Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Stem Cells ; 36(2): 192-205, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29044892

RESUMO

One strategy for stem cell-based therapy of the cerebral cortex involves the generation and transplantation of functional, histocompatible cortical-like neurons from embryonic stem cells (ESCs). Diploid parthenogenetic Pg-ESCs have recently emerged as a promising source of histocompatible ESC derivatives for organ regeneration but their utility for cerebral cortex therapy is unknown. A major concern with Pg-ESCs is genomic imprinting. In contrast with biparental Bp-ESCs derived from fertilized oocytes, Pg-ESCs harbor two maternal genomes but no sperm-derived genome. Pg-ESCs are therefore expected to have aberrant expression levels of maternally expressed (MEGs) and paternally expressed (PEGs) imprinted genes. Given the roles of imprinted genes in brain development, tissue homeostasis and cancer, their deregulation in Pg-ESCs might be incompatible with therapy. Here, we report that, unexpectedly, only one gene out of 7 MEGs and 12 PEGs was differentially expressed between Pg-ESCs and Bp-ESCs while 13 were differentially expressed between androgenetic Ag-ESCs and Bp-ESCs, indicating that Pg-ESCs but not Ag-ESCs, have a Bp-like imprinting compatible with therapy. In vitro, Pg-ESCs generated cortical-like progenitors and electrophysiologically active glutamatergic neurons that maintained the Bp-like expression levels for most imprinted genes. In vivo, Pg-ESCs participated to the cortical lineage in fetal chimeras. Finally, transplanted Pg-ESC derivatives integrated into the injured adult cortex and sent axonal projections in the host brain. In conclusion, mouse Pg-ESCs generate functional cortical-like neurons with Bp-like imprinting and their derivatives properly integrate into both the embryonic cortex and the injured adult cortex. Collectively, our data support the utility of Pg-ESCs for cortical therapy. Stem Cells 2018;36:192-205.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Animais , Metilação de DNA/genética , Metilação de DNA/fisiologia , Eletrofisiologia , Impressão Genômica/genética , Impressão Genômica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , Partenogênese/genética , Partenogênese/fisiologia
2.
Mol Med ; 21: 185-96, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25811991

RESUMO

Parent-of-origin imprints have been implicated in the regulation of neural differentiation and brain development. Previously we have shown that, despite the lack of a paternal genome, human parthenogenetic (PG) embryonic stem cells (hESCs) can form proliferating neural stem cells (NSCs) that are capable of differentiation into physiologically functional neurons while maintaining allele-specific expression of imprinted genes. Since biparental ("normal") hESC-derived NSCs (N NSCs) are targeted by immune cells, we characterized the immunogenicity of PG NSCs. Flow cytometry and immunocytochemistry revealed that both N NSCs and PG NSCs exhibited surface expression of human leukocyte antigen (HLA) class I but not HLA-DR molecules. Functional analyses using an in vitro mixed lymphocyte reaction assay resulted in less proliferation of peripheral blood mononuclear cells (PBMC) with PG compared with N NSCs. In addition, natural killer (NK) cells cytolyzed PG less than N NSCs. At a molecular level, expression analyses of immune regulatory factors revealed higher HLA-G levels in PG compared with N NSCs. In line with this finding, MIR152, which represses HLA-G expression, is less transcribed in PG compared with N cells. Blockage of HLA-G receptors ILT2 and KIR2DL4 on natural killer cell leukemia (NKL) cells increased cytolysis of PG NSCs. Together this indicates that PG NSCs have unique immunological properties due to elevated HLA-G expression.


Assuntos
Diferenciação Celular , Citotoxicidade Imunológica , Células-Tronco Embrionárias/citologia , Expressão Gênica , Antígenos HLA-G/genética , Células Matadoras Naturais/imunologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Apoptose/genética , Apoptose/imunologia , Linhagem Celular , Regulação da Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Antígenos HLA-DR/metabolismo , Antígenos HLA-G/imunologia , Antígenos HLA-G/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , MicroRNAs/genética , Células-Tronco Neurais/citologia
3.
Stem Cells ; 32(7): 1983-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24740448

RESUMO

Parthenogenesis is the development of an oocyte without fertilization. Mammalian parthenogenetic (PG) embryos are not viable, but can develop into blastocysts from which embryonic stem cells (ESCs) have been derived in mouse and human. PG ESCs are frequently homozygous for alleles encoding major histocompatibility complex (MHC) molecules. MHC homozygosity permits much more efficient immune matching than MHC heterozygosity found in conventional ESCs, making PG ESCs a promising cell source for cell therapies requiring no or little immune suppression. However, findings of restricted differentiation and proliferation of PG cells in developmental chimeras have cast doubt on the potential of PG ESC derivatives for organ regeneration. To address this uncertainty, we determined whether PG ESC derivatives are effective in rescuing mice with lethal liver failure due to deficiency of fumarylacetoacetate hydrolase (Fah). In developmental chimeras generated by injecting wild-type PG ESCs into Fah-deficient blastocysts, PG ESCs differentiated into hepatocytes that could repopulate the liver, provide normal liver function, and facilitate long-term survival of adult mice. Moreover, after transplantation into adult Fah-deficient mice, PG ESC-derived hepatocytes efficiently engrafted and proliferated, leading to high-level liver repopulation. Our results show that--despite the absence of a paternal genome--PG ESCs can form therapeutically effective hepatocytes.


Assuntos
Células-Tronco Embrionárias/transplante , Falência Hepática/terapia , Tirosinemias/terapia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Hepatócitos/fisiologia , Humanos , Fígado/patologia , Fígado/fisiopatologia , Regeneração Hepática , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Partenogênese
4.
Proc Natl Acad Sci U S A ; 107(26): 11841-6, 2010 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-20534472

RESUMO

Piwi-interacting RNAs (piRNAs) are essential for silencing of transposable elements in the germline, but their biogenesis is poorly understood. Here we demonstrate that MOV10L1, a germ cell-specific putative RNA helicase, is associated with Piwi proteins. Genetic disruption of the MOV10L1 RNA helicase domain in mice renders both MILI and MIWI2 devoid of piRNAs. Absence of a functional piRNA pathway in Mov10l1 mutant testes causes loss of DNA methylation and subsequent derepression of retrotransposons in germ cells. The Mov10l1 mutant males are sterile owing to complete meiotic arrest. This mouse mutant expresses Piwi proteins but lacks piRNAs, suggesting that MOV10L1 is required for piRNA biogenesis and/or loading to Piwi proteins.


Assuntos
RNA Helicases/genética , RNA Helicases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Animais , Proteínas Argonautas , Sequência de Bases , Proteínas de Ciclo Celular , Metilação de DNA , Primers do DNA/genética , Fertilidade , Masculino , Meiose , Camundongos , Camundongos Knockout , Mutação , Proteínas/metabolismo , RNA Helicases/deficiência , Retroelementos/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Espermatócitos/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Testículo/metabolismo
5.
J Biol Chem ; 285(53): 41337-47, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21047779

RESUMO

Glycogen synthase kinase-3 (Gsk-3) isoforms, Gsk-3α and Gsk-3ß, are constitutively active, largely inhibitory kinases involved in signal transduction. Underscoring their biological significance, altered Gsk-3 activity has been implicated in diabetes, Alzheimer disease, schizophrenia, and bipolar disorder. Here, we demonstrate that deletion of both Gsk-3α and Gsk-3ß in mouse embryonic stem cells results in reduced expression of the de novo DNA methyltransferase Dnmt3a2, causing misexpression of the imprinted genes Igf2, H19, and Igf2r and hypomethylation of their corresponding imprinted control regions. Treatment of wild-type embryonic stem cells and neural stem cells with the Gsk-3 inhibitor, lithium, phenocopies the DNA hypomethylation at these imprinted loci. We show that inhibition of Gsk-3 by phosphatidylinositol 3-kinase (PI3K)-mediated activation of Akt also results in reduced DNA methylation at these imprinted loci. Finally, we find that N-Myc is a potent Gsk-3-dependent regulator of Dnmt3a2 expression. In summary, we have identified a signal transduction pathway that is capable of altering the DNA methylation of imprinted loci.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Regulação Enzimológica da Expressão Gênica , Impressão Genômica , Quinase 3 da Glicogênio Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais
6.
Mol Reprod Dev ; 78(4): 241-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21308854

RESUMO

In eukaryotes, mRNA is actively exported to the cytoplasm by a family of nuclear RNA export factors (NXF). Four Nxf genes have been identified in the mouse: Nxf1, Nxf2, Nxf3, and Nxf7. Inactivation of Nxf2, a germ cell-specific gene, causes defects in spermatogenesis. Here we report that Nxf3 is expressed exclusively in Sertoli cells of the postnatal testis, in a developmentally regulated manner. Expression of Nxf3 coincides with the cessation of Sertoli cell proliferation and the beginning of their differentiation. Continued expression of Nxf3 in mature Sertoli cells of the adult is spermatogenesis stage-independent. Nxf3 is not essential for spermatogenesis, however, suggesting functional redundancy among Nxf family members. With its unique expression pattern in the testis, the promoter of Nxf3 can be used to drive postnatal Sertoli cell-specific expression of other proteins such as Cre recombinase.


Assuntos
Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Feminino , Integrases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Transporte de RNA , RNA Mensageiro/metabolismo , Células de Sertoli/citologia , Testículo/citologia , Testículo/metabolismo
7.
J Cell Biol ; 173(4): 497-507, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16717126

RESUMO

During meiosis, the arrangement of homologous chromosomes is tightly regulated by the synaptonemal complex (SC). Each SC consists of two axial/lateral elements (AEs/LEs), and numerous transverse filaments. SC protein 2 (SYCP2) and SYCP3 are integral components of AEs/LEs in mammals. We find that SYCP2 forms heterodimers with SYCP3 both in vitro and in vivo. An evolutionarily conserved coiled coil domain in SYCP2 is required for binding to SYCP3. We generated a mutant Sycp2 allele in mice that lacks the coiled coil domain. The fertility of homozygous Sycp2 mutant mice is sexually dimorphic; males are sterile because of a block in meiosis, whereas females are subfertile with sharply reduced litter size. Sycp2 mutant spermatocytes exhibit failure in the formation of AEs and chromosomal synapsis. Strikingly, the mutant SYCP2 protein localizes to axial chromosomal cores in both spermatocytes and fetal oocytes, but SYCP3 does not, demonstrating that SYCP2 is a primary determinant of AEs/LEs and, thus, is required for the incorporation of SYCP3 into SCs.


Assuntos
Proteínas Cromossômicas não Histona/genética , Pareamento Cromossômico/genética , Cromossomos/genética , Meiose/genética , Espermatócitos/metabolismo , Complexo Sinaptonêmico/metabolismo , Animais , Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/metabolismo , Sequência Conservada/genética , Proteínas de Ligação a DNA , Feminino , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Oócitos/ultraestrutura , Estrutura Terciária de Proteína/genética , Caracteres Sexuais , Diferenciação Sexual/genética , Espermatócitos/ultraestrutura , Espermatogênese/genética , Complexo Sinaptonêmico/ultraestrutura
8.
Front Genet ; 12: 627050, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33889176

RESUMO

Gene editing of the mitochondrial genome using the CRISPR-Cas9 system is highly challenging mainly due to sub-efficient delivery of guide RNA and Cas9 enzyme complexes into the mitochondria. In this study, we were able to perform gene editing in the mitochondrial DNA by appending an NADH-ubiquinone oxidoreductase chain 4 (ND4) targeting guide RNA to an RNA transport-derived stem loop element (RP-loop) and expressing the Cas9 enzyme with a preceding mitochondrial localization sequence. We observe mitochondrial colocalization of RP-loop gRNA and a marked reduction of ND4 expression in the cells carrying a 11205G variant in their ND4 sequence coincidently decreasing the mtDNA levels. This proof-of-concept study suggests that a stem-loop element added sgRNA can be transported to the mitochondria and functionally interact with Cas9 to mediate sequence-specific mtDNA cleavage. Using this novel approach to target the mtDNA, our results provide further evidence that CRISPR-Cas9-mediated gene editing might potentially be used to treat mitochondrial-related diseases.

9.
Dev Biol ; 330(1): 167-74, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19345203

RESUMO

In eukaryotes, mRNA is actively transported from nucleus to cytoplasm by a family of nuclear RNA export factors (NXF). While yeast harbors only one such factor (Mex67p), higher eukaryotes encode multiple NXFs. In mouse, four Nxf genes have been identified: Nxf1, Nxf2, Nxf3, and Nxf7. To date, the function of mouse Nxf genes has not been studied by targeted gene deletion in vivo. Here we report the generation of Nxf2 null mutant mice by homologous recombination in embryonic stem cells. Nxf2-deficient male mice exhibit fertility defects that differ between mouse strains. One third of Nxf2-deficient males on a mixed (C57BL/6x129) genetic background exhibit meiotic arrest and thus are sterile, whereas the remaining males are fertile. Disruption of Nxf2 in inbred (C57BL/6J) males impairs spermatogenesis, resulting in male subfertility, but causes no meiotic arrest. Testis weight and sperm output in C57BL/6J Nxf2(-/Y) mice are sharply reduced. Mutant epididymal sperm exhibit diminished motility. Importantly, proliferation of spermatogonia in Nxf2(-/Y) mice is significantly decreased. As a result, inactivation of Nxf2 causes depletion of germ cells in a substantial fraction of seminiferous tubules in aged mice. These studies demonstrate that Nxf2 plays a dual function in spermatogenesis: regulation of meiosis and maintenance of spermatogonial stem cells.


Assuntos
Meiose , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Ligação a RNA/genética , Espermatogônias/metabolismo , Fatores Etários , Animais , Proliferação de Células , Citoplasma/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Transporte de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Motilidade dos Espermatozoides/genética , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/crescimento & desenvolvimento
10.
Mol Cell Biol ; 27(7): 2758-64, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17242180

RESUMO

macroH2A histone variants have been implicated to function in gene silencing by several studies, including ones showing a preferential association of macroH2A on the inactive X chromosome. To examine macroH2A function in vivo, we knocked out macroH2A1. macroH2A1 knockout mice are viable and fertile. A broad screen of liver gene expression showed no evidence of defects in X inactivation but did identify genes that have increased expression levels in macroH2A1 knockouts. macroH2A1-containing nucleosomes are enriched on the coding and/or upstream regions of these genes, suggesting that their increased expression levels are a direct effect of the absence of macroH2A1. The concentrations of macroH2A1 nucleosomes on these genes are low in the livers of newborn mice, and the macroH2A1 knockout had little effect on the expression levels of these genes in newborn liver. Our results indicate that an increase in liver macroH2A1 during the transition from newborn to young-adult status contributes to a decrease in the expression levels of these genes. These genes cluster in the area of lipid metabolism, and we observed metabolic effects in macroH2A1 knockouts. Our results indicate that the function of macroH2A1 histones is not restricted to gene silencing but also involves fine tuning the expression of specific genes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histonas/biossíntese , Animais , Animais Recém-Nascidos , Perfilação da Expressão Gênica , Inativação Gênica , Glucose/metabolismo , Histonas/genética , Fígado/metabolismo , Camundongos , Camundongos Knockout , Mutação , Nucleossomos/genética , Nucleossomos/metabolismo , Cromossomo X/genética , Cromossomo X/metabolismo , Inativação do Cromossomo X/genética , Inativação do Cromossomo X/fisiologia
11.
Stem Cells ; 26(6): 1474-83, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18369101

RESUMO

Uniparental zygotes with two paternal (androgenetic [AG]) or two maternal (gynogenetic [GG]; parthenogenetic [PG]) genomes are not able to develop into viable offspring but can form blastocysts from which embryonic stem cells (ESCs) can be derived. Although some aspects of the in vitro and in vivo differentiation potential of PG and GG ESCs of several species have been studied, the developmental capacity of AG ESCs is much less clear. Here, we investigate the potential of murine AG ESCs to undergo neural differentiation. We observed that AG ESCs differentiate in vitro into pan-neural progenitor cells (pnPCs) that further give rise to cells that express neuronal- and astroglial-specific markers. Neural progeny of in vitro-differentiated AG ESCs exhibited fidelity of expression of six imprinted genes analyzed, with the exception of Ube3a. Bisulfite sequencing for two imprinting control regions suggested that pnPCs predominantly maintained their methylation pattern. Following blastocyst injection of AG and biparental (normal fertilized [N]) ESCs, we found widespread and evenly distributed contribution of ESC-derived cells in both AG and N chimeric early fetal brains. AG and N ESC-derived cells isolated from chimeric fetal brains by fluorescence-activated cell sorting exhibited similar neurosphere-initiating cell frequencies and neural multilineage differentiation potential. Our results indicate that AG ESC-derived neural progenitor/stem cells do not differ from N neural progenitor/stem cells in their self-renewal and neural multilineage differentiation potential. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Androgênios/fisiologia , Blastocisto/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Animais , Blastocisto/citologia , Encéfalo/citologia , Encéfalo/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular , Divisão Celular , Feminino , Genes Reporter , Genoma , Impressão Genômica , Masculino , Camundongos , Camundongos Endogâmicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/genética , Zigoto
12.
Methods Mol Biol ; 430: 195-211, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370301

RESUMO

Complementing mutant embryos or embryonic stem cells with normal cells in embryonic chimeras is a valuable tool for investigating phenotypes. Chimera approaches provide a method to examine the phenotype of mutant cells, including hematopoiesis, in mutants with early embryonic lethality. Complementation with normal cells in a chimera can, in most instances, rescue mutant cells to later stages of gestation and beyond, permitting analysis of contribution and function of mutant cells in various organs, both within the chimera, but also by using functional transplantation assays for hematopoietic stem and progenitor cells. This chapter describes principles and methods for the generation of mouse chimeras, for identification and quantitative analysis of cell contribution in chimeras, and for chimeric fetal liver transplantation into adult recipients and analysis of mutant cells in the adult.


Assuntos
Quimera , Hematopoese , Transplante de Fígado , Fígado/embriologia , Animais , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL
13.
J Vis Exp ; (136)2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29985369

RESUMO

CRISPR/Cas9 technology is accelerating genome engineering in many cell types, but so far, gene delivery and stable gene modification have been challenging in primary NK cells. For example, transgene delivery using lentiviral or retroviral transduction resulted in a limited yield of genetically-engineered NK cells due to substantial procedure-associated NK cell apoptosis. We describe here a DNA-free method for genome editing of human primary and expanded NK cells using Cas9 ribonucleoprotein complexes (Cas9/RNPs). This method allowed efficient knockout of the TGFBR2 and HPRT1 genes in NK cells. RT-PCR data showed a significant decrease in gene expression level, and a cytotoxicity assay of a representative cell product suggested that the RNP-modified NK cells became less sensitive to TGFß. Genetically modified cells could be expanded post-electroporation by stimulation with irradiated mbIL21-expressing feeder cells.


Assuntos
Sistemas CRISPR-Cas/genética , Engenharia Genética/métodos , Terapia Genética/métodos , Imunoterapia/métodos , Células Matadoras Naturais/metabolismo , Ribonucleoproteínas/metabolismo , Humanos
14.
JCI Insight ; 3(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30429376

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant or digenic disorder linked to derepression of the toxic DUX4 gene in muscle. There is currently no pharmacological treatment. The emergence of DUX4 enabled development of cell and animal models that could be used for basic and translational research. Since DUX4 is toxic, animal model development has been challenging, but progress has been made, revealing that tight regulation of DUX4 expression is critical for creating viable animals that develop myopathy. Here, we report such a model - the tamoxifen-inducible FSHD mouse model called TIC-DUX4. Uninduced animals are viable, born in Mendelian ratios, and overtly indistinguishable from WT animals. Induced animals display significant DUX4-dependent myopathic phenotypes at the molecular, histological, and functional levels. To demonstrate the utility of TIC-DUX4 mice for therapeutic development, we tested a gene therapy approach aimed at improving muscle strength in DUX4-expressing muscles using adeno-associated virus serotype 1.Follistatin (AAV1.Follistatin), a natural myostatin antagonist. This strategy was not designed to modulate DUX4 but could offer a mechanism to improve muscle weakness caused by DUX4-induced damage. AAV1.Follistatin significantly increased TIC-DUX4 muscle mass and strength even in the presence of DUX4 expression, suggesting that myostatin inhibition may be a promising approach to treat FSHD-associated weakness. We conclude that TIC-DUX4 mice are a relevant model to study DUX4 toxicity and, importantly, are useful in therapeutic development studies for FSHD.


Assuntos
Modelos Animais de Doenças , Folistatina/genética , Terapia Genética , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/terapia , Miostatina/antagonistas & inibidores , Animais , Feminino , Folistatina/uso terapêutico , Masculino , Camundongos Transgênicos , Distrofia Muscular Facioescapuloumeral/induzido quimicamente , Distrofia Muscular Facioescapuloumeral/genética , Fenótipo , Tamoxifeno
15.
Cloning Stem Cells ; 9(4): 630-41, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18154522

RESUMO

The inefficiency of mammalian somatic cell cloning is associated with abnormal gene expression presumably caused by errors in reprogramming of the transplanted genome. In the mouse, aggregation of four-cell stage clones leads to an improvement of both gene expression and development. To determine whether clone-clone aggregation at postgenomic activation stages influences gene expression in bovine clones, we profiled, in single and aggregated embryos at the blastocyst stage, expression of developmentally relevant genes namely Oct4, Dnmt1, Dnmt3, Glut1, Glut3, and a housekeeping gene, Poly(A) polymerase (PolyA) by real-time RT-PCR. Compared to embryos generated by in vitro fertilization (IVF), individual clones more frequently exhibited transcript levels that were more than twofold higher or lower than the average value of IVF embryos. This was observed less often in clone aggregates for Oct4, Dnmt1, Dnmt3, and PolyA, but not for Glut1 and Glut3. The analysis of interferon tau bioactivity as a marker of trophectoderm function in blastocyst outgrowths showed that both single clones and clone aggregates have less extraembryonic potential in vitro compared to IVF embryos, with no apparent consequence of aggregation. These findings indicate that aggregation of bovine clones with each other at later cleavage stages can change gene expression patterns at preimplantation stages, but does not rescue trophectoderm function in vitro.


Assuntos
Blastocisto/citologia , Clonagem de Organismos/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas tau/metabolismo , Animais , Bovinos , Linhagem Celular , Núcleo Celular/metabolismo , Primers do DNA/química , Ectoderma/metabolismo , Implantação do Embrião , Fertilização in vitro , Oócitos/metabolismo , RNA Mensageiro/metabolismo
16.
PLoS One ; 11(12): e0166822, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27926922

RESUMO

Transferring mouse mutations into specific mouse strain backgrounds can be critical for appropriate analysis of phenotypic effects of targeted genomic alterations and quantitative trait loci. Speed congenic breeding strategies incorporating marker-assisted selection of progeny with the highest percentage target background as breeders for the next generation can produce congenic strains within approximately 5 generations. When mating selected donor males to target strain females, this may require more than 1 year, with each generation lasting 10 to 11 weeks including 3 weeks of gestation and 7 to 8 weeks until the males reach sexual maturity. Because ovulation can be induced in female mice as early as 3 weeks of age, superovulation-aided backcrossing of marker-selected females could accelerate the production of congenic animals by approximately 4 weeks per generation, reducing time and cost. Using this approach, we transferred a transgenic strain of undefined genetic background to >99% C57BL/6J within 10 months, with most generations lasting 7 weeks. This involved less than 60 mice in total, with 9 to 18 animals per generation. Our data demonstrate that high-speed backcrossing through the female germline is feasible and practical with small mouse numbers.


Assuntos
Células Germinativas/fisiologia , Reprodução/fisiologia , Animais , Animais Congênicos/fisiologia , Feminino , Endogamia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Locos de Características Quantitativas/fisiologia
17.
Anim Reprod Sci ; 82-83: 97-108, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15271446

RESUMO

In the context of mammalian somatic cell cloning, the term reprogramming refers to the processes that enable a somatic cell nucleus to adopt the role of a zygotic nucleus. Gene re-expression is one measure of reprogramming if correlated with subsequent developmental potential. This paper describes several experiments utilizing pre-implantation gene expression to evaluate reprogramming and clone viability. We have established a direct correlation between Oct4 expression in mouse clones at the blastocyst stage and their potential to maintain pluripotent embryonic cells essential for post-implantation development. Furthermore, the quality of gene expression in clones dramatically improves when genetically identical clones are combined in clone-clone aggregate chimeras. Clone--clone aggregates exhibit a higher developmental potential than single clones both in vitro and in vivo. This could be mediated by complementation between blastomeres from epigenetically different clones within the aggregate rather than by the increase in cell number resulting from aggregation. We also discuss the use of tetraploid embryos as a model to evaluate reprogramming using gene expression and demonstrate that somatic cell nuclei can be reprogrammed by blastomeres to re-express embryonic specific genes but not to contribute to post-implantation development.


Assuntos
Núcleo Celular/genética , Clonagem de Organismos , Expressão Gênica , Técnicas de Transferência Nuclear , Zigoto/ultraestrutura , Animais , Blastocisto/metabolismo , Fase de Clivagem do Zigoto , Clonagem de Organismos/métodos , Clonagem de Organismos/veterinária , Proteínas de Ligação a DNA/genética , Camundongos , Fator 3 de Transcrição de Octâmero , Fatores de Transcrição/genética
18.
PLoS One ; 9(2): e89471, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586802

RESUMO

Tudor domain containing (Tdrd) proteins that are expressed in germ cells are divided into two groups. One group, consisting of TDRD1, TDRKH, TDRD9 and TDRD12, function in piRNA biogenesis and retrotransposon silencing, while the other group including RNF17/TDRD4 and TDRD5-7 are required for spermiogenesis. These Tdrd proteins play distinct roles during male germ cell development. Here, we report the characterization of STK31/TDRD8 in mice. STK31 contains a tudor domain and a serine/threonine kinase domain. We find that STK31 is a cytoplasmic protein in germ cells. STK31 is expressed in embryonic gonocytes of both sexes and postnatal spermatocytes and round spermatids in males. Disruption of the tudor domain and kinase domain of STK31 respectively does not affect fertility in mice. Our data suggest that the function of STK31 may be redundant with other Tdrd proteins in germ cell development.


Assuntos
Células Germinativas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Reprodução/genética , Animais , Western Blotting , Citoplasma/metabolismo , Primers do DNA/genética , Imunofluorescência , Perfilação da Expressão Gênica , Técnicas Histológicas , Masculino , Camundongos , Estrutura Terciária de Proteína/genética , Reprodução/fisiologia
19.
Cell Med ; 5(1): 29-42, 2013 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26858862

RESUMO

Uniparental zygotes with two paternal (androgenetic, AG) or two maternal genomes (gynogenetic, GG) cannot develop into viable offsprings but form blastocysts from which pluripotent embryonic stem (ES) cells can be derived. For most organs, it is unclear whether uniparental ES cells can give rise to stably expandable somatic stem cells that can repair injured tissues. Even if previous reports indicated that the capacity of AG ES cells to differentiate in vitro into pan-neural progenitor cells (pNPCs) and into cells expressing neural markers is similar to biparental [normal fertilized (N)] ES cells, their potential for functional neurogenesis is not known. Here we show that murine AG pNPCs give rise to neuron-like cells, which then generate sodium-driven action potentials while maintaining fidelity of imprinted gene expression. Neural engraftment after intracerebral transplantation was achieved only by late (22 days) AG and N pNPCs with in vitro low colony-forming cell (CFC) capacity. However, persisting CFC formation seen, in particular, in early (13 or 16 days) differentiation cultures of N and AG pNPCs correlated with a high incidence of trigerm layer teratomas. As AG ES cells display functional neurogenesis and in vivo stability similar to N ES cells, they represent a unique model system to study the roles of paternal and maternal genomes on neural development and on the development of imprinting-associated brain diseases.

20.
Cell Cycle ; 10(13): 2091-9, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21606677

RESUMO

Chimeras are organisms composed of at least two genetically distinct cell lineages originating from different zygotes. In the laboratory, mouse chimeras can be produced experimentally; various techniques allow combining different early stage mouse embryos with each other or with pluripotent stem cells. Identification of the progeny of the different lineages in chimeras permits to follow cell fate and function, enabling correlation of genotype with phenotype. Mouse chimeras have become a tool to investigate critical developmental processes, including cell specification, differentiation, patterning, and the function of specific genes. In addition, chimeras can also be generated to address biological processes in the adult, including mechanisms underlying diseases or tissue repair and regeneration. This review summarizes the different types of chimeras and how they have been generated and provides examples of how mouse chimeras offer a unique and powerful system to investigate questions pertaining to cell and tissue function in the developing and adult organism.


Assuntos
Quimera , Doença , Embrião de Mamíferos/fisiologia , Regeneração/fisiologia , Animais , Embrião de Mamíferos/anatomia & histologia , Humanos , Camundongos , Transplante de Órgãos/métodos , Células-Tronco/citologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA