Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Genes Dev ; 37(15-16): 724-742, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37612136

RESUMO

Histidine (His) residues are methylated in various proteins, but their roles and regulation mechanisms remain unknown. Here, we show that carnosine N-methyltransferase 1 (CARNMT1), a known His methyltransferase of dipeptide carnosine (ßAla-His), is a major His N1-position-specific methyltransferase. We found that 52 His sites in 20 proteins underwent CARNMT1-mediated methylation. The consensus methylation site for CARNMT1 was identified as Cx(F/Y)xH, a C3H zinc finger (C3H ZF) motif. CARNMT1-deficient and catalytically inactive mutant mice showed embryonic lethality. Among the CARNMT1 target C3H ZF proteins, RNA degradation mediated by Roquin and tristetraprolin (TTP) was affected by CARNMT1 and its enzymatic activity. Furthermore, the recognition of the 3' splice site of the CARNMT1 target C3H ZF protein U2AF1 was perturbed, and pre-mRNA alternative splicing (AS) was affected by CARNMT1 deficiency. These findings indicate that CARNMT1-mediated protein His methylation, which is essential for embryogenesis, plays roles in diverse aspects of RNA metabolism by targeting C3H ZF-type RNA-binding proteins and modulating their functions, including pre-mRNA AS and mRNA degradation regulation.


Assuntos
Carnosina , Animais , Camundongos , Camundongos Endogâmicos C3H , Histidina/genética , Precursores de RNA , Metiltransferases/genética , Sítios de Splice de RNA , Dedos de Zinco
2.
Genes Dev ; 36(7-8): 483-494, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35483741

RESUMO

Genomic imprinting regulates parental origin-dependent monoallelic gene expression. It is mediated by either germline differential methylation of DNA (canonical imprinting) or oocyte-derived H3K27me3 (noncanonical imprinting) in mice. Depletion of Eed, an essential component of Polycomb repressive complex 2, results in genome-wide loss of H3K27me3 in oocytes, which causes loss of noncanonical imprinting (LOI) in embryos. Although Eed maternal KO (matKO) embryos show partial lethality after implantation, it is unknown whether LOI itself contributes to the developmental phenotypes of these embryos, which makes it unclear whether noncanonical imprinting is developmentally relevant. Here, by combinatorial matKO of Xist, a noncanonical imprinted gene whose LOI causes aberrant transient maternal X-chromosome inactivation (XCI) at preimplantation, we show that prevention of the transient maternal XCI greatly restores the development of Eed matKO embryos. Moreover, we found that the placentae of Eed matKO embryos are remarkably enlarged in a manner independent of Xist LOI. Heterozygous deletion screening of individual autosomal noncanonical imprinted genes suggests that LOI of the Sfmbt2 miRNA cluster chromosome 2 miRNA cluster (C2MC), solute carrier family 38 member 4 (Slc38a4), and Gm32885 contributes to the placental enlargement. Taken together, our study provides evidence that Xist imprinting sustains embryonic development and that autosomal noncanonical imprinting restrains placental overgrowth.


Assuntos
MicroRNAs , RNA Longo não Codificante , Animais , Desenvolvimento Embrionário/genética , Feminino , Histonas/metabolismo , Camundongos , Placenta , Gravidez , RNA Longo não Codificante/genética , Proteínas Repressoras/genética , Inativação do Cromossomo X
3.
Genes Dev ; 36(1-2): 84-102, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34992147

RESUMO

The placenta is a highly evolved, specialized organ in mammals. It differs from other organs in that it functions only for fetal maintenance during gestation. Therefore, there must be intrinsic mechanisms that guarantee its unique functions. To address this question, we comprehensively analyzed epigenomic features of mouse trophoblast stem cells (TSCs). Our genome-wide, high-throughput analyses revealed that the TSC genome contains large-scale (>1-Mb) rigid heterochromatin architectures with a high degree of histone H3.1/3.2-H3K9me3 accumulation, which we termed TSC-defined highly heterochromatinized domains (THDs). Importantly, depletion of THDs by knockdown of CAF1, an H3.1/3.2 chaperone, resulted in down-regulation of TSC markers, such as Cdx2 and Elf5, and up-regulation of the pluripotent marker Oct3/4, indicating that THDs maintain the trophoblastic nature of TSCs. Furthermore, our nuclear transfer technique revealed that THDs are highly resistant to genomic reprogramming. However, when H3K9me3 was removed, the TSC genome was fully reprogrammed, giving rise to the first TSC cloned offspring. Interestingly, THD-like domains are also present in mouse and human placental cells in vivo, but not in other cell types. Thus, THDs are genomic architectures uniquely developed in placental lineage cells, which serve to protect them from fate reprogramming to stably maintain placental function.


Assuntos
Histonas , Trofoblastos , Animais , Diferenciação Celular/genética , Feminino , Histonas/genética , Histonas/metabolismo , Mamíferos , Camundongos , Placenta , Gravidez , Células-Tronco , Trofoblastos/metabolismo
4.
Cell ; 159(4): 884-95, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25417163

RESUMO

Mammalian oocytes can reprogram somatic cells into a totipotent state enabling animal cloning through somatic cell nuclear transfer (SCNT). However, the majority of SCNT embryos fail to develop to term due to undefined reprogramming defects. Here, we identify histone H3 lysine 9 trimethylation (H3K9me3) of donor cell genome as a major barrier for efficient reprogramming by SCNT. Comparative transcriptome analysis identified reprogramming resistant regions (RRRs) that are expressed normally at 2-cell mouse embryos generated by in vitro fertilization (IVF) but not SCNT. RRRs are enriched for H3K9me3 in donor somatic cells and its removal by ectopically expressed H3K9me3 demethylase Kdm4d not only reactivates the majority of RRRs, but also greatly improves SCNT efficiency. Furthermore, use of donor somatic nuclei depleted of H3K9 methyltransferases markedly improves SCNT efficiency. Our study thus identifies H3K9me3 as a critical epigenetic barrier in SCNT-mediated reprogramming and provides a promising approach for improving mammalian cloning efficiency.


Assuntos
Desenvolvimento Embrionário , Código das Histonas , Histonas/metabolismo , Técnicas de Transferência Nuclear , Animais , Clonagem de Organismos/métodos , Embrião de Mamíferos/metabolismo , Feminino , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Metilação , Metiltransferases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas Repressoras/metabolismo , Zigoto
5.
Cell ; 154(6): 1380-9, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23992846

RESUMO

Targeted genome editing technologies have enabled a broad range of research and medical applications. The Cas9 nuclease from the microbial CRISPR-Cas system is targeted to specific genomic loci by a 20 nt guide sequence, which can tolerate certain mismatches to the DNA target and thereby promote undesired off-target mutagenesis. Here, we describe an approach that combines a Cas9 nickase mutant with paired guide RNAs to introduce targeted double-strand breaks. Because individual nicks in the genome are repaired with high fidelity, simultaneous nicking via appropriately offset guide RNAs is required for double-stranded breaks and extends the number of specifically recognized bases for target cleavage. We demonstrate that using paired nicking can reduce off-target activity by 50- to 1,500-fold in cell lines and to facilitate gene knockout in mouse zygotes without sacrificing on-target cleavage efficiency. This versatile strategy enables a wide variety of genome editing applications that require high specificity.


Assuntos
Quebras de DNA de Cadeia Dupla , Marcação de Genes/métodos , Genoma , Animais , Sequência de Bases , Camundongos , Dados de Sequência Molecular , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/genética , Zigoto/metabolismo , Pequeno RNA não Traduzido
6.
Development ; 150(20)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36897562

RESUMO

Reactive oxygen species (ROS) are generated from NADPH oxidases and mitochondria; they are generally harmful for stem cells. Spermatogonial stem cells (SSCs) are unique among tissue-stem cells because they undergo ROS-dependent self-renewal via NOX1 activation. However, the mechanism by which SSCs are protected from ROS remains unknown. Here, we demonstrate a crucial role for Gln in ROS protection using cultured SSCs derived from immature testes. Measurements of amino acids required for SSC cultures revealed the indispensable role of Gln in SSC survival. Gln induced Myc expression to drive SSC self-renewal in vitro, whereas Gln deprivation triggered Trp53-dependent apoptosis and impaired SSC activity. However, apoptosis was attenuated in cultured SSCs that lacked NOX1. In contrast, cultured SSCs lacking Top1mt mitochondria-specific topoisomerase exhibited poor mitochondrial ROS production and underwent apoptosis. Gln deprivation reduced glutathione production; supra-molar Asn supplementation allowed offspring production from SSCs cultured without Gln. Therefore, Gln ensures ROS-dependent SSC-self-renewal by providing protection against NOX1 and inducing Myc.


Assuntos
Glutamina , Espermatogônias , Masculino , Camundongos , Animais , Espermatogônias/metabolismo , Glutamina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proliferação de Células , Células-Tronco , Células Cultivadas
7.
Development ; 148(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33766931

RESUMO

During spermatogenesis, intricate gene expression is coordinately regulated by epigenetic modifiers, which are required for differentiation of spermatogonial stem cells (SSCs) contained among undifferentiated spermatogonia. We have previously found that KMT2B conveys H3K4me3 at bivalent and monovalent promoters in undifferentiated spermatogonia. Because these genes are expressed late in spermatogenesis or during embryogenesis, we expect that many of them are potentially programmed by KMT2B for future expression. Here, we show that one of the genes targeted by KMT2B, Tsga8, plays an essential role in spermatid morphogenesis. Loss of Tsga8 in mice leads to male infertility associated with abnormal chromosomal distribution in round spermatids, malformation of elongating spermatid heads and spermiation failure. Tsga8 depletion leads to dysregulation of thousands of genes, including the X-chromosome genes that are reactivated in spermatids, and insufficient nuclear condensation accompanied by reductions of TNP1 and PRM1, key factors for histone-to-protamine transition. Intracytoplasmic sperm injection (ICSI) of spermatids rescued the infertility phenotype, suggesting competency of the spermatid genome for fertilization. Thus, Tsga8 is a KMT2B target that is vitally necessary for spermiogenesis and fertility.


Assuntos
Fertilidade , Nucleoproteínas/metabolismo , Espermátides/metabolismo , Espermatogênese , Células-Tronco/metabolismo , Animais , Feminino , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Nucleoproteínas/genética , Espermatogônias/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(14): 7837-7844, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32229564

RESUMO

The blood-testis barrier (BTB) is thought to be indispensable for spermatogenesis because it creates a special environment for meiosis and protects haploid cells from the immune system. The BTB divides the seminiferous tubules into the adluminal and basal compartments. Spermatogonial stem cells (SSCs) have a unique ability to transmigrate from the adluminal compartment to the basal compartment through the BTB upon transplantation into the seminiferous tubule. Here, we analyzed the role of Cldn11, a major component of the BTB, in spermatogenesis using spermatogonial transplantation. Cldn11-deficient mice are infertile due to the cessation of spermatogenesis at the spermatocyte stage. Cldn11-deficient SSCs failed to colonize wild-type testes efficiently, and Cldn11-deficient SSCs that underwent double depletion of Cldn3 and Cldn5 showed minimal colonization, suggesting that claudins on SSCs are necessary for transmigration. However, Cldn11-deficient Sertoli cells increased SSC homing efficiency by >3-fold, suggesting that CLDN11 in Sertoli cells inhibits transmigration of SSCs through the BTB. In contrast to endogenous SSCs in intact Cldn11-deficient testes, those from WT or Cldn11-deficient testes regenerated sperm in Cldn11-deficient testes. The success of this autologous transplantation appears to depend on removal of endogenous germ cells for recipient preparation, which reprogrammed claudin expression patterns in Sertoli cells. Consistent with this idea, in vivo depletion of Cldn3/5 regenerated endogenous spermatogenesis in Cldn11-deficient mice. Thus, coordinated claudin expression in both SSCs and Sertoli cells expression is necessary for SSC homing and regeneration of spermatogenesis, and autologous stem cell transplantation can rescue congenital defects of a self-renewing tissue.


Assuntos
Fertilidade/genética , Infertilidade/terapia , Espermatogônias/transplante , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Fertilidade/fisiologia , Humanos , Infertilidade/genética , Infertilidade/patologia , Masculino , Camundongos , Espermatogênese/genética , Espermatogônias/crescimento & desenvolvimento , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/transplante , Células-Tronco/citologia , Transplante Autólogo/métodos
9.
Proc Natl Acad Sci U S A ; 117(5): 2513-2518, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964830

RESUMO

During natural fertilization, mammalian spermatozoa must pass through the zona pellucida before reaching the plasma membrane of the oocyte. It is assumed that this step involves partial lysis of the zona by sperm acrosomal enzymes, but there has been no unequivocal evidence to support this view. Here we present evidence that acrosin, an acrosomal serine protease, plays an essential role in sperm penetration of the zona. We generated acrosin-knockout (KO) hamsters, using an in vivo transfection CRISPR/Cas9 system. Homozygous mutant males were completely sterile. Acrosin-KO spermatozoa ascended the female genital tract and reached ovulated oocytes in the oviduct ampulla, but never fertilized them. In vitro fertilization (IVF) experiments revealed that mutant spermatozoa attached to the zona, but failed to penetrate it. When the zona pellucida was removed before IVF, all oocytes were fertilized. This indicates that in hamsters, acrosin plays an indispensable role in allowing fertilizing spermatozoa to penetrate the zona. This study also suggests that the KO hamster system would be a useful model for identifying new gene functions or analyzing human and animal disorders because of its technical facility and reproducibility.


Assuntos
Acrosina/metabolismo , Cricetinae/metabolismo , Interações Espermatozoide-Óvulo , Espermatozoides/enzimologia , Acrosina/genética , Acrossomo/metabolismo , Animais , Cricetinae/genética , Feminino , Fertilização in vitro , Técnicas de Inativação de Genes , Masculino , Espermatozoides/fisiologia , Zona Pelúcida/metabolismo
10.
Methods ; 191: 23-31, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32334080

RESUMO

Genetically modified mouse models are essential for in vivo investigation of gene function and human disease research. Targeted mutations can be introduced into mouse embryos using genome editing technology such as CRISPR-Cas. Although mice with small indel mutations can be produced, the production of mice carrying large deletions or gene fragment knock-in alleles remains inefficient. We introduced the nuclear localisation property of Cdt1 protein into the CRISPR-Cas system for efficient production of genetically engineered mice. Mouse Cdt1-connected Cas9 (Cas9-mC) was present in the nucleus of HEK293T cells and mouse embryos. Cas9-mC induced a bi-allelic full deletion of Dmd, GC-rich fragment knock-in, and floxed allele knock-in with high efficiency compared to standard Cas9. These results indicate that Cas9-mC is a useful tool for producing mouse models carrying targeted mutations.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Animais , Sistemas CRISPR-Cas/genética , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Camundongos , Zigoto
11.
Proc Natl Acad Sci U S A ; 116(42): 21047-21053, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31570606

RESUMO

The placenta is critical in mammalian embryonic development because the embryo's supply of nutrients, including amino acids, depends solely on mother-to-embryo transport through it. However, the molecular mechanisms underlying this amino acid supply are poorly understood. In this study, we focused on system A amino acid transporters Slc38a1/SNAT1, Slc38a2/SNAT2, and Slc38a4/SNAT4, which carry neutral, short-side-chain amino acids, to determine their involvement in placental or embryonic development. A triple-target CRISPR screen identified Slc38a4/SNAT4 as the critical amino acid transporter for placental development in mice. We established mouse lines from the CRISPR founders with large deletions in Slc38a4 and found that, consistent with the imprinted paternal expression of Slc38a4/SNAT4 in the placenta, paternal knockout (KO) but not maternal KO of Slc38a4/SNAT4 caused placental hypoplasia associated with reduced fetal weight. Immunostaining revealed that SNAT4 was widely expressed in differentiating cytotrophoblasts and maturing trophoblasts at the maternal-fetal interface. A blood metabolome analysis revealed that amino acid concentrations were globally reduced in Slc38a4/SNAT4 mutant embryos. These results indicated that SNAT4-mediated amino acid transport in mice plays a major role in placental and embryonic development. Given that expression of Slc38a4 in the placenta is conserved in other species, our Slc38a4/SNAT4 mutant mice could be a promising model for the analysis of placental defects leading to intrauterine growth restriction in mammals.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Placenta/metabolismo , Placenta/patologia , Útero/metabolismo , Útero/patologia , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Animais , Feminino , Camundongos , Placentação/fisiologia , Gravidez , Trofoblastos/metabolismo , Trofoblastos/patologia
12.
Biol Reprod ; 104(1): 223-233, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-32965494

RESUMO

Conditional knockout (cKO) mice have contributed greatly to understanding the tissue- or stage-specific functions of genes in vivo. However, the current cKO method requires considerable time and effort because of the need to generate two gene-modified mouse strains (Cre transgenic and loxP knockin) for crossing. Here, we examined whether we could analyze the germ cell-related functions of embryonic lethal genes in F0 chimeric mice by restricting the origin of germ cells to mutant embryonic stem cells (ESCs). We confirmed that the full ESC origin of spermatozoa in fertile chimeric mice was achieved by the CRISPR/Cas9 system using three guide RNAs targeting Nanos3, which induced germ cell depletion in the host blastocyst-derived tissues. Among these fertile chimeric mice, those from male ESCs with a Dnmt3b mutation, which normally causes embryo death, also produced F1 mice derived exclusively from the mutant ESCs. Thus, our new chimeric strategy readily revealed that Dnmt3b is dispensable for male germ cell development, in agreement with a previous cKO study. Our new approach enables us to analyze the germ cell functions of embryonic lethal genes in the F0 generation without using the current cKO method.


Assuntos
Quimerismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células-Tronco Embrionárias/citologia , Células Germinativas/citologia , Espermatozoides/citologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Quimera , Células-Tronco Embrionárias/metabolismo , Células Germinativas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Espermatozoides/metabolismo
13.
Biol Reprod ; 104(1): 234-243, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-32990726

RESUMO

The genus Mus consists of many species with high genetic diversity. However, only one species, Mus musculus (the laboratory mouse), is common in biomedical research. The unavailability of assisted reproductive technologies (ARTs) for other Mus species might be a major reason for their limited use in laboratories. Here, we devised ARTs for Mus spretus (the Algerian mouse), a commonly used wild-derived Mus species. We found that in vitro production of M. spretus embryos was difficult because of low efficacies of superovulation with equine chorionic gonadotropin or anti-inhibin serum (AIS) (5-8 oocytes per female) and a low fertilization rate following in vitro fertilization (IVF; 15.2%). The primary cause of this was the hardening of the zona pellucida but not the sperm's fertilizing ability, as revealed by reciprocal IVF with laboratory mice. The largest number of embryos (16 per female) were obtained when females were injected with AIS followed by human chorionic gonadotropin and estradiol injections 24 h later, and then by natural mating. These in vivo-derived 2-cell embryos could be vitrified/warmed with a high survival rate (94%) using an ethylene glycol-based solution. Importantly, more than 60% of such embryos developed into healthy offspring following interspecific embryo transfer into (C57BL/6 × C3H) F1 female mice. Thus, we have devised practical ARTs for Mus spretus mice, enabling efficient production of embryos and animals, with safe laboratory preservation of their strains. In addition, we have demonstrated that interspecific embryo transfer is possible in murine rodents.


Assuntos
Transferência Embrionária/veterinária , Técnicas de Reprodução Assistida/veterinária , Superovulação , Animais , Criopreservação/veterinária , Feminino , Masculino , Camundongos
14.
Reproduction ; 162(1): F45-F58, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33635828

RESUMO

Twenty-five years have passed since the birth of Dolly the sheep, the first mammalian clone produced by adult somatic cell nuclear transfer (SCNT). During that time, the main thrust of SCNT-related research has been the elucidation of SCNT-associated epigenetic abnormalities and their correction, with the aim of improving the efficiency of cloned animal production. Through these studies, it has become clear that some epigenomic information can be reprogrammed by the oocyte, while some cannot. Now we know that the imprinting memories in the donor genome, whether canonical (DNA-methylation-dependent) or noncanonical (H3K27me3-dependent), are not reprogrammed by SCNT. Thus, SCNT-derived embryos have the normal canonical imprinting and the erased noncanonical imprinting, both being inherited from the donor cells. The latter can cause abnormal phenotypes in SCNT-derived placentas arising from biallelic expressions of noncanonically imprinted genes. By contrast, repressive epigenomic information, such as DNA methylation and histone modifications, might be more variably reprogrammed, leaving room for technical improvements. Low-input analytical technologies now enable us to analyze the genome of gametes and embryos in a high-throughput, genome-wide manner. These technologies are being applied rapidly to the SCNT field, providing evidence for incomplete reprogramming of the donor genome in cloned embryos or offspring. Insights from the study of epigenetic phenomena in SCNT are highly relevant for our understanding of the mechanisms of genomic reprogramming that can induce totipotency in the mammalian genome.


Assuntos
Animais Geneticamente Modificados/genética , Núcleo Celular/genética , Reprogramação Celular , Clonagem de Organismos/veterinária , Epigênese Genética , Gado/genética , Técnicas de Transferência Nuclear/veterinária , Animais , Animais Geneticamente Modificados/crescimento & desenvolvimento , Aniversários e Eventos Especiais , Clonagem de Organismos/métodos , Clonagem de Organismos/tendências , Gado/crescimento & desenvolvimento
15.
Mol Reprod Dev ; 85(5): 376-386, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29457675

RESUMO

The common marmoset (Callithrix jacchus) represents a promising nonhuman primate model for the study of human diseases because of its small size, ease of handling, and availability of gene-modified animals. Here, we aimed to devise reproductive technology for marmoset spermatid injection using immature males for a possible rapid generational turnover. Spermatids at each step could be identified easily by their morphology under differential interference microscopy: thus, early round spermatids had a round nucleus with a few nucleolus-like structures and abundant cytoplasm, as in other mammals. The spermatids acquired oocyte-activating capacity at the late round spermatid stage, as confirmed by the resumption of meiosis and Ca2+ oscillations upon injection into mouse oocytes. The spermatids could be cryopreserved efficiently with a simple medium containing glycerol and CELL BANKER®. Late round or elongated spermatids first appeared at 10-12 months of age, 6-8 months before sexual maturation. Marmoset oocytes microinjected with frozen-thawed late round or elongated spermatids retrieved from a 12-month-old male marmoset developed to the 8-cell stage without the need for artificial oocyte activation stimulation. Thus, it might be possible to shorten the intergeneration time by spermatid injection, from 2 years (by natural mating) to 13-15 months including gestation.


Assuntos
Sinalização do Cálcio , Núcleo Celular/metabolismo , Criopreservação , Técnicas de Transferência Nuclear , Oócitos/metabolismo , Espermátides/metabolismo , Animais , Callithrix , Feminino , Masculino , Camundongos , Microinjeções , Oócitos/citologia , Espermátides/citologia
16.
Nature ; 486(7403): 415-9, 2012 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-22722204

RESUMO

The modification of DNA by 5-methylcytosine (5mC) has essential roles in cell differentiation and development through epigenetic gene regulation. 5mC can be converted to another modified base, 5-hydroxymethylcytosine (5hmC), by the tet methylcytosine dioxygenase (Tet) family of enzymes. Notably, the balance between 5hmC and 5mC in the genome is linked with cell-differentiation processes such as pluripotency and lineage commitment. We have previously reported that the maternal factor PGC7 (also known as Dppa3, Stella) is required for the maintenance of DNA methylation in early embryogenesis, and protects 5mC from conversion to 5hmC in the maternal genome. Here we show that PGC7 protects 5mC from Tet3-mediated conversion to 5hmC by binding to maternal chromatin containing dimethylated histone H3 lysine 9 (H3K9me2) in mice. In addition, imprinted loci that are marked with H3K9me2 in mature sperm are protected by PGC7 binding in early embryogenesis. This type of regulatory mechanism could be involved in DNA modifications in somatic cells as well as in early embryos.


Assuntos
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Embrião de Mamíferos/metabolismo , Histonas/química , Histonas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Cromatina/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona , Citosina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Feminino , Impressão Genômica/genética , Lisina/química , Lisina/metabolismo , Masculino , Metilação , Camundongos , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , RNA Longo não Codificante , RNA não Traduzido/genética , Espermatozoides/metabolismo , ras-GRF1/genética
17.
J Reprod Dev ; 64(4): 319-326, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-29731504

RESUMO

In mammals, spermatozoa activate oocytes by triggering a series of intracellular Ca2+ oscillations with phospholipase C zeta (PLCζ), a sperm-borne oocyte-activating factor. Because the introduction of PLCζ alone can induce oocyte activation, it might be a promising reagent for assisted reproductive technologies. To test this possibility, we injected human PLCζ (hPLCζ) mRNA into mouse oocytes at different concentrations. We observed the oocyte activation and subsequent embryonic development. Efficient oocyte activation and embryonic development to the blastocyst stage was achieved only with a limited range of mRNA concentrations (0.1 ng/µl). Higher concentrations of mRNA caused developmental arrest of most embryos, suggesting that excessive PLCζ protein might be harmful at this stage. In a second series of experiments, we aimed to regulate the PLCζ protein concentration in oocytes by applying auxin-inducible degron (AID) technology that allows rapid degradation of the target protein tagged with AID induced by auxin. Injection of the hPLCζ protein tagged with AID and enhanced green fluorescent protein (hPLCζ-AID-EGFP) demonstrated that high EGFP expression levels at the late 1-cell stage were efficiently reduced by auxin treatment, suggesting efficient hPLCζ degradation by this system. Furthermore, the defective development observed with higher concentrations of hPLCζ-AID-EGFP mRNA was rescued following auxin treatment. Full-term offspring were obtained by round spermatid injection with optimized hPLCζ-AID activation. Our results indicate that this AID technology can be applied to regulate the protein levels in mouse oocytes and that our optimized PLCζ system could be used for assisted fertilization in mammals.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Fosfoinositídeo Fosfolipase C/metabolismo , RNA Mensageiro/farmacologia , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Animais , Feminino , Camundongos , Oócitos/metabolismo
18.
Biol Reprod ; 96(1): 221-231, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28395324

RESUMO

Spermatogenesis is a complicated process that originates from spermatogonial stem cells (SSCs), which have self-renewal activity. Because SSCs are the only stem cells in the body that transmit genetic information to the next generation, they are an attractive target for germline modification. Although several virus vectors have been successfully used to transduce SSCs, cell toxicity or insertional mutagenesis of the transgene has limited their usage. Adeno-associated virus (AAV) is unique among virus vectors because of its target specificity and low toxicity in somatic cells, and clinical trials have shown that it has promise for gene therapy. However, there are conflicting reports on the possibility of germline integration of AAV into the genome of male germ cells, including SSCs. Here, we examined the usefulness of AAV vectors for exploring germline gene modification in SSCs. AAV1 infected cultured SSCs without apparent toxicity. Moreover, SSCs that were infected in fresh testis cells generated normal appearing spermatogenic colonies after spermatogonial transplantation. A microinsemination experiment produced offspring that underwent excision of the floxed target gene by AAV1-mediated Cre expression. Analysis of the offspring DNA showed no evidence of AAV integration, suggesting a low risk of germline integration by AAV infection. Although more extensive experiments are required to assess the risk of germline integration, our results show that AAV1 is useful for genetic manipulation of SSCs, and gene transduction by AAV will provide a useful approach to overcome potential problems associated with previous virus vector-mediated gene transduction.


Assuntos
Adenoviridae , Células-Tronco Germinativas Adultas , Técnicas de Transferência de Genes , Animais , Masculino , Camundongos Endogâmicos , Sorogrupo
19.
Hum Mol Genet ; 23(4): 992-1001, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24105465

RESUMO

Animals cloned by somatic cell nuclear transfer (SCNT) provide a unique model for understanding the mechanisms of nuclear epigenetic reprogramming to a state of totipotency. Though many phenotypic abnormalities have been demonstrated in cloned animals, the underlying mechanisms are not well understood. In this study, we performed transcriptome-wide allelic expression analyses in brain and placental tissues of cloned mice. We found that Gab1, Sfmbt2 and Slc38a4 showed loss of imprinting in all cloned mice analyzed, which might be involved in placentomegaly of cloned mice. These three genes did not require de novo DNA methylation in growing oocytes for the establishment of imprinting, implying the involvement of a de novo DNA methylation-independent mechanism. Loss of Dlk1-Dio3 imprinting was also observed in nearly half of cloned mouse embryos and showed a strong correlation with embryonic lethality. Our findings are essential to understand the underlying mechanisms of developmental abnormalities of cloned animals. We also emphasize that particular attention should be paid to specific imprinted genes for therapeutic and agricultural applications of SCNT.


Assuntos
Clonagem de Organismos , Impressão Genômica , Proteínas Adaptadoras de Transdução de Sinal , Sistema A de Transporte de Aminoácidos/genética , Animais , Sequência de Bases , Encéfalo/metabolismo , Feminino , Iodeto Peroxidase/genética , Camundongos , Fosfoproteínas/genética , Placenta/metabolismo , Gravidez , Proteínas Repressoras , Análise de Sequência de RNA , Fatores de Transcrição/genética , Transcriptoma
20.
Biochem Biophys Res Commun ; 478(2): 592-8, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27473657

RESUMO

D1Pas1 is a mouse autosomal DEAD-box RNA helicase expressed predominantly in the testis. To assess its possible function, we generated D1Pas1-deficient mice using embryonic stem cells with a targeted D1Pas1 allele. Deletion of D1Pas1 did not cause noticeable embryonic defects or death, indicating that D1Pas1 is not essential for embryogenesis. Whereas homozygous knockout female mice showed normal reproductive performance, homozygous knockout male mice were completely sterile. The seminiferous epithelium of D1Pas1-deficient males contained no spermatids or spermatozoa because of spermatogenic arrest at the late pachytene stage. Upregulation of retrotransposons such as LINE-1 was not found in D1Pas1-deficient males, unlike males lacking Mvh, another testicular DEAD-box RNA helicase. Meiotic chromosome behavior in developing spermatocytes of D1Pas1-deficient males was indistinguishable from that in wild-type males, at least until synaptonemal complex formation. Thus, mouse D1Pas1 is the first-identified DEAD-box RNA helicase that plays critical roles in the final step of the first meiotic prophase in male germ cells.


Assuntos
RNA Helicases DEAD-box/genética , Meiose , Espermatogênese , Animais , RNA Helicases DEAD-box/metabolismo , Feminino , Técnicas de Inativação de Genes , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retroelementos , Espermatócitos/citologia , Espermatócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA