Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cornea ; 38 Suppl 1: S34-S41, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31403532

RESUMO

In its early stages, an embryo polarizes to form cell subpopulations that subsequently produce specific organ cell types. These cell subpopulations are defined by transcription factors (TFs) that activate or repress specific genes. Although an embryo comprises thousands of TFs, surprisingly few are needed to determine the fate of a given cell. The ectoderm divides into the neuroectoderm and surface ectoderm, the latter of which gives rise to epidermal keratinocytes and corneal epithelial cells (CECs). Meanwhile, neuroectoderm cells give rise to other parts of the eye such as the corneal endothelium and retina. To investigate the regulatory role of TFs in CECs, we overexpressed the "core TFs" (PAX6, OVOL2, and KLF4) in human fibroblasts and found that the cells adopted a CEC-like quality. OVOL2 overexpression was even able to directly induce cells with a neuroectoderm fate toward a surface ectoderm fate, designated "direct reprogramming." Conversely, suppression of OVOL2 or PAX6 expression induced CECs to show qualities consistent with neural lineage cells or epidermal keratinocytes, respectively. This suggests that these core TFs can maintain the CEC phenotype through reciprocal gene regulation. Direct reprogramming has important implications for cell therapies. The potential benefits of cells derived by direct reprogramming compared with induced pluripotent stem cells include the fact that it requires less time than reprogramming a cell back to the pluripotent state and then to another cell type. Further understanding of the reciprocally repressive mechanism of action for core TFs could lead to alternative treatments for regenerative medicine not requiring cell transplantation.


Assuntos
Epitélio Corneano/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Fator de Transcrição PAX6/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Linhagem da Célula , Epitélio Corneano/citologia , Redes Reguladoras de Genes , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Fator de Transcrição PAX6/biossíntese , Fatores de Transcrição/biossíntese
2.
Nat Commun ; 9(1): 1387, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643333

RESUMO

Multicellular organisms consist of multiple cell types. The identity of these cells is primarily maintained by cell-type-specific gene expression programs; however, mechanisms that suppress these programs are poorly defined. Here we show that serum response factor (Srf), a transcription factor that is activated by various extracellular stimuli, can repress cell-type-specific genes and promote cellular reprogramming to pluripotency. Manipulations that decrease ß-actin monomer quantity result in the nuclear accumulation of Mkl1 and the activation of Srf, which downregulate cell-type-specific genes and alter the epigenetics of regulatory regions and chromatin organization. Mice overexpressing Srf exhibit various pathologies including an ulcerative colitis-like symptom and a metaplasia-like phenotype in the pancreas. Our results demonstrate an unexpected function of Srf via a mechanism by which extracellular stimuli actively destabilize cell identity and suggest Srf involvement in a wide range of diseases.


Assuntos
Cromatina/química , Colite Ulcerativa/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Metaplasia/genética , Células-Tronco Neurais/metabolismo , Pâncreas/metabolismo , Fator de Resposta Sérica/genética , Actinas/genética , Actinas/metabolismo , Animais , Diferenciação Celular , Reprogramação Celular/genética , Cromatina/metabolismo , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Pâncreas/patologia , Fator de Resposta Sérica/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo
3.
Exp Eye Res ; 154: 30-38, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27818314

RESUMO

PAX6, a paired box transcription factor, is necessary for eye development. However, how it regulates the cell identity of human corneal epithelial cells (CECs) is not well understood. We aimed to clarify the function of PAX6 in human CECs using gene knockout via the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR associated protein 9 (Cas9) system. We designed guide RNAs for different targets in PAX6. PAX6-depleted CECs maintained the epithelial morphology, but became larger. Global analyses using microarray revealed that down-regulated genes were primarily CEC-specific and included keratin 12, keratin 3, clusterin (CLU), aldehyde dehydrogenase 3 family member A1 (ALDH3A1), angiopoietin-like 7 (ANGPTL7) and transketolase (TKT), while up-regulated genes were primarily epidermis-related and included keratin 10, keratin 1, involucrin (IVL), filaggrin (FLG). These findings suggest that PAX6 maintains CEC identity by regulating differentiation.


Assuntos
Epitélio Corneano/metabolismo , Regulação da Expressão Gênica , Fator de Transcrição PAX6/genética , RNA/genética , Western Blotting , Diferenciação Celular , Epitélio Corneano/citologia , Proteínas Filagrinas , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Análise em Microsséries , Fator de Transcrição PAX6/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Cell Rep ; 15(6): 1359-68, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27134177

RESUMO

In development, embryonic ectoderm differentiates into neuroectoderm and surface ectoderm using poorly understood mechanisms. Here, we show that the transcription factor OVOL2 maintains the transcriptional program of human corneal epithelium cells (CECs), a derivative of the surface ectoderm, and that OVOL2 may regulate the differential transcriptional programs of the two lineages. A functional screen identified OVOL2 as a repressor of mesenchymal genes to maintain CECs. Transduction of OVOL2 with several other transcription factors induced the transcriptional program of CECs in fibroblasts. Moreover, neuroectoderm derivatives were found to express mesenchymal genes, and OVOL2 alone could induce the transcriptional program of CECs in neural progenitors by repressing these genes while activating epithelial genes. Our data suggest that the difference between the transcriptional programs of some neuroectoderm- and surface ectoderm-derivative cells may be regulated in part by a reciprocally repressive mechanism between epithelial and mesenchymal genes, as seen in epithelial-to-mesenchymal transition.


Assuntos
Transição Epitelial-Mesenquimal/genética , Epitélio Corneano/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células Epiteliais/metabolismo , Epitélio Corneano/citologia , Epitélio Corneano/crescimento & desenvolvimento , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesoderma/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(16): 6412-7, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23550161

RESUMO

Transcription factors (TFs) are able to regulate differentiation-related processes, including dedifferentiation and direct conversion, through the regulation of cell type-specific transcriptional profiles. However, the functional interactions between the TFs regulating different transcriptional profiles are not well understood. Here, we show that the TFs capable of inducing cell type-specific transcriptional profiles prevent the dedifferentiation induced by TFs for pluripotency. Of the large number of TFs expressed in a neural-lineage cell line, we identified a subset of TFs that, when overexpressed, strongly interfered with the dedifferentiation triggered by the procedure to generate induced pluripotent stem cells. This interference occurred through a maintenance mechanism of the cell type-specific transcriptional profile. Strikingly, the maintenance activity of the interfering TF set was strong enough to induce the cell line-specific transcriptional profile when overexpressed in a heterologous cell type. In addition, the TFs that interfered with dedifferentiation in hepatic-lineage cells involved TFs with known induction activity for hepatic-lineage cells. Our results suggest that dedifferentiation suppresses a cell type-specific transcriptional profile, which is primarily maintained by a small subset of TFs capable of inducing direct conversion. We anticipate that this functional correlation might be applicable in various cell types and might facilitate the identification of TFs with induction activity in efforts to understand differentiation.


Assuntos
Desdiferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Primers do DNA/genética , Perfilação da Expressão Gênica , Hepatócitos/citologia , Camundongos , Microscopia Eletrônica de Transmissão , Neurônios/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/genética , RNA Interferente Pequeno/genética
6.
Development ; 137(17): 2841-7, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20659973

RESUMO

Mammalian parthenogenetic embryos invariably die in mid-gestation from imprinted gene defects and placental hypoplasia. Based on chimera experiments, trophoblastic proliferation is supposed to be inhibited in the absence of a male genome. Here, we show that parthenogenetic mouse embryonic cell nuclei can be reprogrammed by serial rounds of nuclear transfer without using any genetic modification. The durations of survival in uteri of cloned foetuses derived from green fluorescent protein (GFP)-labelled parthenogenetic cell nuclei were extended with repeated nuclear transfers. After five repeats, live cloned foetuses were obtained up to day 14.5 of gestation; however, they did not survive longer even when we repeated nuclear transfer up to nine times. All foetuses showed intestinal herniation and possessed well-expanded large placentas. When embryonic stem (ES) cells derived from fertilised embryos were aggregated with the cloned embryos, full-term offspring with large placentas were obtained from the chimeric embryos. Those placentas were derived from parthenogenetic cell nuclei, judging from GFP expression. The patterns of imprinted gene expression and methylation status were similar to their parthenogenetic origin, except for Peg10, which showed the same level as in the normal placenta. These results suggest that there is a limitation for foetal development in the ability to reprogramme imprinted genes by repeated rounds of nuclear transfer. However, the placentas of parthenogenetic embryos can escape epigenetic regulation when developed using nuclear transfer techniques and can support foetal development to full gestation.


Assuntos
Técnicas de Transferência Nuclear , Partenogênese/fisiologia , Placentação , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Clonagem de Organismos , Metilação de DNA , Primers do DNA/genética , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Feminino , Expressão Gênica , Impressão Genômica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Partenogênese/genética , Placenta/citologia , Placenta/fisiologia , Células-Tronco Pluripotentes/citologia , Gravidez , Células-Tronco Totipotentes/citologia , Quimeras de Transplante
7.
Reproduction ; 138(2): 309-17, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19433501

RESUMO

Since the birth of Cumulina, the first mouse clone produced by somatic cell nuclear transfer (SCNT), the success rate of cloning in mice has been extremely low compared with other species and most of the inbred mouse strains have never been cloned. Recently, our laboratory has found that treatment of SCNT mouse embryos with trichostatin A, a histone deacetylase inhibitor (HDACi), improved the full-term development of B6D2F1 mouse clones significantly. However, this was not effective for the inbred strains. Here, we show for the first time that by treating SCNT embryos with another HDACi, scriptaid, all the important inbred mouse strains can be cloned, such as C57BL/6, C3H/He, DBA/2, and 129/Sv. Moreover, the success of somatic nuclear reprogramming and cloning efficiency via nuclear transfer technique is clearly linked to the competent de novo synthesis of nascent mRNA in cloned mouse embryos.


Assuntos
Clonagem de Organismos/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inibidores de Histona Desacetilases , Hidroxilaminas/farmacologia , Quinolinas/farmacologia , RNA Mensageiro/biossíntese , Animais , Desenvolvimento Embrionário , Inibidores Enzimáticos/farmacologia , Feminino , Imunofluorescência , Genoma , Camundongos , Camundongos Endogâmicos , Repetições de Microssatélites , Microscopia Confocal , Técnicas de Transferência Nuclear , RNA Mensageiro/análise , Estimulação Química
8.
Hum Mol Genet ; 18(8): 1424-38, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19174477

RESUMO

Mice with maternal duplication of proximal Chromosome 11 (MatDp(prox11)), where Meg1/Grb10 is located, exhibit pre- and postnatal growth retardation. To elucidate the responsible imprinted gene for the growth abnormality, we examined the precise structure and regulatory mechanism of this imprinted region and generated novel model mice mimicking the pattern of imprinted gene expression observed in the MatDp(prox11) by deleting differentially methylated region of Meg1/Grb10 (Meg1-DMR). It was found that Cobl and Ddc, the neighboring genes of Meg1/Grb10, also comprise the imprinted region. We also found that the mouse-specific repeat sequence consisting of several CTCF-binding motifs in the Meg1-DMR functions as a silencer, suggesting that the Meg1/Grb10 imprinted region adopted a different regulatory mechanism from the H19/Igf2 region. Paternal deletion of the Meg1-DMR (+/DeltaDMR) caused both upregulation of the maternally expressed Meg1/Grb10 Type I in the whole body and Cobl in the yolk sac and loss of paternally expressed Meg1/Grb10 Type II and Ddc in the neonatal brain and heart, respectively, demonstrating maternalization of the entire Meg1/Grb10 imprinted region. We confirmed that the +/DeltaDMR mice exhibited the same growth abnormalities as the MatDp(prox11) mice. Fetal and neonatal growth was very sensitive to the expression level of Meg1/Grb10 Type I, indicating that the 2-fold increment of the Meg1/Grb10 Type I is one of the major causes of the growth retardation observed in the MatDp(prox11) and +/DeltaDMR mice. This suggests that the corresponding human GRB10 Type I plays an important role in the etiology of Silver-Russell syndrome caused by partial trisomy of 7p11-p13.


Assuntos
Cromossomos Humanos Par 11/metabolismo , Proteína Adaptadora GRB10/genética , Impressão Genômica , Transtornos do Crescimento/genética , Animais , Feminino , Inativação Gênica , Humanos , Masculino , Camundongos
9.
Proc Natl Acad Sci U S A ; 105(45): 17318-22, 2008 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-18981419

RESUMO

Cloning animals by nuclear transfer provides an opportunity to preserve endangered mammalian species. However, it has been suggested that the "resurrection" of frozen extinct species (such as the woolly mammoth) is impracticable, as no live cells are available, and the genomic material that remains is inevitably degraded. Here we report production of cloned mice from bodies kept frozen at -20 degrees C for up to 16 years without any cryoprotection. As all of the cells were ruptured after thawing, we used a modified cloning method and examined nuclei from several organs for use in nuclear transfer attempts. Using brain nuclei as nuclear donors, we established embryonic stem cell lines from the cloned embryos. Healthy cloned mice were then produced from these nuclear transferred embryonic stem cells by serial nuclear transfer. Thus, nuclear transfer techniques could be used to "resurrect" animals or maintain valuable genomic stocks from tissues frozen for prolonged periods without any cryopreservation.


Assuntos
Clonagem de Organismos/métodos , Criopreservação/métodos , Camundongos , Técnicas de Transferência Nuclear , Animais , Encéfalo/citologia , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/citologia
10.
Stem Cells ; 26(3): 783-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18192228

RESUMO

Recent cloning technology has been demonstrated successfully using nuclear transfer (NT) techniques to generate embryonic stem (ES) cells. Mice can be cloned from adult somatic cells or ES cells by NT, and such cloned embryos can be used to establish new NT-ES cell lines. However, ES cells derived from parthenogenetic embryos show epigenetic disorders and low potential for normal differentiation unless used to produce subsequent generations of NT-ES lines. Thus, enucleated oocytes can initialize epigenetic modification, but the extent and efficacy of this remain unclear. In this study, our goal was to clarify why the contribution rate of ES cells derived from parthenogenetic embryos (pES) cells appears to improve after NT. We compared gene expression profiles between pES and NT-pES cell lines using DNA microarray analysis and allele-specific DNA methylation analysis. Although changes in expression level were observed for 4% of 34,967 genes, only 81 (0.2%) showed common changes across multiple cell lines. In particular, the expression level of a paternally expressed gene, U2af1-rs1, was significantly increased in all NT-pES cell lines investigated. The methylation status at the upstream differentially methylated region of U2af1-rs1 was also changed significantly after NT. This was observed in NT-pES cells, but also in conventionally produced NT-ES cells, which has never been reported previously. These results suggest that NT affects the epigenetic status of a few gene regions in common and that a change in the methylation status of U2af1-rs1 could be used as a genetic marker to investigate the effects of NT.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Fertilização/genética , Técnicas de Transferência Nuclear , Partenogênese/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Proteínas/genética , Proteínas/metabolismo
11.
Reproduction ; 133(2): 371-82, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17307905

RESUMO

Oocyte growth, maturation, and activation are complex processes that include transcription, heterochromatin formation, chromosome condensation and decondensation, two consecutive chromosome separations, and genomic imprinting. The objective of this study was to investigate changes in histone H3 modifications in relation to chromatin/chromosome morphology in pig oocytes during their growth, maturation, and activation. During the growth phase, histone H3 was acetylated at lysines 9, 14, and 18 (K9, K14, and K18), and became methylated at K9 when the follicles developed to the antral stage (oocyte diameter, 90 mum). When the fully grown oocytes (diameter, 120 mum) started their maturation, histone H3 became phosphorylated at serine 28 (S28) and then at S10, and deacetylated at K9, K14, and K18 as the chromosomes condensed. After the electroactivation of mature oocytes, histone H3 was reacetylated and dephosphorylated concomitant with the decondensation of the chromosomes. Histone H3 kinase activity increased over a similar time course to that of the phosphorylation of histone H3-S28 during oocyte maturation, and this activity decreased as histone H3-S10 and H3-S28 began to be dephosphorylated after the activation of the mature oocytes. These results suggest that the chromatin morphology of pig oocytes is regulated by the acetylation/deacetylation and the phosphorylation/dephosphorylation of histone H3, and the phosphorylation of histone H3 is the key event in meiotic chromosome condensation in oocytes. The inhibition of histone deacetylase with trichostatin A (TSA) inhibited the deacetylation and phosphorylation of histone H3, and chromosome condensation. Therefore, the deacetylation of histone H3 is thought to be required for its phosphorylation in meiosis. Although histone H3 acetylation and phosphorylation were reversible, the histone methylation that was established during the oocyte growth phase was stable throughout the course of oocyte maturation and activation.


Assuntos
Montagem e Desmontagem da Cromatina , Cromossomos/ultraestrutura , Histonas/metabolismo , Oócitos/metabolismo , Oogênese/fisiologia , Acetilação , Animais , Células Cultivadas , Cromatina/genética , Metilação de DNA , Feminino , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Microscopia de Fluorescência , Fosforilação , Suínos
12.
J Reprod Dev ; 53(1): 165-70, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17077581

RESUMO

Although the somatic cloning technique has been used for numerous applications and basic research of reprogramming in various species, extremely low success rates have plagued this technique for a decade. Further in mice, the "clonable" strains have been limited to mainly hybrid F1 strains such as B6D2F1. Recently, we established a new efficient cloning technique using trichostatin A (TSA) which leads to a 2-5 fold increase in success rates for mouse cloning of B6D2F1 cumulus cells. To further test the validity of this TSA cloning technique, we tried to clone the adult ICR mouse, an outbred strain, which has never been directly cloned before. Only when TSA was used did we obtain both male and female cloned mice from cumulus and fibroblast cells of adult ICR mice with 4-5% success rates, which is comparable to 5-7% of B6D2F1. Thus, the TSA treatment is the first cloning technique to allow us to successfully clone outbred mice, demonstrating that this technique not only improves the success rates of cloning from hybrid strains, but also enables mouse cloning from normally "unclonable" strains.


Assuntos
Clonagem de Organismos/métodos , Inibidores Enzimáticos , Ácidos Hidroxâmicos , Técnicas de Transferência Nuclear , Animais , Animais não Endogâmicos , Feminino , Fibroblastos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Especificidade da Espécie
13.
Stem Cells ; 25(4): 986-93, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17185608

RESUMO

Recently, ES cell lines were established from single blastomeres taken from eight-cell embryos in mice and humans with success rates of 4% and 2%, respectively, which suggests that the method could be used in regenerative medicine to reduce ethical concerns over harm to embryos. However, those studies used other ES cells as supporting cells. Here, we report a simple and highly efficient method of establishing mouse ES cell lines from single blastomeres, in which single blastomeres are simply plated onto a feeder layer of mouse embryonic fibroblasts with modified ES cell medium. A total of 112 ES cell lines were established from two-cell (establishment rate, 50%-69%), early four-cell (28%-40%), late four-cell (22%), and eight-cell (14%-16%) stage embryos. We also successfully established 18 parthenogenetic ES cell lines from first (36%-40%) and second polar bodies (33%), the nuclei of which were reconstructed to embryos by nuclear transfer. Most cell lines examined maintained normal karyotypes and expressed markers of pluripotency, including germline transmission in chimeric mice. Our results suggest that the single cells of all early-stage embryos or polar bodies have the potential to be converted into ES cells without any special treatment.


Assuntos
Blastômeros/citologia , Células-Tronco Embrionárias/citologia , Oócitos/citologia , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Técnicas de Transferência Nuclear , Partenogênese
14.
Stem Cells ; 25(1): 46-53, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008422

RESUMO

Parthenogenesis is the process by which an oocyte develops into an embryo without being fertilized by a spermatozoon. Although such embryos lack the potential to develop to full term, they can be used to establish parthenogenetic embryonic stem (pES) cells for autologous cell therapy in females without needing to destroy normally competent embryos. Unfortunately, the capacity for further differentiation of these pES cells in vivo is very poor. In this study, we succeeded in improving the potential of pES cells using a nuclear transfer (NT) technique. The original pES cell nuclei were transferred into enucleated oocytes, and the resulting NT embryos were used to establish new NT-pES cell lines. We established 84 such lines successfully (78% from blastocysts, 12% from oocytes). All examined cell lines were positive for several ES cell markers and had a normal extent of karyotypes, except for one original pES cell line and its NT-pES cell derivatives, in which all nuclei were triploid. The DNA methylation status of the differentially methylated domain H19 and differentially methylated region IG did not change after NT. However, the in vivo and in vitro differentiation potentials of NT-pES cells were significantly (two to five times) better than the original pES cells, judged by the production of chimeric mice and by in vitro differentiation into neuronal and mesodermal cell lines. Thus, NT could be used to improve the potential of pES cells and may enhance that of otherwise poor-quality ES cells. It also offers a new tool for studying epigenetics.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Técnicas de Transferência Nuclear , Oócitos/fisiologia , Partenogênese/fisiologia , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Quimera/genética , Feminino , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Cariotipagem , Camundongos , Oócitos/citologia
15.
Reproduction ; 132(6): 849-57, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17127745

RESUMO

The success rate is generally higher when cloning mice from embryonic stem (ES) cell nuclei than from somatic cell nuclei, suggesting that the embryonic nature or the undifferentiated state of the donor cell increases cloning efficiency. We assessed the developmental ability of cloned embryos derived from cultured neural stem cell (NSC) nuclei and compared the success rate with that of embryos cloned from other donor cells such as differentiated NSCs, cumulus cells, Sertoli cells and ES cells in the mouse. The transfer of two-cell cloned embryos derived from cultured NSC nuclei into surrogate mothers produced five live cloned mice. However, the success rate (0.5%) was higher in embryos cloned from cultured NSC nuclei than from differentiated NSCs (0%), but lower than that obtained by cloning mice from other cell nuclei (2.2-3.5%). Although the in vitro developmental potential to the two-cell stage of the cloned embryos derived from NSC nuclei (73%) was similar to that of the cloned embryos derived from other somatic cell nuclei (e.g., 85% in Sertoli cells and 75% in cumulus cells), the developmental rate to the morula-blastocyst stage was only 7%. This rate is remarkably lower than that produced from other somatic cells (e.g., 50% in Sertoli cells and 54% in cumulus cells). These results indicate that the undifferentiated state of neural cells does not enhance the cloning efficiency in mice and that the arrest point for in vitro development of cloned embryos depends on the donor cell type.


Assuntos
Clonagem de Organismos/métodos , Células-Tronco Embrionárias , Neurônios , Técnicas de Transferência Nuclear , Oócitos , Animais , Células Cultivadas , Transferência Embrionária , Desenvolvimento Embrionário , Feminino , Masculino , Camundongos , Camundongos Endogâmicos ICR , Células de Sertoli , Células-Tronco
16.
Biochem Biophys Res Commun ; 349(1): 106-14, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16938271

RESUMO

Somatic cell nuclear transfer technology has been applied to produce live clones successfully in several mammalian species, but the success rates are very low. In mice, about half of the nuclear transfer embryos undergo implantation, but very few survive to term. We undertook detailed histological analyses of placentas from cloned mouse embryos generated from cumulus cells at 10.5 dpc of pregnancy, by which stage most clones have terminated their development. At 10.5 dpc, the extraembryonic tissues displayed several defined histological patterns, each reflecting their stage of developmental arrest. The most notable abnormality was the poor development of the spongiotrophoblast layer of diploid cells. This is in contrast to the placental hyperplasia frequently observed in somatic clones at 12.5 dpc or later stages. A variety of structural abnormalities were also observed in the embryos. Both placental and embryonic defects likely contribute to the low success rate of the mouse clones.


Assuntos
Membrana Corioalantoide/metabolismo , Clonagem de Organismos/métodos , Técnicas de Cultura Embrionária , Placenta/metabolismo , Animais , Núcleo Celular/metabolismo , Desenvolvimento Embrionário , Feminino , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Doenças Placentárias/genética , Gravidez , Prenhez
17.
Stem Cells ; 24(9): 2023-33, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16690779

RESUMO

Therapeutic cloning, whereby nuclear transfer (NT) is used to generate embryonic stem cells (ESCs) from blastocysts, has been demonstrated successfully in mice and cattle. However, if NT-ESCs have abnormalities, such as those associated with the offspring produced by reproductive cloning, their scientific and medical utilities might prove limited. To evaluate the characteristics of NT-ESCs, we established more than 150 NT-ESC lines from adult somatic cells of several mouse strains. Here, we show that these NT-ESCs were able to differentiate into all functional embryonic tissues in vivo. Moreover, they were identical to blastocyst-derived ESCs in terms of their expression of pluripotency markers in the presence of tissue-dependent differentially DNA methylated regions, in DNA microarray profiles, and in high-coverage gene expression profiling. Importantly, the NT procedure did not cause irreversible damage to the nuclei. These similarities of NT-ESCs and ESCs indicate that murine therapeutic cloning by somatic cell NT can provide a reliable model for preclinical stem cell research.


Assuntos
Blastocisto/metabolismo , Núcleo Celular/metabolismo , Embrião de Mamíferos/citologia , Fertilização/fisiologia , Células-Tronco Pluripotentes/citologia , Criação de Embriões para Pesquisa , Animais , Blastocisto/citologia , Bovinos , Diferenciação Celular , Metilação de DNA , Feminino , Perfilação da Expressão Gênica , Cariotipagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Análise em Microsséries
18.
FEBS Lett ; 580(7): 1801-6, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16513118

RESUMO

To examine the establishment and maintenance of trophectoderm (TE) lineage in somatic cloned blastocysts, the expression of Cdx2, a key molecule for specification of TE fate, was immunohistochemically examined simultaneously with Oct4 expression. Cloned mouse embryos were made by nuclear transfer using cumulus cells, tail-tip fibroblasts, and embryonic stem cells. After 96 h of culture, the rates of Oct4-expressing blastocysts were as low as 50% and 60% for cumulus and fibroblast clones, respectively. However, regardless of Oct4 expression, the majority of those cloned blastocysts (> 90%) normally expressed Cdx2. Thus, even though somatic cloned embryos have reduced potential to produce the inner cell mass lineage, the TE lineage can be established and maintained.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/citologia , Animais , Blastocisto/citologia , Fator de Transcrição CDX2 , Técnicas de Cultura de Células , Núcleo Celular , Células Clonais/citologia , Proteínas de Homeodomínio/análise , Camundongos , Camundongos Endogâmicos C57BL , Fator 3 de Transcrição de Octâmero/análise , Células-Tronco/citologia , Fatores de Transcrição/análise
19.
Biol Reprod ; 74(5): 865-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16436529

RESUMO

This study investigated the effects on fertilized embryo development of somatic cytoplasm after its injection into intact mouse oocytes. Mature oocytes collected from female B6D2F1 mice were injected with cumulus cell cytoplasm of different volumes and from different mouse strains (B6D2F1, ICR, and C57BL/6), or with embryonic cytoplasm. After culture for 1 h, B6D2F1 sperm were injected into those oocytes by intracytoplasmic sperm injection (ICSI). The oocytes were examined for pre- and postimplantation developmental competence. Increases in the volume of the somatic cytoplasm from onefold to fourfold resulted in an impairment of blastocyst development and full-term development (28% and 7%, respectively, vs. 96% and 63%, respectively, in the control group; P < 0.01). An increase in the volume of somatic cytoplasm reduced the expression of POU5F1 (more commonly known as OCT4) in expanded blastocysts. The frequency of embryos that developed to the blastocyst stage did not differ when B6D2F1 or ICR somatic cytoplasm was injected, but injection of C57BL/6 somatic cytoplasm induced a two-cell block in embryo development. Injection of the cytoplasm from fertilized embryos did not reduce the frequency of embryos attaining full-term development. Interestingly, somatic cytoplasm significantly increased the placental weight of ICSI embryos, even the injection of onefold cytoplasm (0.20 +/- 0.02 [n = 32] vs. 0.12 +/- 0.02 in the control group [n = 87]; P < 0.01). These findings indicate that the injection of somatic cytoplasm into oocytes before ICSI causes a decrease in preimplantation development, clearly impairs full-term development, and causes placental overgrowth in fertilized embryos. To our knowledge, placental overgrowth phenotypes are only caused by interspecies hybridization and cloning, and in genetically modified mice. Here, we report for the first time that somatic cytoplasm causes abnormal placentas in fertilized embryos. This study suggests that somatic cell cytoplasmic material is one cause of the low rate of full-term development in cloned mammals.


Assuntos
Citoplasma/transplante , Desenvolvimento Embrionário/fisiologia , Oócitos/crescimento & desenvolvimento , Placentação , Animais , Blastocisto/metabolismo , Blastômeros/fisiologia , Citoplasma/fisiologia , Feminino , Desenvolvimento Fetal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Microinjeções , Fator 3 de Transcrição de Octâmero/metabolismo , Gravidez , Injeções de Esperma Intracitoplásmicas
20.
Biochem Biophys Res Commun ; 340(1): 183-9, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16356478

RESUMO

The low success rate of animal cloning by somatic cell nuclear transfer (SCNT) is believed to be associated with epigenetic errors including abnormal DNA hypermethylation. Recently, we elucidated by using round spermatids that, after nuclear transfer, treatment of zygotes with trichostatin A (TSA), an inhibitor of histone deacetylase, can remarkably reduce abnormal DNA hypermethylation depending on the origins of transferred nuclei and their genomic regions [S. Kishigami, N. Van Thuan, T. Hikichi, H. Ohta, S. Wakayama. E. Mizutani, T. Wakayama, Epigenetic abnormalities of the mouse paternal zygotic genome associated with microinsemination of round spermatids, Dev. Biol. (2005) in press]. Here, we found that 5-50 nM TSA-treatment for 10 h following oocyte activation resulted in more efficient in vitro development of somatic cloned embryos to the blastocyst stage from 2- to 5-fold depending on the donor cells including tail tip cells, spleen cells, neural stem cells, and cumulus cells. This TSA-treatment also led to more than 5-fold increase in success rate of mouse cloning from cumulus cells without obvious abnormality but failed to improve ES cloning success. Further, we succeeded in establishment of nuclear transfer-embryonic stem (NT-ES) cells from TSA-treated cloned blastocyst at a rate three times higher than those from untreated cloned blastocysts. Thus, our data indicate that TSA-treatment after SCNT in mice can dramatically improve the practical application of current cloning techniques.


Assuntos
Blastocisto/efeitos dos fármacos , Clonagem de Organismos/métodos , Epigênese Genética/efeitos dos fármacos , Ácidos Hidroxâmicos/administração & dosagem , Técnicas de Transferência Nuclear , Oócitos/efeitos dos fármacos , Animais , Núcleo Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...