Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Reprod Fertil Dev ; 36(2): 112-123, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38064192

RESUMO

CONTEXT: Genome editing enables the introduction of beneficial sequence variants into the genomes of animals with high genetic merit in a single generation. This can be achieved by introducing variants into primary cells followed by producing a live animal from these cells by somatic cell nuclear transfer cloning. The latter step is associated with low efficiencies and developmental problems due to incorrect reprogramming of the donor cells, causing animal welfare concerns. Direct editing of fertilised one-cell embryos could circumvent this issue and might better integrate with genetic improvement strategies implemented by the industry. METHODS: In vitro fertilised zygotes were injected with TALEN editors and repair template to introduce a known coat colour dilution mutation in the PMEL gene. Embryo biopsies of injected embryos were screened by polymerase chain reaction and sequencing for intended biallelic edits before transferring verified embryos into recipients for development to term. Calves were genotyped and their coats scanned with visible and hyperspectral cameras to assess thermal energy absorption. KEY RESULTS: Multiple non-mosaic calves with precision edited genotypes were produced, including calves from high genetic merit parents. Compared to controls, the edited calves showed a strong coat colour dilution which was associated with lower thermal energy absorbance. CONCLUSIONS: Although biopsy screening was not absolutely accurate, non-mosaic, precisely edited calves can be readily produced by embryo-mediated editing. The lighter coat colouring caused by the PMEL mutation can lower radiative heat gain which might help to reduce heat stress. IMPLICATIONS: The study validates putative causative sequence variants to rapidly adapt grazing cattle to changing environmental conditions.


Assuntos
Edição de Genes , Genoma , Animais , Bovinos , Genótipo , Embrião de Mamíferos , Mutação
2.
Mol Reprod Dev ; 88(1): 3-14, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33251684

RESUMO

Multiplying the germline would increase the number of offspring that can be produced from selected animals, accelerating genetic improvement for livestock breeding. This could be achieved by producing multiple chimaeric animals, each carrying a mix of donor and host germ cells in their gonads. However, such chimaeric germlines would produce offspring from both donor and host genotypes, limiting the rate of genetic improvement. To resolve this problem, we disrupted the RNA-binding protein DAZL and generated germ cell-deficient host animals. Using Cas9-mediated homology-directed repair (HDR), we introduced a DAZL loss-of-function mutation in male ovine fetal fibroblasts. Following manual single cell isolation, 4/48 (8.3%) of donor cell strains were homozygously HDR-edited. Sequence-validated strains were used as nuclear donors for somatic cell cloning to generate three lambs, which died at birth. All DAZL null male neonatal sheep lacked germ cells on histological sections and showed greatly reduced germ cell markers. Somatic cells within their testes were morphologically intact and expressed normal levels of lineage-specific markers, suggesting that the germ cell niche remained intact. This extends the DAZL mutant phenotype beyond mice into agriculturally relevant ruminants, providing a pathway for using absolute germline transmitters in rapid livestock improvement.


Assuntos
Fibroblastos/metabolismo , Mutação com Perda de Função , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ovinos/metabolismo , Espermatogônias/metabolismo , Testículo/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Sequência de Bases , Biomarcadores/metabolismo , Cruzamento/métodos , Células Cultivadas , Edição de Genes/métodos , Expressão Gênica , Masculino , Camundongos , Fenótipo , Reparo de DNA por Recombinação/genética , Ovinos/genética
3.
Biol Reprod ; 103(1): 114-125, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32318688

RESUMO

Correct reprogramming of epigenetic marks in the donor nucleus is a prerequisite for successful cloning by somatic cell transfer (SCT). In several mammalian species, repressive histone (H) lysine (K) trimethylation (me3) marks, in particular H3K9me3, form a major barrier to somatic cell reprogramming into pluripotency and totipotency. We engineered bovine embryonic fibroblasts (BEFs) for the doxycycline-inducible expression of a biologically active, truncated form of murine Kdm4b, a demethylase that removes H3K9me3 and H3K36me3 marks. Upon inducing Kdm4b, H3K9me3 and H3K36me3 levels were reduced about 3-fold and 5-fold, respectively, compared with noninduced controls. Donor cell quiescence has been previously associated with reduced somatic trimethylation levels and increased cloning efficiency in cattle. Simultaneously inducing Kdm4b expression (via doxycycline) and quiescence (via serum starvation) further reduced global H3K9me3 and H3K36me3 levels by a total of 18-fold and 35-fold, respectively, compared with noninduced, nonstarved control fibroblasts. Following SCT, Kdm4b-BEFs reprogrammed significantly better into cloned blastocysts than noninduced donor cells. However, detrimethylated donors and sustained Kdm4b-induction during embryo culture did not increase the rates of postblastocyst development from implantation to survival into adulthood. In summary, overexpressing Kdm4b in donor cells only improved their reprogramming into early preimplantation stages, highlighting the need for alternative experimental approaches to reliably improve somatic cloning efficiency in cattle.


Assuntos
Blastocisto/fisiologia , Bovinos/embriologia , Reprogramação Celular/fisiologia , Clonagem de Organismos , Histonas/metabolismo , Técnicas de Transferência Nuclear , Animais , Reprogramação Celular/genética , Desmetilação , Desenvolvimento Embrionário/fisiologia , Epigênese Genética , Feminino , Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/fisiologia , Camundongos , Regulação para Cima
4.
Mol Biol Rep ; 46(2): 1737-1746, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30694456

RESUMO

Episomal plasmids based on a scaffold/matrix attachment region (S/MAR) are extrachromosomal DNA entities that replicate once per cell cycle and are stably maintained in cells or tissue. We generated minicircles, episomal plasmids devoid of bacterial sequences, and show that they are stably transmitted in clonal primary bovine fibroblasts without selection pressure over more than two months. Total DNA, plasmid extraction and fluorescence in situ hybridization (FISH) analyses suggest that the minicircles remained episomal and were not integrated into the genome. Minicircles survived extended periods in serum-starved cells, which indicates that ongoing transcription in non-proliferating cells is not necessary for the maintenance of S/MAR-episomes. To test whether minicircles endure the process of somatic cell nuclear transfer (SCNT), we used cell-cycle synchronized, serum-starved, minicircle-containing cells. Analysis of cells outgrown from SCNT-derived blastocysts shows that the minicircles are maintained through SCNT and early embryonic development, which raises the prospect of using cell lines with episomal minicircles for the generation of transgenic animals.


Assuntos
DNA Circular/fisiologia , Plasmídeos/genética , Plasmídeos/fisiologia , Animais , Animais Geneticamente Modificados/genética , Blastocisto , Bovinos , DNA Circular/genética , Vetores Genéticos/genética , Hibridização in Situ Fluorescente , Técnicas de Transferência Nuclear/veterinária
5.
Biol Reprod ; 95(1): 16, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27281704

RESUMO

Reprogramming by nuclear transfer (NT) cloning forces cells to lose their lineage-specific epigenetic marks and reacquire totipotency. This process often produces molecular anomalies that compromise clone development. We hypothesized that quiescence alters the epigenetic status of somatic NT donor cells and elevates their reprogrammability. To test this idea, we compared chromatin composition and cloning efficiency of serum-starved quiescent (G0) fibroblasts versus nonstarved mitotically selected (G1) controls. We show that G0 chromatin contains reduced levels of Polycomb group proteins EED, SUZ12, PHC1, and RING2, as well as histone variant H2A.Z. Using quantitative confocal immunofluorescence microscopy and fluorometric enzyme-linked immunosorbent assay, we further show that G0 induced DNA and histone hypomethylation, specifically at H3K4me3, H3K9me2/3 and H3K27me3, but not H3K9me1. Collectively, these changes resulted in a more relaxed G0 chromatin state. Following NT, G0 donors developed into blastocysts that retained H3K9me3 hypomethylation, both in the inner cell mass and trophectoderm. G0 blastocysts from different cell types and cell lines developed significantly better into adult offspring. In conclusion, serum starvation induced epigenetic changes, specifically hypotrimethylation, that provide a mechanistic correlate for increased somatic cell reprogrammability.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Reprogramação Celular/fisiologia , Epigênese Genética , Fibroblastos/citologia , Mitose/fisiologia , Animais , Bovinos , Proteínas de Ciclo Celular/genética , Fibroblastos/metabolismo , Histonas/metabolismo , Técnicas de Transferência Nuclear
6.
Biol Reprod ; 93(6): 132, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26510863

RESUMO

The inner cell mass (ICM) of mammalian blastocysts consists of pluripotent epiblast and hypoblast lineages, which develop into embryonic and extraembryonic tissues, respectively. We conducted a chemical screen for regulators of epiblast identity in bovine Day 8 blastocysts. From the morula stage onward, in vitro fertilized embryos were cultured in the presence of cell-permeable small molecules targeting nine principal signaling pathway components, including TGFbeta1, BMP, EGF, VEGF, PDGF, FGF, cAMP, PI3K, and JAK signals. Using 1) blastocyst quality (by morphological grading), 2) cell numbers (by differential stain), and 3) epiblast (FGF4, NANOG) and hypoblast (PDGFRa, SOX17) marker gene expression (by quantitative PCR), we identified positive and negative regulators of ICM development and pluripotency. TGFbeta1, BMP, and cAMP and combined VEGF/PDGF/FGF signals did not affect blastocyst development while PI3K was important for ICM growth but did not alter lineage-specific gene expression. Stimulating cAMP specifically increased NANOG expression, while combined VEGF/PDGF/FGF inhibition up-regulated epiblast and hypoblast markers. The strongest effects were observed by suppressing JAK1/2 signaling with AZD1480. This treatment interfered with ICM formation, but trophectoderm cell numbers and markers (CDX2, KTR8) were not altered. JAK inhibition repressed both epiblast and hypoblast transcripts as well as naive pluripotency-related genes (KLF4, TFCP2L1) and the JAK substrate STAT3. We found that tyrosine (Y) 705-phosphorylated STAT3 (pSTAT3(Y705)) was restricted to ICM nuclei, where it colocalized with SOX2 and NANOG. JAK inhibition abolished this ICM-exclusive pSTAT3(Y705) signal and strongly reduced the number of SOX2-positive nuclei. In conclusion, JAK/STAT3 activation is required for bovine ICM formation and acquisition of naive pluripotency markers.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Desenvolvimento Embrionário/fisiologia , Janus Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Animais , Massa Celular Interna do Blastocisto/citologia , Massa Celular Interna do Blastocisto/efeitos dos fármacos , Bovinos , Desenvolvimento Embrionário/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Janus Quinases/antagonistas & inibidores , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
7.
Biol Reprod ; 91(2): 49, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25009207

RESUMO

Mammalian blastocysts comprise three distinct lineages, namely, trophoblast, hypoblast, and epiblast, which develop into fetal placenta, extraembryonic yolk sac, and embryo proper, respectively. Pluripotent embryonic stem cells, capable of forming all adult cell types, can only be derived from the epiblast. In mouse and rat, this process is promoted by the double inhibition (2i) of mitogen-activated protein kinase kinase (MAP2K), which antagonizes FGF signaling, and glycogen synthase kinase 3 (GSK3), which stimulates the WNT pathway. We investigated variations of the 2i treatment on lineage segregation and pluripotency-related gene expression in bovine blastocysts. In vitro-fertilized embryos were cultured either in the presence of inhibitors of GSK3 (3 µM CHIR) and MAP2K (0.4 vs. 10 µM PD0325901, designated 2i and 2i+, respectively) or in 2i/2i+ with FGFR inhibitor (0.1 µM PD173074, designated 3i [2i and PD173074] and 3i+ [2i+ and PD173074]). Compared with 2i, both 2i+ and 3i+ potentiated the improvement in blastocyst morphology. Using an automated platform for multiplexed digital mRNA profiling, we simultaneously counted transcripts of 76 candidate genes in bovine blastocysts treated with multiple kinase inhibitors. We show that 2i+ medium specifically increased FGF4 and NANOG while reducing PDGFRalpha and SOX17 levels. The shift from a hypoblast to an epiblast gene expression signature was confirmed by quantitative PCR. A wide range of functionally related genes, including candidates involved in DNA methylation, were not significantly changed. This well-defined 2i+ effect was not observed after pharmacologically inhibiting FGF receptor or related MAP kinases (p38, JNK, and ERK5). In summary, our data suggest that increased MAP2K inhibition exerts its pluripotency-promoting effects through as yet unidentified signals.


Assuntos
Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Bovinos/embriologia , Camadas Germinativas/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Animais , Benzamidas/farmacologia , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Técnicas de Cultura Embrionária , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Pirimidinas/farmacologia
8.
Biol Reprod ; 88(3): 74, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23390165

RESUMO

Cells in the mammalian blastocyst segregate into three distinct lineages, namely, trophoblast, hypoblast, and epiblast. During development, these will form extraembryonic and embryonic tissues, respectively. In mouse, only epiblast cells can be directly converted into cultured pluripotent embryonic stem cells, capable of forming all adult cell types. This conversion is promoted by the double inhibition (i.e., 2i) of mitogen-activated protein kinase kinase (Map2k), antagonizing Fgf signaling, and of glycogen synthase kinase 3 (Gsk3), stimulating the Wnt pathway. We investigated the effect of 2i treatment on lineage segregation and pluripotency-related gene expression in bovine blastocysts. In vitro fertilized (IVF) embryos were cultured in the presence of dimethyl sulfoxide or inhibitors of MAP2K (0.4 µM PD0325901) and GSK3 (3 µM CHIR99021) from the zygote (Day 1) stage. Compared to vehicle controls, 2i conditions increased the abundance of cumulus cells in bovine IVF cultures, which compromised blastocyst formation. Following cumulus removal, 2i accelerated blastocyst development and increased inner cell mass (ICM) and trophoblast cell numbers by 30% and 27%, respectively. These developmental and morphological changes were accompanied by alterations in gene expression. Signal inhibition increased transcription of putative epiblast markers NANOG and SOX2 while repressing putative hypoblast marker GATA4. Using microsurgical blastocyst dissection, we found that the increase in NANOG and SOX2 levels was specific to the ICM and not due to ectopic expression in the trophoblast. Expression of other pluripotency-related (POU5F1, KLF4, DPPA3) or trophoblast-enriched (CDX2) genes was not affected. In summary, 2i conditions reprogrammed the transcriptional profile of bovine ICM but not trophoblast cells. By shifting the balance from hypoblast- to epiblast-associated gene expression, 2i culture may prime bovine epiblast for subsequent derivation of pluripotent stem cell cultures.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Camadas Germinativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Bovinos , Feminino , Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Fator 4 Semelhante a Kruppel , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Trofoblastos/metabolismo
9.
Reproduction ; 145(1): 97-108, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23137934

RESUMO

Transgenic mammals have been produced using sperm as vectors for exogenous DNA (sperm-mediated gene transfer (SMGT)) in combination with artificial insemination. Our study evaluated whether SMGT could also be achieved in combination with IVF to efficiently produce transgenic bovine embryos. We assessed binding and uptake of fluorescently labelled plasmids into sperm in the presence of different concentrations of dimethyl sulphoxide or lipofectamine. Live motile sperm displayed a characteristic punctuate fluorescence pattern across their entire surface, while uniform postacrosomal fluorescence was only apparent in dead sperm. Association with sperm or lipofection reagent protected exogenous DNA from DNase I digestion. Following IVF, presence and expression of episomal and non-episomal green fluorescent protein (GFP)-reporter plasmids was monitored in oocytes and embryos. We found no evidence of intracellular plasmid uptake and none of the resulting zygotes (n=96) and blastocysts were GFP positive by fluorescence microscopy or genomic PCR (n=751). When individual zona-free oocytes were matured, fertilised and continuously cultured in the presence of episomal reporter plasmids until the blastocyst stage, most embryos (38/68=56%) were associated with the exogenous DNA. Using anti-GFP immunocytochemistry (n=48) or GFP fluorescence (n=94), no GFP expression was detected in blastocysts. By contrast, ICSI resulted in 18% of embryos expressing the GFP reporter. In summary, exposure to DNA was an inefficient technique to produce transgenic bovine sperm or blastocysts in vitro.


Assuntos
Animais Geneticamente Modificados/genética , Bovinos/genética , DNA/farmacologia , Embrião de Mamíferos/efeitos dos fármacos , Técnicas de Transferência de Genes , Espermatozoides/efeitos dos fármacos , Animais , Células Cultivadas , DNA/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Fertilização in vitro/métodos , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Inseminação Artificial/métodos , Masculino , Plasmídeos , Injeções de Esperma Intracitoplásmicas/métodos , Espermatozoides/metabolismo , Transfecção
10.
Trends Biotechnol ; 41(10): 1237-1247, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37173191

RESUMO

Animal breeding drives genetic progress mainly through the male germline. This process is slow to respond to rapidly mounting environmental pressures that threaten sustainable food security from animal protein production. New approaches promise to accelerate breeding by producing chimaeras, which comprise sterile host and fertile donor genotypes, to exclusively transmit elite male germlines. Following gene editing to generate sterile host cells, the missing germline can be restored by transplanting either: (i) spermatogonial stem cells (SSCs) into the testis; or (ii) embryonic stem cells (ESCs) into early embryos. Here we compare these alternative germline complementation strategies and their impact on agribiotechnology and species conservation. We propose a novel breeding platform that integrates embryo-based complementation with genomic selection, multiplication, and gene modification.


Assuntos
Espermatogônias , Testículo , Masculino , Animais , Espermatogônias/metabolismo , Espermatogônias/transplante , Testículo/metabolismo , Edição de Genes
11.
Front Cell Dev Biol ; 11: 1112069, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37745294

RESUMO

Mutations in the transcription factor gene grainyhead-like 2 (GRHL2) are associated with progressive non-syndromic sensorineural deafness autosomal dominant type 28 (DFNA28) in humans. Since complete loss of Grhl2 is lethal in mouse embryos, we studied its role during inner ear pathology and hearing loss in vitro. To this end, we generated different homozygous deletions to knockout Grhl2 in mouse embryonic stem cells (Grhl2-KO ESCs), including some mimicking naturally occurring truncations in the dimerisation domain related to human DFNA28. Under naïve culture conditions, Grhl2-KO cells in suspension were more heterogenous in size and larger than wild-type controls. Adherent Grhl2-KO cells were also larger, with a less uniform shape, flattened, less circular morphology, forming loose monolayer colonies with poorly defined edges. These changes correlated with lower expression of epithelial cadherin Cdh1 but no changes in tight junction markers (Ocln, Tjp2) or other Grhl isoforms (Grhl1, Grhl3). Clonogenicity from single cells, proliferation rates of cell populations and proliferation markers were reduced in Grhl2-KO ESCs. We next induced stepwise directed differentiation of Grhl2-KO ESCs along an otic pathway, giving rise to three-dimensional inner ear-like organoids (IELOs). Quantitative morphometry revealed that Grhl2-KO cells initially formed larger IELOs with a less compacted structure, more eccentric shape and increased surface area. These morphological changes persisted for up to one week. They were partially rescued by forced cell aggregation and fully restored by stably overexpressing exogenous Grhl2 in Grhl2-KO ESCs, indicating that Grhl2 alters cell-cell interactions. On day 8, aggregates were transferred into minimal maturation medium to allow self-guided organogenesis for another two weeks. During this period, Grhl2-KO cells and wild-type controls developed similarly, expressing neural, neuronal and sensory hair cell markers, while maintaining their initial differences in size and shape. In summary, Grhl2 is required for morphological maintenance of ESCs and orderly formation of IELOs, consistent with an essential role in organising epithelial integrity during inner ear development. Our findings validate quantitative morphometry as a useful, non-invasive screening method for molecular phenotyping of candidate mutations during organoid development.

12.
Front Genet ; 13: 933534, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36246653

RESUMO

Cloning multiple animals from genomically selected donor embryos is inefficient but would accelerate genetic gain in dairy cattle breeding. To improve embryo cloning efficiency, we explored the idea that epigenetic reprogramming improves when donor cells are in mitosis. We derived primary cultures from bovine inner cell mass (ICM) cells of in vitro fertilized (IVF) embryos. Cells were grown feeder-free in a chemically defined medium with increased double kinase inhibition (2i+). Adding recombinant bovine interleukin 6 to 2i+ medium improved plating efficiency, outgrowth expansion, and expression of pluripotency-associated epiblast marker genes (NANOG, FGF4, SOX2, and DPPA3). For genotype multiplication by embryonic cell transfer (ECT) cloning, primary colonies were treated with nocodazole, and single mitotic donors were harvested by mechanical shake-off. Immunofluorescence against phosphorylated histone 3 (P-H3) showed 37% of nocodazole-treated cells in metaphase compared to 6% in DMSO controls (P < 1 × 10-5), with an average of 53% of P-H3-positive cells expressing the pluripotency marker SOX2. We optimized several parameters (fusion buffer, pronase treatment, and activation timing) for ECT with mitotic embryonic donors. Sequential double cytoplast ECT, whereby another cytoplast was fused to the first cloned reconstruct, doubled cloned blastocyst development and improved morphological embryo quality. However, in situ karyotyping revealed that over 90% of mitotic ECT-derived blastocysts were tetraploid or aneuploid with extra chromosomes, compared to less than 2% in the original ICM donor cells. Following the transfer of single vs. double cytoplast embryos, there was no difference between the two methods in pregnancy establishment at D35 (1/22 = 5% vs. 4/53 = 8% for single vs. double ECT, respectively). Overall, post-implantation development was drastically reduced from embryonic mitotic clones when compared to somatic interphase clones and IVF controls. We conclude that mitotic donors cause ploidy errors during in vitro development that cannot be rescued by enhanced epigenetic reprogramming through double cytoplast cloning.

13.
Cell Reprogram ; 23(1): 14-25, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33529123

RESUMO

Zona-free somatic cell transfer (SCT) and embryo aggregation increase throughput and efficiency of cloned embryo and offspring production, respectively, but both approaches have not been widely adopted. Cloning efficiency is further improved by cell cycle coordination between the interphase donor cell and metaphase-arrested recipient cytoplast. This commonly involves inclusion of caffeine and omission of calcium to maintain high mitotic cyclin-dependent kinase activity and low calcium levels, respectively, in the nonactivated cytoplast. The aim of our study was to integrate these various methodological improvements into a single work stream that increases sheep cloning success. We show that omitting calcium during zona-free SCT improved blastocyst development from 6% to 13%, while caffeine treatment reduced spontaneous oocyte activation from 17% to 8%. In a retrospective analysis, morula aggregation produced high morphological quality blastocysts with better in vivo survival to term than nonaggregated controls (15% vs. 9%), particularly after vitrification (14% vs. 0%). By combining cytoplast cell cycle control with zona-free embryo reconstruction and aggregation, this novel SCT protocol maximizes the benefits of vitrification by producing more cryoresilient blastocysts. The presented cloning methodology is relatively easy to operate and further increases throughput and efficiency of cloned embryo and offspring production. Integration of additional reprogramming steps or alternate donor cells is straightforward, providing a flexible workflow that can be adapted to changing experimental requirements.


Assuntos
Blastocisto/citologia , Clonagem de Organismos/métodos , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Mórula/citologia , Técnicas de Transferência Nuclear/veterinária , Oócitos/citologia , Animais , Ovinos , Vitrificação
14.
Biomed Microdevices ; 12(5): 777-86, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20499188

RESUMO

Nuclear transfer (NT) cloning involves manual positioning of individual donor-recipient cell couplets for electrofusion. This is time-consuming and introduces operator-dependent variation as a confounding parameter in cloning trials. In order to automate the NT procedure, we developed a micro-fluidic device that integrates automated cell positioning and electrofusion of isolated cell couplets. A simple two layer micro-fluidic device was fabricated. Thin film interdigitated titanium electrodes (300 nm thick, 250 microm wide and 250 microm apart) were deposited on a solid borosilicate glass substrate. They were coated with a film of electrically insulating photosensitive epoxy polymer (SU-8) of either 4 or 22 microm thickness. Circular holes ("micropits") measuring 10, 20, 30, 40 or 80 microm in diameter were fabricated above the electrodes. The device was immersed in hypo-osmolar fusion buffer and manually loaded with somatic donor cells and recipient oocytes. Dielectrophoresis (DEP) was used to attract cells towards the micropit and form couplets on the same side of the insulating film. Fusion pulses between 80 V and 120 V were applied to each couplet and fusion scored under a stereomicroscope. Automated couplet formation between oocytes and somatic cells was achieved using DEP. Bovine oocyte-oocyte, oocyte-follicular cells and oocyte-fibroblast couplets fused with up to 69% (n = 13), 50% (n = 30) and 78% (n = 9) efficiency, respectively. Fusion rates were comparable to parallel plate or film electrodes that are conventionally used for bovine NT. This demonstrates proof-of-principle that a micropit device is capable of both rapid cell positioning and fusion.


Assuntos
Fusão Celular/instrumentação , Eletroforese/instrumentação , Técnicas Analíticas Microfluídicas , Técnicas de Transferência Nuclear/instrumentação , Animais , Automação , Bovinos , Eletrodos , Feminino , Modelos Teóricos , Oocistos/citologia , Integração de Sistemas
15.
Biomed Microdevices ; 11(4): 851-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19365731

RESUMO

Automated lab on chip systems offer increased throughput and reproducibility, but the implementation of microelectrodes presently relies on miniaturization of parallel plate electrodes that are time consuming and costly to fabricate. Electric field modelling of open electrofusion chambers suggested that widely spaced (> or =2 mm) coplanar film electrodes should result in similar cell fusion rates as parallel plate electrodes provided the cell positioning was roughly midway between the electrodes. This hypothesis was investigated by electrofusion trials of bovine oocyte-donor cell couplets used in nuclear transfer (NT) cloning. Comparative experiments with reference parallel plate electrodes were conducted as controls. Coplanar fusion rates > or = 90% were demonstrated for embryonic blastomeres, follicular cells and fetal and adult fibroblasts as NT donor cells. For embryonic and adult cell types, there was no significant difference in fusion rate between coplanar and parallel plate electrodes. For both electrode geometries, fusion efficiency with adult fibroblasts was highest at a calculated field strength of 2.33 kV/cm. The coplanar electrodes required a voltage pi/2 times greater than parallel plate electrodes to achieve equivalent field strength when the couplets are placed midway between the electrodes.


Assuntos
Microeletrodos , Técnicas de Transferência Nuclear/instrumentação , Animais , Blastômeros/citologia , Bovinos , Linhagem Celular , Feminino , Feto/citologia , Fibroblastos/citologia , Masculino , Oócitos/citologia
16.
Reprod Fertil Dev ; 21(1): 83-94, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19152749

RESUMO

Following nuclear transfer (NT), the most stringent measure of extensive donor cell reprogramming is development into viable offspring. This is referred to as cloning efficiency and quantified as the proportion of cloned embryos transferred into surrogate mothers that survive into adulthood. Cloning efficiency depends on the ability of the enucleated recipient cell to carry out the reprogramming reactions ('reprogramming ability') and the ability of the nuclear donor cell to be reprogrammed ('reprogrammability'). It has been postulated that reprogrammability of the somatic donor cell epigenome is inversely proportional to its differentiation status. In order to test this hypothesis, reprogrammability was compared between undifferentiated stem cells and their differentiated isogenic progeny. In the mouse, cells of divergent differentiation status from the neuronal, haematopoietic and skin epithelial lineage were tested. In cattle and deer, skeletal muscle and antler cells, respectively, were used as donors. No conclusive correlation between differentiation status and cloning efficiency was found, indicating that somatic donor cell type may not be the limiting factor for cloning success. This may reflect technical limitations of the NT-induced reprogramming assay. Alternatively, differentiation status and reprogrammability may be unrelated, making all cells equally difficult to reprogramme once they have left the ground state of pluripotency.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Clonagem de Organismos/métodos , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Animais , Camundongos , Especificidade da Espécie
17.
Reprod Domest Anim ; 43 Suppl 2: 407-16, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18638154

RESUMO

Despite more than a decade of research efforts, farm animal cloning by somatic cell nuclear transfer (SCNT) is still frustratingly inefficient. Inefficiency manifests itself at different levels, which are currently not well integrated. At the molecular level, it leads to widespread genetic, epigenetic and transcriptional aberrations in cloned embryos. At the organismal level, these genome-wide abnormalities compromise development of cloned foetuses and offspring. Specific molecular defects need to be causally linked to specific cloned phenotypes, in order to design specific treatments to correct them. Cloning efficiency depends on the ability of the nuclear donor cell to be fully reprogrammed into an embryonic state and the ability of the enucleated recipient cell to carry out the reprogramming reactions. It has been postulated that reprogrammability of the somatic donor cell epigenome is influenced by its differentiation status. However, direct comparisons between cells of divergent differentiation status within several somatic lineages have found no conclusive evidence for this. Choosing somatic stem cells as donors has not improved cloning efficiency, indicating that donor cell type may be less critical for cloning success. Different recipient cells, on the other hand, vary in their reprogramming ability. In bovine, using zygotes instead of oocytes has increased cloning success. Other improvements in livestock cloning efficiency include better coordinating donor cell type with cell cycle stage and aggregating cloned embryos. In the future, it will be important to demonstrate if these small increases at every step are cumulative, adding up to an integrated cloning protocol with greatly improved efficiency.


Assuntos
Animais Geneticamente Modificados , Clonagem de Organismos/veterinária , Epigênese Genética , Técnicas de Transferência Nuclear/veterinária , Animais , Animais Domésticos , Clonagem de Organismos/métodos , Técnicas de Transferência Nuclear/normas , Partenogênese
18.
Biol Reprod ; 76(2): 268-78, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17050861

RESUMO

Our objectives were to compare the cellular and molecular effects of aggregating bovine embryonic vs. somatic cell nuclear transfer (ECNT vs. SCNT) embryos and to determine whether aggregation can improve cattle cloning efficiency. We reconstructed cloned embryos from: 1) morula-derived blastomeres, 2) six adult male ear skin fibroblast lines, 3) one fetal female lung fibroblast line (BFF), and 4) two transgenic clonal strains derived from BFF. Embryos were cultured either singularly (1X) or as aggregates of three (3X). In vitro-fertilized (IVF) 1X and 3X embryos served as controls. After aggregation, the in vitro development of ECNT but not that of SCNT or IVF embryos was strongly compromised. The inner cell mass (ICM), total cell (TC) numbers, and ICM:TC ratios significantly increased for all the aggregates. The relative concentration of the key embryonic transcript POU5F1 (or OCT4) did not correlate with these increases, remaining unchanged in the ECNT and IVF aggregates and decreasing significantly in the SCNT aggregates. Overall, the IVF and 3X ECNT but not the 1X ECNT embryos had significantly higher relative POU5F1 levels than the SCNT embryos. High POU5F1 levels correlated with high in vivo survival, while no such correlation was noted for the ICM:TC ratios. Development to weaning was more than doubled in the ECNT aggregates (10/51 or 20% vs. 7/85 or 8% for 3X vs. 1X, respectively; P < 0.05). In contrast, the SCNT and IVF controls showed no improvement in survival. These data reveal striking biological differences between embryonic and somatic clones in response to aggregation.


Assuntos
Bovinos/embriologia , Clonagem de Organismos , Técnicas de Transferência Nuclear , Técnicas de Reprodução Assistida , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos/crescimento & desenvolvimento , Blastocisto , Massa Celular Interna do Blastocisto , Células Cultivadas , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Feminino , Fertilização in vitro , Técnicas In Vitro , Masculino , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sobrevida , Trofoblastos/citologia , Desmame
19.
Adv Exp Med Biol ; 591: 30-57, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17176553

RESUMO

Somatic cell nuclear transfer (SCNT) is much more widely and efficiently practiced in cattle than in any other species, making this arguably the most important mammal cloned to date. While the initial objective behind cattle cloning was commercially driven--in particular to multiply genetically superior animals with desired phenotypic traits and to produce genetically modified animals-researchers have now started to use bovine SCNT as a tool to address diverse questions in developmental and cell biology. In this paper, we review current cattle cloning methodologies and their potential technical or biological pitfalls at any step of the procedure. In doing so, we focus on one methodological parameter, namely donor cell selection. We emphasize the impact of epigenetic and genetic differences between embryonic, germ, and somatic donor cell types on cloning efficiency. Lastly, we discuss adult phenotypes and fitness of cloned cattle and their offspring and illustrate some of the more imminent commercial cattle cloning applications.


Assuntos
Bovinos/genética , Núcleo Celular/genética , Clonagem de Organismos/métodos , Clonagem de Organismos/tendências , Técnicas de Transferência Nuclear/tendências , Animais , Clonagem de Organismos/normas , Desenvolvimento Embrionário/genética , Epigênese Genética/genética , Genótipo , Células Germinativas/metabolismo , Humanos , Técnicas de Transferência Nuclear/normas , Criação de Embriões para Pesquisa/métodos , Criação de Embriões para Pesquisa/normas
20.
Sci Rep ; 7(1): 7514, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28790329

RESUMO

Correct reprogramming of epigenetic marks is essential for somatic cells to regain pluripotency. Repressive histone (H) lysine (K) methylation marks are known to be stable and difficult to reprogram. In this study, we generated transgenic mice and mouse embryonic fibroblasts (MEFs) for the inducible expression of KDM4B, a demethylase that removes H3 K9 and H3K36 trimethylation (me3) marks (H3K9/36me3). Upon inducing Kdm4b, H3K9/36me3 levels significantly decreased compared to non-induced controls. Concurrently, H3K9me1 levels significantly increased, while H3K9me2 and H3K27me3 remained unchanged. The global transcriptional impact of Kdm4b-mediated reduction in H3K9/36me3 levels was examined by comparative microarray analysis and mRNA-sequencing of three independent transgenic MEF lines. We identified several commonly up-regulated targets, including the heterochromatin-associated zinc finger protein 37 and full-length endogenous retrovirus repeat elements. Following optimized zona-free somatic nuclear transfer, reduced H3K9/36me3 levels were restored within hours. Nevertheless, hypo-methylated Kdm4b MEF donors reprogrammed six-fold better into cloned blastocysts than non-induced donors. They also reprogrammed nine-fold better into induced pluripotent stem cells that gave rise to teratomas and chimeras. In summary, we firmly established H3K9/36me3 as a major roadblock to somatic cell reprogramming and identified transcriptional targets of derestricted chromatin that could contribute towards improving this process in mouse.


Assuntos
Reprogramação Celular , Fibroblastos/metabolismo , Histonas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Processamento de Proteína Pós-Traducional , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Hibridização Genômica Comparativa , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Fibroblastos/citologia , Histonas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metilação , Camundongos , Camundongos Transgênicos , Técnicas de Transferência Nuclear , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Fatores de Transcrição , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA