Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Am J Physiol Renal Physiol ; 326(6): F1066-F1077, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634134

RESUMEN

The epithelial Na+ channel (ENaC) γ subunit is essential for homeostasis of Na+, K+, and body fluid. Dual γ subunit cleavage before and after a short inhibitory tract allows dissociation of this tract, increasing channel open probability (PO), in vitro. Cleavage proximal to the tract occurs at a furin recognition sequence (143RKRR146, in the mouse γ subunit). Loss of furin-mediated cleavage prevents in vitro activation of the channel by proteolysis at distal sites. We hypothesized that 143RKRR146 mutation to 143QQQQ146 (γQ4) in 129/Sv mice would reduce ENaC PO, impair flow-stimulated flux of Na+ (JNa) and K+ (JK) in perfused collecting ducts, reduce colonic amiloride-sensitive short-circuit current (ISC), and impair Na+, K+, and body fluid homeostasis. Immunoblot of γQ4/Q4 mouse kidney lysates confirmed loss of a band consistent in size with the furin-cleaved proteolytic fragment. However, γQ4/Q4 male mice on a low Na+ diet did not exhibit altered ENaC PO or flow-induced JNa, though flow-induced JK modestly decreased. Colonic amiloride-sensitive ISC in γQ4/Q4 mice was not altered. γQ4/Q4 males, but not females, exhibited mildly impaired fluid volume conservation when challenged with a low Na+ diet. Blood Na+ and K+ were unchanged on a regular, low Na+, or high K+ diet. These findings suggest that biochemical evidence of γ subunit cleavage should not be used in isolation to evaluate ENaC activity. Furthermore, factors independent of γ subunit cleavage modulate channel PO and the influence of ENaC on Na+, K+, and fluid volume homeostasis in 129/Sv mice, in vivo.NEW & NOTEWORTHY The epithelial Na+ channel (ENaC) is activated in vitro by post-translational proteolysis. In vivo, low Na+ or high K+ diets enhance ENaC proteolysis, and proteolysis is hypothesized to contribute to channel activation in these settings. Using a mouse expressing ENaC with disruption of a key proteolytic cleavage site, this study demonstrates that impaired proteolytic activation of ENaC's γ subunit has little impact upon channel open probability or the ability of mice to adapt to low Na+ or high K+ diets.


Asunto(s)
Canales Epiteliales de Sodio , Proteolisis , Sodio , Animales , Canales Epiteliales de Sodio/metabolismo , Canales Epiteliales de Sodio/genética , Masculino , Femenino , Sodio/metabolismo , Túbulos Renales Colectores/metabolismo , Homeostasis , Furina/metabolismo , Furina/genética , Ratones , Colon/metabolismo , Potasio/metabolismo , Dieta Hiposódica , Ratones de la Cepa 129 , Mutación , Amilorida/farmacología
2.
bioRxiv ; 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38405735

RESUMEN

The ENaC gamma subunit is essential for homeostasis of Na + , K + , and body fluid. Dual subunit cleavage before and after a short inhibitory tract allows dissociation of this tract, increasing channel open probability (P O ), in vitro . Cleavage proximal to the tract occurs at a furin recognition sequence ( 143 RKRR 146 in mouse). Loss of furin-mediated cleavage prevents in vitro activation of the channel by proteolysis at distal sites. We hypothesized that 143 RKRR 146 mutation to 143 QQQQ 146 ( Q4 ) in 129/Sv mice would reduce ENaC P O , impair flow-stimulated flux of Na + (J Na ) and K + (J K ) in perfused collecting ducts, reduce colonic amiloride-sensitive short circuit current (I SC ), and impair Na + , K + , and body fluid homeostasis. Immunoblot of Q4/Q4 mouse kidney lysates confirmed loss of a band consistent in size with the furin-cleaved proteolytic fragment. However, Q4/Q4 male mice on a low Na + diet did not exhibit altered ENaC P O or flow-induced J Na , though flow-induced J K modestly decreased. Colonic amiloride-sensitive I SC in Q4/Q4 mice was not altered. Q4/Q4 males, but not females, exhibited mildly impaired fluid volume conservation when challenged with a low Na + diet. Blood Na + and K + were unchanged on a regular, low Na + , or high K + diet. These findings suggest that biochemical evidence of gamma subunit cleavage should not be used in isolation to evaluate ENaC activity. Further, factors independent of gamma subunit cleavage modulate channel P O and the influence of ENaC on Na + , K + , and fluid volume homeostasis in 129/Sv mice, in vivo .

3.
Cell Rep ; 21(13): 3957-3969, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281840

RESUMEN

Pre-implantation embryo development is an intricate and precisely regulated process orchestrated by maternally inherited proteins and newly synthesized proteins following zygotic genome activation. Although genomic and transcriptomic studies have enriched our understanding of the genetic programs underlying this process, the protein expression landscape remains unexplored. Using quantitative mass spectrometry, we identified nearly 5,000 proteins from 8,000 mouse embryos of each stage (zygote, 2-cell, 4-cell, 8-cell, morula, and blastocyst). We found that protein expression in zygotes, morulas, and blastocysts is distinct from 2- to 8-cell embryos. Analysis of protein phosphorylation identified critical kinases and signal transduction pathways. We highlight key factors and their important roles in embryo development. Combined analysis of transcriptomic and proteomic data reveals coordinated control of RNA degradation, transcription, and translation and identifies previously undefined exon-junction-derived peptides. Our study provides an invaluable resource for further mechanistic studies and suggests core factors regulating pre-implantation embryo development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Proteínas/metabolismo , Animales , Desarrollo Embrionario/genética , Exones/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Masculino , Ratones Endogámicos C57BL , Fosfoproteínas/metabolismo , Fosforilación , Biosíntesis de Proteínas , Mapas de Interacción de Proteínas , Proteoma/metabolismo , Proteómica , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transcriptoma/genética
4.
Oncotarget ; 8(29): 47344-47355, 2017 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-28476045

RESUMEN

Somatic cell nuclear transfer and transcription factor mediated reprogramming are two widely used techniques for somatic cell reprogramming. Both fully reprogrammed nuclear transfer embryonic stem cells and induced pluripotent stem cells hold potential for regenerative medicine, and evaluation of the stem cell pluripotency state is crucial for these applications. Previous reports have shown that the Dlk1-Dio3 region is associated with pluripotency in induced pluripotent stem cells and the incomplete somatic cell reprogramming causes abnormally elevated levels of genomic 5-methylcytosine in induced pluripotent stem cells compared to nuclear transfer embryonic stem cells and embryonic stem cells. In this study, we compared pluripotency associated genes Rian and Gtl2 in the Dlk1-Dio3 region in exactly syngeneic nuclear transfer embryonic stem cells and induced pluripotent stem cells with same genomic insertion. We also assessed 5-methylcytosine and 5-hydroxymethylcytosine levels and performed high-throughput sequencing in these cells. Our results showed that Rian and Gtl2 in the Dlk1-Dio3 region related to pluripotency in induced pluripotent stem cells did not correlate with the genes in nuclear transfer embryonic stem cells, and no significant difference in 5-methylcytosine and 5-hydroxymethylcytosine levels were observed between fully and partially reprogrammed nuclear transfer embryonic stem cells and induced pluripotent stem cells. Through syngeneic comparison, our study identifies for the first time that Grb10 is associated with the pluripotency state in nuclear transfer embryonic stem cells.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteína Adaptadora GRB10/genética , Regulación del Desarrollo de la Expresión Génica , Tejido Adiposo/citología , Animales , Biomarcadores , Transdiferenciación Celular/genética , Técnicas de Reprogramación Celular , Metilación de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Ratones , Fenotipo
6.
PLoS Genet ; 10(10): e1004589, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25340332

RESUMEN

Distal arthrogryposis type 2B (DA2B) is an important genetic disorder in humans. However, the mechanisms governing this disease are not clearly understood. In this study, we generated knock-in mice carrying a DA2B mutation (K175del) in troponin I type 2 (skeletal, fast) (TNNI2), which encodes a fast-twitch skeletal muscle protein. Tnni2K175del mice (referred to as DA2B mice) showed typical DA2B phenotypes, including limb abnormality and small body size. However, the current knowledge concerning TNNI2 could not explain the small body phenotype of DA2B mice. We found that Tnni2 was expressed in the osteoblasts and chondrocytes of long bone growth plates. Expression profile analysis using radii and ulnae demonstrated that Hif3a expression was significantly increased in the Tnni2K175del mice. Chromatin immunoprecipitation assays indicated that both wild-type and mutant tnni2 protein can bind to the Hif3a promoter using mouse primary osteoblasts. Moreover, we showed that the mutant tnni2 protein had a higher capacity to transactivate Hif3a than the wild-type protein. The increased amount of hif3a resulted in impairment of angiogenesis, delay in endochondral ossification, and decrease in chondrocyte differentiation and osteoblast proliferation, suggesting that hif3a counteracted hif1a-induced Vegf expression in DA2B mice. Together, our data indicated that Tnni2K175del mutation led to abnormally increased hif3a and decreased vegf in bone, which explain, at least in part, the small body size of Tnni2K175del mice. Furthermore, our findings revealed a new function of tnni2 in the regulation of bone development, and the study of gain-of-function mutation in Tnni2 in transgenic mice opens a new avenue to understand the pathological mechanism of human DA2B disorder.


Asunto(s)
Artrogriposis/genética , Desarrollo Óseo/genética , Factores de Transcripción/biosíntesis , Troponina I/genética , Animales , Proteínas Reguladoras de la Apoptosis , Artrogriposis/fisiopatología , Calcio/metabolismo , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Ratones , Contracción Muscular/genética , Mutación , Proteínas Represoras , Sarcómeros/patología , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis
7.
Stem Cells Dev ; 23(19): 2283-96, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24805295

RESUMEN

The deficiency of X-inactive specific transcript (XIST) on the inactive X chromosome affects the behavior of female human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), and further chromosomal erosion can occur with continued passaging of these cells. However, X chromosome instability has not been identified in other species. In the present study, we investigated three female rabbit ESC (rbESC) lines and found that two of them expressed Xist normally and obtained both Xist RNA coating and H3K27me3 foci, thus defined as Xi(Xist)Xa. Interestingly, the third female rbESC line lacked Xist expression during ESC maintenance and differentiation. This line showed H3K27me3 foci but no Xist RNA coating in the early passages and was thus defined as Xi(w/oXist)Xa. Similar to Xi(w/oXist)Xa hESCs or hiPSCs, Xi(w/oXist)Xa rbESCs lose H3K27me3 and undergo Xi erosion (Xe) with passaging. Moreover, Xist-deficient rbESCs also exhibit impaired differentiation ability and upregulation of cancer-related genes. By overexpressing OCT4, SOX2, KLF4, and c-MYC in Xist-deficient rbESCs under optimized culture conditions, we successfully obtained mouse ESC-like (mESC-like) cells. The mESC-like rbESCs displayed dome-shaped colony morphology, activation of the LIF/STAT3-dependent pathway, and conversion of disordered X chromosome. Importantly, the defective differentiation potential was also greatly improved. Our data demonstrate that variations in X chromosome inactivation occur in early passage of rbESCs; thus, Xi disorders are conserved across species and are reversible using the proper epigenetic reprogramming and culture conditions. These findings may be very useful for future efforts toward deriving fully pluripotent rbESCs or rabbit iPSCs (rbiPSCs).


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción/metabolismo , Cromosoma X/metabolismo , Animales , Diferenciación Celular/fisiología , Humanos , Factor 4 Similar a Kruppel , Conejos , Transcripción Genética/fisiología , Regulación hacia Arriba
8.
Cell Stem Cell ; 14(1): 27-39, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24268696

RESUMEN

Although somatic cell nuclear transfer (SCNT) and induction of pluripotency (to form iPSCs) are both recognized reprogramming methods, there has been relatively little comparative analysis of the resulting pluripotent cells. Here, we examine the capacity of these two reprogramming approaches to rejuvenate telomeres using late-generation telomerase-deficient (Terc(-/-)) mice that exhibit telomere dysfunction and premature aging. We found that embryonic stem cells established from Terc(-/-) SCNT embryos (Terc(-/-) ntESCs) have greater differentiation potential and self-renewal capacity than Terc(-/-) iPSCs. Remarkably, SCNT results in extensive telomere lengthening in cloned embryos and improved telomere capping function in the established Terc(-/-) ntESCs. In addition, mitochondrial function is severely impaired in Terc(-/-) iPSCs and their differentiated derivatives but significantly improved in Terc(-/-) ntESCs. Thus, our results suggest that SCNT-mediated reprogramming mitigates telomere dysfunction and mitochondrial defects to a greater extent than iPSC-based reprogramming. Understanding the basis of this differential could help optimize reprogramming strategies.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Técnicas de Transferencia Nuclear , ARN/fisiología , Telomerasa/fisiología , Telómero/genética , Adenosina Trifosfato/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Hibridación Fluorescente in Situ , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Placa Neural/metabolismo , Placa Neural/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Stem Cells ; 31(4): 729-40, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23307593

RESUMEN

Leukemia inhibitory factor and bone morphogenetic protein signaling pathways play important roles in maintaining the self-renewal of mouse embryonic stem cells (ESCs). In contrast, the supplementation of fibroblast growth factor 2 (FGF2) in culture promotes mouse ESC differentiation. It has been proposed that factors that are adverse for maintaining the self-renewal of ESCs might play detrimental roles in the transcription factor-mediated reprogramming of somatic cells to pluripotency. However, recent evidence has revealed that reprogramming efficiency could be improved by FGF2, while the underlying molecular mechanism remains unknown. In this study, we dissected the roles of FGF2 in promoting mouse fibroblast reprogramming and disclosed the molecular mechanism behind this process. We used both primary induction and secondary inducible reprogramming systems and demonstrated that supplementation with FGF2 in the early phase of induced pluripotent stem cell induction could significantly increase reprogramming efficiency. Moreover, we discovered that many extracellular matrix candidate genes were significantly downregulated in fibroblasts treated with FGF2, and in particular, the synthesis of collagen could be greatly reduced by FGF2 treatment. Subsequently, we demonstrated that collagen is a barrier for reprogramming fibroblast cells to pluripotency, and the decreasing of collagen either by collagenase treatment or downregulation of collagen gene expression could significantly improve the reprogramming efficiency. Our results reveal a novel role of the extracellular matrix in mediating fibroblasts reprogramming.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Animales , Western Blotting , Células Cultivadas , Reprogramación Celular/genética , Fibroblastos/efectos de los fármacos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Stem Cells Dev ; 21(14): 2630-41, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22471963

RESUMEN

The mammalian target of the rapamycin (mTOR) signaling pathway functions in many cellular processes, including cell growth, proliferation, differentiation, and survival. Recent advances have demonstrated that differentiated somatic cells can be directly reprogrammed into the pluripotent state by overexpression of several pluripotency transcription factors. However, whether the mTOR signaling pathway is involved in this somatic cell-reprogramming process remains unknown. Here, we provide evidence that an elaborate regulation of the mTOR activity is required for the successful reprogramming of somatic cells to pluripotency. The reprogramming of somatic cells collected from the Tsc2(-/-) embryo, in which the mTOR activity is hyperactivated, is entirely inhibited. By taking advantage of the secondary inducible pluripotent stem (iPS) system, we demonstrate that either elevating the mTOR activity by Tsc2 shRNA knockdown or using high concentrations of rapamycin to completely block the mTOR activity in cells derived from iPS mice greatly impairs somatic cell reprogramming. Secondary iPS induction efficiency can only be elevated by elaborately regulating the mTOR activity. Taken together, our data demonstrate that the precise regulation of the mTOR activity plays a critical role in the successful reprogramming of somatic cells to form iPS cells.


Asunto(s)
Diferenciación Celular , Regulación Enzimológica de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proliferación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/enzimología , Activación Enzimática , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Células Madre Pluripotentes Inducidas/enzimología , Masculino , Ratones , Ratones de la Cepa 129 , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Sirolimus/farmacología , Organismos Libres de Patógenos Específicos , Serina-Treonina Quinasas TOR/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Stem Cells ; 29(5): 755-63, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21305674

RESUMEN

ESCs and trophoblast stem (TS) cells are both derived from early embryos, yet these cells have distinct differentiation properties. ESCs can differentiate into all three germ layer cell types, whereas TS cells can only differentiate into placental cells. It has not been determined whether TS cells can be converted into ES-like pluripotent stem (PS) cells. Here, we report that overexpression of a single transcription factor, Oct4, in TS cells is sufficient to reprogram TS cells into a pluripotent state. These Oct4-induced PS (OiPS) cells have the epigenetic characteristics of ESCs, including X chromosome reactivation, elevated H3K27 me3 modifications, and hypomethylation of promoter regions in Oct4 and Nanog genes. Meanwhile, methylation of promoter region in the Elf5 gene occurred during reprogramming of TS cells. The gene expression profile of OiPS cells was very similar to ESCs. Moreover, OiPS cells can differentiate into the three germ layer cell types in vitro and in vivo. More importantly, chimeric mice with germline transmission could be efficiently produced from OiPS cells. Our results demonstrate that one single transcription factor, Oct4, could reprogram the nonembryonic TS cells into PS cells.


Asunto(s)
Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre/citología , Trofoblastos/citología , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Femenino , Genotipo , Cariotipificación , Ratones , Microscopía Confocal , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Trofoblastos/metabolismo
13.
Stem Cells Dev ; 20(11): 1951-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21322785

RESUMEN

Derivation of embryonic stem cells from patient-specific cloned blastocysts by somatic cell nuclear transfer (SCNT) holds promise for both regenerative medicine and cell-based drug discovery. However, the efficiency of blastocyst formation after human SCNT is very low. The developmental competence of SCNT embryos has been previously demonstrated in several species to be enhanced by treatment with histone deacetylase inhibitors, such as trichostatin A (TSA), to increase histone acetylation. In this study, we report that treatment of SCNT embryos with 5 nM TSA for 10 h following activation incubation increased the developmental competence of human SCNT embryos constructed from ß-thalassemia fibroblast cells. The efficiency of blastocyst formation from SCNT human embryos treated with TSA was approximately 2 times greater than that from untreated embryos. Cloned blastocysts were confirmed to be generated through SCNT by DNA and mitochondrial DNA fingerprinting analyses. Further, treatment of SCNT embryos with TSA improved the acetylation of histone H3 at lysine 9 in a manner similar to that observed in in vitro fertilized embryos.


Asunto(s)
Blastómeros/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Talasemia beta/patología , Acetilación , Secuencia de Bases , Blastocisto/patología , Blastómeros/metabolismo , Forma de la Célula , Células Cultivadas , Clonación de Organismos , Técnicas de Cocultivo , ADN Mitocondrial/genética , Femenino , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Cariotipo , Masculino , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Técnicas de Transferencia Nuclear , Análisis de Secuencia de ADN , Inyecciones de Esperma Intracitoplasmáticas
15.
Proc Natl Acad Sci U S A ; 107(41): 17639-44, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20876089

RESUMEN

The mammalian oocyte possesses powerful reprogramming factors, which can reprogram terminally differentiated germ cells (sperm) or somatic cells within a few cell cycles. Although it has been suggested that use of oocyte-derived transcripts may enhance the generation of induced pluripotent stem cells, the reprogramming factors in oocytes are undetermined, and even the identified proteins composition of oocytes is very limited. In the present study, 7,000 mouse oocytes at different developmental stages, including the germinal vesicle stage, the metaphase II (MII) stage, and the fertilized oocytes (zygotes), were collected. We successfully identified 2,781 proteins present in germinal vesicle oocytes, 2,973 proteins in MII oocytes, and 2,082 proteins in zygotes through semiquantitative MS analysis. Furthermore, the results of the bioinformatics analysis indicated that different protein compositions are correlated with oocyte characteristics at different developmental stages. For example, specific transcription factors and chromatin remodeling factors are more abundant in MII oocytes, which may be crucial for the epigenetic reprogramming of sperm or somatic nuclei. These results provided important knowledge to better understand the molecular mechanisms in early development and may improve the generation of induced pluripotent stem cells.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Oocitos/crecimiento & desarrollo , Proteoma/genética , Cigoto/metabolismo , Animales , Biología Computacional , ADN/metabolismo , Epigénesis Genética/genética , Espectrometría de Masas , Ratones , Oocitos/metabolismo , Proteómica/métodos , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
J Biol Chem ; 285(43): 33113-33122, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20699224

RESUMEN

Nuclear importing system and nuclear factors play important roles in mediating nuclear reprogramming and zygotic gene activation. However, the components and mechanisms that mediate nuclearly specific targeting of the nuclear proteins during nuclear reprogramming and zygotic gene activation remain largely unknown. Here, we identified a novel member of the importin-α family, AW146299(KPNA7), which is predominantly expressed in mouse oocytes and zygotes and localizes to the nucleus or spindle. Mutation of Kpna7 gene caused reproductivity reduction and sex imbalance by inducing preferential fetal lethality in females. Parthenogenesis analysis showed that the cell cycle of activated one-cell embryos is loss of control and ahead of schedule but finally failed to develop into blastocyst stage. Further RT-PCR and epigenetic modification analysis showed that knocking out of Kpna7 induced abnormalities of gene expression (dppa2, dppa4, and piwil2) and epigenetic modifications (down-regulation of histone H3K27me3). Biochemical analysis showed that KPNA7 interacts with KPNB1 (importin-ß1). In summary, we identified a novel Kpna7 gene that is required for normal fertility and fecundity.


Asunto(s)
Fertilidad/fisiología , Cigoto/metabolismo , alfa Carioferinas/metabolismo , Animales , Proteínas Argonautas , Secuencia de Bases , Núcleo Celular/genética , Núcleo Celular/metabolismo , Femenino , Muerte Fetal/genética , Muerte Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Oocitos/citología , Oocitos/metabolismo , Proteínas/genética , Proteínas/metabolismo , Huso Acromático/genética , Huso Acromático/metabolismo , Factores de Transcripción , Cigoto/citología , alfa Carioferinas/genética , beta Carioferinas
17.
J Genet Genomics ; 37(7): 415-21, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20659705

RESUMEN

Embryonic stem cells (ESCs) derived from the early embryos possess two important characteristics: self-renewal and pluripotency, which make ESCs ideal seed cells that could be potentially utilized for curing a number of degenerative and genetic diseases clinically. However, ethical concerns and immune rejection after cell transplantation limited the clinical application of ESCs. Fortunately, the recent advances in induced pluripotent stem cell (iPSC) research have clearly shown that differentiated somatic cells from various species could be reprogrammed into pluripotent state by ectopically expressing a combination of several transcription factors, which are highly enriched in ESCs. This ground-breaking achievement could circumvent most of the limitations that ESCs faced. However, it remains challenging if the iPS cell lines, especially the human iPSCs lines, available are fully pluripotent. Therefore, it is prerequisite to establish a molecular standard to distinguish the better quality iPSCs from the inferior ones.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes/trasplante , Humanos
18.
J Genet Genomics ; 37(7): 431-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20659707

RESUMEN

Embryonic stem cells (ESCs) are a potential source of generating transplantable hematopoietic stem and progenitor cells, which in turn can serve as "seed" cells for hematopoietic regeneration. In this study, we aimed to gauge the ability of mouse ESCs directly differentiating into hematopoietic cells in adult bone marrow (BM). To this end, we first derived a new mouse ESC line that constitutively expressed the green fluorescent protein (GFP) and then injected the ESCs into syngeneic BM via intra-tibia. The progeny of the transplanted ESCs were then analyzed at different time points after transplantation. Notably, however, most injected ESCs differentiated into non-hematopoietic cells in the BM whereas only a minority of the cells acquired hematopoietic cell surface markers. This study provides a strategy for evaluating the differentiation potential of ESCs in the BM micro-environment, thereby having important implications for the physiological maintenance and potential therapeutic applications of ESCs.


Asunto(s)
Médula Ósea/fisiología , Diferenciación Celular , Células Madre Embrionarias/citología , Animales , Células Madre Embrionarias/trasplante , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Ratones
19.
PLoS One ; 5(4): e10320, 2010 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-20436676

RESUMEN

Somatic cell nuclear transfer (SCNT) has been proved capable of reprogramming various differentiated somatic cells into pluripotent stem cells. Recently, induced pluripotent stem cells (iPS) have been successfully derived from mouse and human somatic cells by the over-expression of a combination of transcription factors. However, the molecular mechanisms underlying the reprogramming mediated by either the SCNT or iPS approach are poorly understood. Increasing evidence indicates that many tumor pathways play roles in the derivation of iPS cells. Embryonal carcinoma (EC) cells have the characteristics of both stem cells and cancer cells and thus they might be the better candidates for elucidating the details of the reprogramming process. Although previous studies indicate that EC cells cannot be reprogrammed into real pluripotent stem cells, the reasons for this remain unclear. Here, nuclei from mouse EC cells (P19) were transplanted into enucleated oocytes and pluripotent stem cells (P19 NTES cells) were subsequently established. Interestingly, P19 NTES cells prolonged the development of tetraploid aggregated embryos compared to EC cells alone. More importantly, we found that the expression recovery of the imprinted H19 gene was dependent on the methylation state in the differential methylation region (DMR). The induction of Nanog expression, however, was independent of the promoter region DNA methylation state in P19 NTES cells. A whole-genome transcriptome analysis further demonstrated that P19 NTES cells were indeed the intermediates between P19 cells and ES cells and many interesting genes were uncovered that may be responsible for the failed reprogramming of P19 cells. To our knowledge, for the first time, we linked incomplete reprogramming to the improved pluripotency of EC cell-derived pluripotent stem cells. The candidate genes we discovered may be useful not only for understanding the mechanisms of reprogramming, but also for deciphering the transition between tumorigenesis and pluripotency.


Asunto(s)
Reprogramación Celular , Células Madre de Carcinoma Embrionario/ultraestructura , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes/citología , Animales , Núcleo Celular , Metilación de ADN , Células Madre de Carcinoma Embrionario/citología , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Oocitos , Células Madre Pluripotentes/metabolismo , Activación Transcripcional
20.
Biol Reprod ; 83(2): 238-43, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20427755

RESUMEN

Differentiated somatic cells of various species can be reprogrammed into induced pluripotent stem cells (iPSCs) by ectopically expressing a combination of several transcription factors that are highly enriched in embryonic stem cells (ESCs). The generation of iPSCs in large animals has raised the possibility of producing genetically modified large animals through the nuclear transplantation approach. However, it remains unknown whether iPSCs could be used for generating cloned animals through the nuclear transfer method. Here, we show the successful production of viable cloned mice from inducible iPSCs through the nuclear transfer approach, and the efficiency is similar to that of using ESCs derived via normal fertilization. Furthermore, the cloned mice are fertile and can produce second-generation offspring. These efforts strengthen the possibility of utilizing iPSCs to generate gene-modified large animals for pharmaceutical purposes in the future.


Asunto(s)
Clonación de Organismos/métodos , Técnicas de Transferencia Nuclear , Células Madre Pluripotentes/ultraestructura , Animales , Blastocisto/fisiología , Diferenciación Celular/genética , Desarrollo Embrionario , Células Madre Embrionarias , Femenino , Fertilidad , Expresión Génica , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Factor 3 de Transcripción de Unión a Octámeros/genética , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA