reprogramming efficiency, proving that SEPHS1 is required for acquisition of pluripotency. However, SEPHS1 knockdown did not affect the expression of significant pluripotency genes, suggesting that SEPHS1 may be involved in the survival of pluripotent stem cells rather than in the regulation of pluripotency genes. Transcriptome analysis revealed altered expression of the gene set related to the ROS pathway and apoptosis in SEPHS1-knockdown cells. We also demonstrated the role of SEPHS1 in human ESC clonogenicity, and we found improved single-cell survival of hESCs by selenium treatment in a concentration-dependent manner. Our study implies that hSEPHS1 is a regulator of selenium-mediated redox-signaling in human pluripotent stem cells and plays a role in their survival.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Fosfotransferasas/genética , Fosfotransferasas/metabolismo , Selenio/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular , Células Cultivadas , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/enzimología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Selenio/farmacología , Transducción de Señal
6.
Biochem Biophys Res Commun ; 493(1): 723-730, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28859981

RESUMEN

Large-scale production of human pluripotent stem cells (hPSCs) in an efficient and safe manner is crucial to the successful application of hPSCs in biomedical research and regenerative medicine. Three-dimensional culture methods for hPSCs have been extensively studied using single-cell passaging approaches; however, these techniques have been challenged by the induction of massive cell death and accumulation of genomic abnormalities. In this work, we developed and optimized a novel, simple clump-passaging method for in vitro hPSCs 3-dimensional (3D) culture that can be exploited for large-scale production. Fully grown hPSC spheroids were dissociated into smaller-sized spheroid clumps by simple treatment with enzyme-free dissociation buffer, and clumped hPSCs were inoculated and maintained for 3D suspension culture. Our clump-passaging method effectively increased the hPSCs survival rate after subculture and supported scalable hPSCs 3D expansion. We also tested and selected chemically defined media formulations that are suitable for 3D culture and commercially available. Overall, our clump-passaging and expansion method demonstrated high survival and expansion rates for hPSC spheroids compared with conventional methods and may also have the advantage of maintaining genomic stability.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Ingeniería de Tejidos/métodos , Diferenciación Celular/fisiología , Medios de Cultivo/metabolismo , Humanos
7.
Stem Cells ; 34(12): 2840-2851, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27428041

RESUMEN

The fundamental tenet that aging is irreversible has been challenged by the development of reprogramming technology that can restore molecular and cellular age by reversing the progression of aging. The use of cells from aged individuals as sources for reprogramming or transplantation creates a major barrier in stem cell therapy with respect to cell quality and quantity. Here, we investigated the molecular features underlying senescence and rejuvenation during aged cell reprogramming and identified novel factors that can overcome age-associated barriers. Enzymes, such as nicotinamide nucleotide transhydrogenase (NNT) and nicotinamide mononucleotide adenylyltransferase 3 (NMNAT3), that control mitochondrial NAD+ levels appear to be susceptible to aging. In aged cells, mitochondrial NAD+ levels decrease, accompanied by reduced SIRT3 activity; these changes severely impede cell fate transition. However, in cells collected from aged p16 knockout mice, which exhibit delayed cellular senescence, no changes in NNT or NMNAT3 expression were found. Importantly, restoring mitochondrial NAD+ levels by overexpressing NNT and NMNAT3 enhanced reprogramming efficiency of aged somatic cells and extended the lifespan of human mesenchymal stem cells by delaying replicative senescence. These results demonstrate that maintenance of mitochondrial NAD+ levels is critical for reversing the mechanisms of aging and ensuring that cells collected from aged individuals are of high quality. Stem Cells 2016;34:2840-2851.


Asunto(s)
Reprogramación Celular , Senescencia Celular , Mitocondrias/metabolismo , NAD/metabolismo , Células Madre/citología , Células Madre/metabolismo , Envejecimiento , Animales , Linaje de la Célula , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones Noqueados , NADP Transhidrogenasas , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Sirtuina 3/metabolismo
8.
Hum Mol Genet ; 23(7): 1802-16, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24218367

RESUMEN

The extensive molecular characterization of human pluripotent stem cells (hPSCs), human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) is required before they can be applied in the future for personalized medicine and drug discovery. Despite the efforts that have been made with kinome analyses, we still lack in-depth insights into the molecular signatures of receptor tyrosine kinases (RTKs) that are related to pluripotency. Here, we present the first detailed and distinct repertoire of RTK characteristic for hPSC pluripotency by determining both the expression and phosphorylation profiles of RTKs in hESCs and hiPSCs using reverse transcriptase-polymerase chain reaction with degenerate primers that target conserved tyrosine kinase domains and phospho-RTK array, respectively. Among the RTKs tested, the up-regulation of EPHA1, ERBB2, FGFR4 and VEGFR2 and the down-regulation of AXL, EPHA4, PDGFRB and TYRO3 in terms of both their expression and phosphorylation levels were predominantly related to the maintenance of hPSC pluripotency. Notably, the specific inhibition of AXL was significantly advantageous in maintaining undifferentiated hESCs and hiPSCs and for the overall efficiency and kinetics of hiPSC generation. Additionally, a global phosphoproteomic analysis showed that ∼30% of the proteins (293 of 970 phosphoproteins) showed differential phosphorylation upon AXL inhibition in undifferentiated hPSCs, revealing the potential contribution of AXL-mediated phosphorylation dynamics to pluripotency-related signaling networks. Our findings provide a novel molecular signature of AXL in pluripotency control that will complement existing pluripotency-kinome networks.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Secuencia de Aminoácidos , Ciclo Celular/genética , Células Cultivadas , Cromatografía Liquida , Activación Enzimática , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Datos de Secuencia Molecular , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Fosforilación , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética , Transducción de Señal/genética , Espectrometría de Masas en Tándem , Tirosina Quinasa del Receptor Axl
9.
J Pathol ; 237(1): 98-110, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25925601

RESUMEN

GM1 gangliosidosis (GM1) is an inherited neurodegenerative disorder caused by mutations in the lysosomal ß-galactosidase (ß-gal) gene. Insufficient ß-gal activity leads to abnormal accumulation of GM1 gangliosides in tissues, particularly in the central nervous system, resulting in progressive neurodegeneration. Here, we report an in vitro human GM1 model, based on induced pluripotent stem cell (iPSC) technology. Neural progenitor cells differentiated from GM1 patient-derived iPSCs (GM1-NPCs) recapitulated the biochemical and molecular phenotypes of GM1, including defective ß-gal activity and increased lysosomes. Importantly, the characterization of GM1-NPCs established that GM1 is significantly associated with the activation of inflammasomes, which play a critical role in the pathogenesis of various neurodegenerative diseases. Specific inflammasome inhibitors potently alleviated the disease-related phenotypes of GM1-NPCs in vitro and in vivo. Our data demonstrate that GM1-NPCs are a valuable in vitro human GM1 model and suggest that inflammasome activation is a novel target pathway for GM1 drug development.


Asunto(s)
Gangliosidosis GM1/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Inflamasomas/metabolismo , Células-Madre Neurales/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Forma de la Célula , Reprogramación Celular , Gangliosidosis GM1/inmunología , Gangliosidosis GM1/patología , Genotipo , Humanos , Factores Inmunológicos/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Inflamasomas/antagonistas & inhibidores , Inflamasomas/inmunología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/inmunología , Células-Madre Neurales/patología , Células-Madre Neurales/trasplante , Fenotipo , Transducción de Señal , Factores de Tiempo , beta-Galactosidasa/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(35): E3281-90, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23918355

RESUMEN

The future of safe cell-based therapy rests on overcoming teratoma/tumor formation, in particular when using human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Because the presence of a few remaining undifferentiated hPSCs can cause undesirable teratomas after transplantation, complete removal of these cells with no/minimal damage to differentiated cells is a prerequisite for clinical application of hPSC-based therapy. Having identified a unique hESC signature of pro- and antiapoptotic gene expression profile, we hypothesized that targeting hPSC-specific antiapoptotic factor(s) (i.e., survivin or Bcl10) represents an efficient strategy to selectively eliminate pluripotent cells with teratoma potential. Here we report the successful identification of small molecules that can effectively inhibit these antiapoptotic factors, leading to selective and efficient removal of pluripotent stem cells through apoptotic cell death. In particular, a single treatment of hESC-derived mixed population with chemical inhibitors of survivin (e.g., quercetin or YM155) induced selective and complete cell death of undifferentiated hPSCs. In contrast, differentiated cell types (e.g., dopamine neurons and smooth-muscle cells) derived from hPSCs survived well and maintained their functionality. We found that quercetin-induced selective cell death is caused by mitochondrial accumulation of p53 and is sufficient to prevent teratoma formation after transplantation of hESC- or hiPSC-derived cells. Taken together, these results provide the "proof of concept" that small-molecule targeting of hPSC-specific antiapoptotic pathway(s) is a viable strategy to prevent tumor formation by selectively eliminating remaining undifferentiated pluripotent cells for safe hPSC-based therapy.


Asunto(s)
Células Madre Pluripotentes/citología , Bibliotecas de Moléculas Pequeñas , Teratoma/patología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Apoptosis , Proteína 10 de la LLC-Linfoma de Células B , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Mitocondrias/metabolismo , Naftoquinonas/farmacología , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Survivin , Teratoma/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Proteomics ; 15(13): 2220-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25736782

RESUMEN

Recent studies have suggested that REX1 (reduced expression 1) plays an important role in pluripotency, proliferation, and differentiation. However, the molecular mechanisms involved in REX1-dependent regulation of diverse cellular processes remain unclear. To elucidate the regulatory functions of REX1 in human embryonic stem cells (hESCs), comparative proteomic analysis was performed on REX1 RNAi specifically silenced hESCs. Analysis of the proteome via nano-LC-MS/MS identified 140 differentially expressed proteins (DEPs) displaying a >2-fold difference in expression level between control and REX1 knockdown (KD) hESCs, which were then compared with transcriptome data and validated by quantitative real-time RT-PCR and Western blotting. These DEPs were analyzed by GO, pathway, and functional clustering analyses to determine the molecular functions of the proteins and pathways regulated by REX1. The REX1 KD-mediated DEPs mapped to major biological processes involved in the regulation of ribosome-mediated translation and mitochondrial function. Functional network analysis revealed a highly interconnected network among these DEPs and indicated that these interconnected proteins are predominantly involved in translation and the regulation of mitochondrial organization. These findings regarding REX1-mediated regulatory network have revealed the contributions of REX1 to maintaining the status of hESCs and have improved our understanding of the molecular events that underlie the fundamental properties of hESCs.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteómica , Western Blotting , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Interferencia de ARN
12.
Biochem Biophys Res Commun ; 457(4): 554-60, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600812

RESUMEN

Lysosomes are cytoplasmic compartments that contain many acid hydrolases and play critical roles in the metabolism of a wide range of macromolecules. Deficiencies in lysosomal enzyme activities cause genetic diseases, called lysosomal storage disorders (LSDs). Many mutations have been identified in the genes responsible for LSDs, and the identification of mutations is required for the accurate molecular diagnoses. Here, we analyzed cell lines that were derived from two different LSDs, GM1 gangliosidosis and sialidosis. GM1 gangliosidosis is caused by mutations in the GLB1 gene that encodes ß-galactosidase. A lack of ß-galactosidase activity leads to the massive accumulation of GM1 ganglioside, which results in neurodegenerative pathology. Mutations in the NEU1 gene that encodes lysosomal sialidase cause sialidosis. Insufficient activity of lysosomal sialidase progressively increases the accumulation of sialylated molecules, and various clinical symptoms, including mental retardation, appear. We sequenced the entire coding regions of GLB1 and NEU1 in GM1 gangliosidosis and sialidosis patient cells, respectively. We found the novel mutations p.E186A in GLB1 and p.R347Q in NEU1, as well as many other mutations that have been previously reported. We also demonstrated that patient cells containing the novel mutations showed the molecular phenotypes of the corresponding disease. Further structural analysis suggested that these novel mutation sites are highly conserved and important for enzyme activity.


Asunto(s)
Gangliosidosis GM1/enzimología , Gangliosidosis GM1/genética , Mucolipidosis/enzimología , Mucolipidosis/genética , Neuraminidasa/genética , beta-Galactosidasa/genética , Secuencia de Aminoácidos , Animales , Fibroblastos/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Neuraminidasa/química , Neuraminidasa/metabolismo , Alineación de Secuencia , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo
13.
Stem Cells ; 32(12): 3126-36, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25185564

RESUMEN

Oct4 has been implicated in regulation of pluripotency in embryonic stem cells (ESCs) and reprogramming of somatic cells into induced pluripotent stem cells. However, the molecular mechanisms involved in Oct4-dependent regulation of pluripotency and reprogramming have not been clear. To gain insight into the mechanism of regulation of Oct4-mediated self-renewal of ESCs and reprogramming of somatic cells, we attempted to identify Oct4-binding proteins using affinity purification and mass spectrometry. We identified Reptin, a key component of ATP-dependent chromatin remodeling complexes, as an Oct4-binding protein. Depletion of endogenous Reptin using lentiviral short hairpin RNA (shRNA) led to a decrease in the number and size of alkaline phosphatase-positive colonies of mouse ESCs. In addition, shRNA-mediated silencing of Reptin resulted in decreased expression of pluripotency-specific marker genes, including Oct4, Sox2, Nanog, and SSEA-1. Results of the Oct4 reporter assay showed synergism between Oct4 and Reptin, and depletion of endogenous Reptin abolished Oct4 transcriptional activity. Results of a chromatin immunoprecipitation assay showed the overlapping interaction of Reptin and Oct4 to CR4 in the Oct4 enhancer in ESCs. Knockdown of Reptin using shRNA suppressed the reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells, whereas overexpression of Reptin resulted in enhanced efficiency of induced pluripotent stem cell generation. These results strongly suggest that Reptin plays a key role in maintaining the pluripotency of ESCs and in establishing the pluripotency during reprogramming of somatic cells by regulation of Oct4-mediated gene regulation.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular , ADN Helicasas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Células Cultivadas , Reprogramación Celular/fisiología , Ratones
14.
Biochem Biophys Res Commun ; 446(2): 493-8, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24613840

RESUMEN

Genomic imprinting is an epigenetic phenomenon by which a subset of genes is asymmetrically expressed in a parent-of-origin manner. However, little is known regarding the epigenetic behaviors of imprinted genes during human development. Here, we show dynamic epigenetic changes in imprinted genes in hESCs during in vitro differentiation into specialized cell types. Out of 9 imprinted genes with single nucleotide polymorphisms, mono-allelic expression for three imprinted genes (H19, KCNQ1OT1, and IPW), and bi- or partial-allelic expression for three imprinted genes (OSBPL5, PPP1R9A, and RTL1) were stably retained in H9-hESCs throughout differentiation, representing imprinting stability. Three imprinted genes (KCNK9, ATP10A, and SLC22A3) showed a loss and a gain of imprinting in a lineage-specific manner during differentiation. Changes in allelic expression of imprinted genes were observed in another hESC line during in vitro differentiation. These findings indicate that the allelic expression of imprinted genes may be vulnerable in a lineage-specific manner in human pluripotent stem cells during differentiation.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Frecuencia de los Genes/genética , Variación Genética/genética , Impresión Genómica/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Línea Celular , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética
15.
Stem Cells ; 31(11): 2374-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23939908

RESUMEN

Reduced expression 1 (REX1) is a widely used pluripotency marker, but little is known about its roles in pluripotency. Here, we show that REX1 is functionally important in the reacquisition and maintenance of pluripotency. REX1-depleted human pluripotent stem cells (hPSCs) lose their self-renewal capacity and full differentiation potential, especially their mesoderm lineage potential. Cyclin B1/B2 expression was found to parallel that of REX1. REX1 positively regulates the transcriptional activity of cyclin B1/B2 through binding to their promoters. REX1 induces the phosphorylation of DRP1 at Ser616 by cyclin B/CDK1, which leads to mitochondrial fission and appears to be important for meeting the high-energy demands of highly glycolytic hPSCs. During reprogramming to pluripotency by defined factors (OCT4, SOX2, KLF4, and c-MYC), the reprogramming kinetics and efficiency are markedly improved by adding REX1 or replacing KLF4 with REX1. These improvements are achieved by lowering reprogramming barriers (growth arrest and apoptosis), by enhancing mitochondrial fission, and by conversion to glycolytic metabolism, dependent on the cyclin B1/B2-DRP1 pathway. Our results show that a novel pluripotency regulator, REX1, is essential for pluripotency and reprogramming.


Asunto(s)
Células Madre Embrionarias/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Células Madre Embrionarias/citología , Técnicas de Silenciamiento del Gen , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Microscopía Electrónica , Células Madre Pluripotentes/citología , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , Transfección
16.
Stem Cells ; 31(6): 1121-35, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526681

RESUMEN

Crosstalk between intracellular signaling pathways has been extensively studied to understand the pluripotency of human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells (hiPSCs); however, the contribution of NAD(+) -dependent pathways remains largely unknown. Here, we show that NAD(+) depletion by FK866 (a potent inhibitor of NAD(+) biosynthesis) was fatal in hPSCs, particularly when deriving pluripotent cells from somatic cells and maintaining pluripotency. NAD and its precursors (nicotinamide [NAM] and nicotinic acid) fully replenished the NAD(+) depletion by FK866 in hPSCs. However, only NAM effectively enhanced the reprogramming efficiency and kinetics of hiPSC generation and was also significantly advantageous for the maintenance of undifferentiated hPSCs. Our molecular and functional studies reveal that NAM lowers the barriers to reprogramming by accelerating cell proliferation and protecting cells from apoptosis and senescence by alleviating oxidative stress, reactive oxygen species accumulation, and subsequent mitochondrial membrane potential collapse. We provide evidence that the positive effects of NAM (occurring at concentrations well above the physiological range) on pluripotency control are molecularly associated with the repression of p53, p21, and p16. Our findings establish that adequate intracellular NAD(+) content is crucial for pluripotency; the distinct effects of NAM on pluripotency may be dependent not only on its metabolic advantage as a NAD(+) precursor but also on the ability of NAM to enhance resistance to cellular stress.


Asunto(s)
Reprogramación Celular/genética , Niacinamida/genética , Niacinamida/metabolismo , Células Madre Pluripotentes/metabolismo , Apoptosis/genética , Línea Celular , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Potencial de la Membrana Mitocondrial/genética , NAD/genética , NAD/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Niacina/genética , Niacina/metabolismo , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Anal Chem ; 85(15): 7462-70, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23834277

RESUMEN

Glycans, which decorate cell surfaces, play crucial roles in various physiological events involving cell surface recognition. Despite the importance of surface glycans, most analyses have been performed using total cells or whole membranes rather than plasma membranes due to difficulties related to isolation. In the present study, we employed an adhesion-based method for plasma membrane isolation to analyze N-glycans on cell surfaces. Cells were attached to polylysine-coated glass plates and then ruptured by hypotonic pressure. After washing to remove intracellular organelles, only a plasma membrane fraction remained attached to the plates, as confirmed by fluorescence imaging using organelle-specific probes. The plate was directly treated with trypsin to digest and detach the glycoproteins from the plasma membrane. From the resulting glycopeptides, N-glycans were released and analyzed using MALDI-TOF mass spectrometry and HPLC. When N-glycan profiles obtained by this method were compared to those by other methods, the amount of high-mannose type glycans mainly contaminated from the endoplasmic reticulum was dramatically reduced, which enabled the efficient detection of complex type glycans present on the cell surface. Moreover, this method was successfully used to analyze the increase of high-mannose glycans on the surface as induced by a mannosidase inhibitor treatment.


Asunto(s)
Membrana Celular/metabolismo , Polisacáridos/metabolismo , Animales , Células CHO , Adhesión Celular , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Manosa/química , Polisacáridos/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
Stem Cells ; 29(12): 2094-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21957033

RESUMEN

Despite the recent identification of surface markers of undifferentiated human embryonic stem cells (hESCs), the crucial cell-surface molecules that regulate the self-renewal capacity of hESCs remain largely undefined. Here, we generated monoclonal antibodies (MAbs) that specifically bind to undifferentiated hESCs but not to mouse embryonic stem cells. Among these antibodies, we selected a novel MAb, 4-63, and identified its target antigen as the L1 cell adhesion molecule (L1CAM) isoform 2. Notably, L1CAM expressed in hESCs lacked the neuron-specific YEGHH and RSLE peptides encoded by exons 2 and 27, respectively. L1CAM colocalized with hESC-specific cell-surface markers, and its expression was markedly downregulated on differentiation. Stable L1CAM depletion markedly decreased hESC proliferation, whereas L1CAM overexpression increased proliferation. In addition, the expression of octamer-binding transcription factor 4, Nanog, sex-determining region Y-box 2, and stage-specific embryonic antigen (SSEA)-3 was markedly downregulated, whereas lineage-specific markers and SSEA-1 were upregulated in L1CAM-depleted hESCs. Interestingly, the actions of L1CAM in regulating the proliferation and differentiation of hESCs were exerted predominantly through the fibroblast growth factor receptor 1 signaling pathway. Taken together, our results suggest that L1CAM is a novel cell-surface molecule that plays an important role in the maintenance of self-renewal and pluripotency in hESCs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/genética , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Biomarcadores/química , Línea Celular , Proliferación Celular , Células Madre Embrionarias/citología , Exones , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunoprecipitación , Ratones , Proteína Homeótica Nanog , Isoformas de Proteínas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Análisis de Secuencia de Proteína , Transducción de Señal , Antígenos Embrionarios Específico de Estadio/genética , Antígenos Embrionarios Específico de Estadio/metabolismo
19.
Arthritis Rheum ; 63(10): 3010-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21953087

RESUMEN

OBJECTIVE: This study was undertaken to generate and characterize human induced pluripotent stem cells (PSCs) from patients with osteoarthritis (OA) and to examine whether these cells can be developed into disease-relevant cell types for use in disease modeling and drug discovery. METHODS: Human synovial cells isolated from two 71-year-old women with advanced OA were characterized and reprogrammed into induced PSCs by ectopic expression of 4 transcription factors (Oct-4, SOX2, Klf4, and c-Myc). The pluripotency status of each induced PSC line was validated by comparison with human embryonic stem cells (ESCs). RESULTS: We found that OA patient-derived human synovial cells had human mesenchymal stem cell (MSC)-like characteristics, as indicated by the expression of specific markers, including CD14-, CD19-, CD34-, CD45-, CD44+, CD51+, CD90+, CD105+, and CD147+. Microarray analysis of human MSCs and human synovial cells further determined their unique and overlapping gene expression patterns. The pluripotency of established human induced PSCs was confirmed by their human ESC-like morphology, expression of pluripotency markers, gene expression profiles, epigenetic status, normal karyotype, and in vitro and in vivo differentiation potential. The potential of human induced PSCs to differentiate into distinct mesenchymal cell lineages, such as osteoblasts, adipocytes, and chondrocytes, was further confirmed by positive expression of markers for respective cell types and positive staining with alizarin red S (osteoblasts), oil red O (adipocytes), or Alcian blue (chondrocytes). Functional chondrocyte differentiation of induced PSCs in pellet culture and 3-dimensional polycaprolactone scaffold culture was assessed by chondrocyte self-assembly and histology. CONCLUSION: Our findings indicate that patient-derived synovial cells are an attractive source of MSCs as well as induced PSCs and have the potential to advance cartilage tissue engineering and cell-based models of cartilage defects.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Osteoartritis/patología , Membrana Sinovial/patología , Adipocitos/citología , Adipocitos/metabolismo , Anciano , Antígenos CD/metabolismo , Linaje de la Célula , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Osteoartritis/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Membrana Sinovial/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA