Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Reprod Sci ; 24(2): 300-312, 2017 02.
Article in English | MEDLINE | ID: mdl-27342271

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs), which are well characterized and widely utilized adult stem cells, encompass the capacity to commit to a variety of cell types. This study was conducted to develop an effective way to induce goat BMSCs (gBMSCs) to transdifferentiate toward putative male germ cells by overexpressing STRA8 (stimulated by RA-8), BOULE (also called BOLL), and DAZL (deleted in azoospermia-like). First, we found that the expression levels of these 3 genes gradually increased during development of the goat testis from 10 days postnatal to 8 months old. Therefore, we hypothesized that overexpressing these genes might contribute to the transdifferentiation of gBMSCs toward germ cells. We then overexpressed, separately and in combination, STRA8, BOULE, and DAZL in gBMSCs. Our results showed that a small population of transfected gBMSCs transdifferentiated into early goat germ cell-like cells and that these cells expressed primordial germ cell specification genes STELLA (also known as DPPA3, developmental pluripotency associated 3) and C-KIT (tyrosine kinase receptor) as well as premeiotic genes MVH (mouse vasa homolog), DAZL, BOULE, STRA8, PIWIL2 (piwi-like RNA-mediated gene silencing 2), and RNF17 (ring finger protein 17). Importantly, results from quantitative reverse transcription polymerase chain reaction, immunofluorescence, and Western blot analysis showed that the meiotic marker synaptonemal complex protein 3 (SCP3) significantly increased in transfected cells compared to untransfected control cells ( P < .05). Additionally, the co-overexpression group cells had the highest SCP3 messenger RNA and protein expression levels, which indicated that 3-gene co-overexpression had the highest potential to transdifferentiate gBMSCs to germ cells. Taken together, these results demonstrate that the overexpression of STRA8, BOULE, and DAZL was able to promote the transdifferentiation of gBMSCs to early goat germ cell-like cells in vitro, which probably enhanced maturation and progression through meiosis. This approach would be important to generating gametes for future basic science as well as for potential clinical applications.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transdifferentiation/physiology , Mesenchymal Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Spermatozoa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Goats , Male , Meiosis/genetics , Mesenchymal Stem Cells/cytology , RNA-Binding Proteins/genetics , Spermatozoa/cytology
2.
PLoS One ; 9(9): e107071, 2014.
Article in English | MEDLINE | ID: mdl-25244645

ABSTRACT

Myostatin functions as a negative regulator of skeletal muscle growth by suppressing proliferation and differentiation of myoblasts. Dysfunction of the myostatin gene, either due to natural mutation or genetic manipulations such as knockout or knockdown, has been reported to increase muscle mass in mammalian species. RNA interference (RNAi) mediated by microRNAs (miRNAs) is a promising method for gene knockdown studies. In the present study, transient and stable silencing of the myostatin gene in caprine fetal fibroblasts (CFF) was evaluated using the two most effective constructs selected from four different miRNA expression constructs screened in 293FT cells. Using these two miRNA constructs, we achieved up to 84% silencing of myostatin mRNA in transiently transfected CFF cells and up to 31% silencing in stably transfected CFF cells. Moreover, off-target effects due to induction of interferon (IFN) response genes, such as interferon beta (IFN-ß) and 2'-5'-oligoadenylate synthetase 2 (OAS2), were markedly fewer in stably transfected CFF cells than in transiently transfected cells. Stable expression of anti-myostatin miRNA with minimal induction of interferon shows great promise for increasing muscle mass in transgenic goats.


Subject(s)
Fibroblasts/metabolism , Gene Silencing , MicroRNAs/metabolism , Myostatin/genetics , Animals , Gene Knockdown Techniques , Goats , HEK293 Cells , Humans , MicroRNAs/genetics , Myostatin/metabolism , RNA Interference
3.
Biochem Biophys Res Commun ; 443(2): 389-94, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24309099

ABSTRACT

Stearoyl-CoA desaturase-1 (Scd1) is a rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids. Overexpression of Scd1 in transgenic animals would modify the nutritional value of ruminant-derived foods by increasing the monounsaturated fatty acid (MUFA) and decreasing the saturated fatty acid (SFA) content. The aim of this study was to develop an effective Scd1 vector that is specifically expressed in dairy goat mammary glands. We successfully amplified the goat full length Scd1 cDNA and evaluated its activity in goat ear skin-derived fibroblast cells (GEFCs) by lipid analysis. In addition, we constructed a mammary gland-specific expression vector and confirmed efficient expression of Scd1 in goat mammary epithelial cells (GMECs) by qRT-PCR and Western blot analysis. Fatty acid analysis showed that Scd1-overexpression resulted in an increase in levels of palmitoleic acid (16:1n-7) and oleic acid (18:1n-9), from 1.73 ± 0.02% to 2.54 ± 0.02% and from 27.25 ± 0.13% to 30.37 ± 0.04%, respectively (both p<0.01) and the ratio of MUFA to SFA was increased. This work lays a foundation for the generation of Scd1 transgenic goats.


Subject(s)
Cloning, Molecular/methods , Fatty Acids, Monounsaturated/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Goats/physiology , Oleic Acid/biosynthesis , Stearoyl-CoA Desaturase/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Fatty Acids, Monounsaturated/isolation & purification , Genetic Enhancement/methods , Genetic Vectors/genetics , Oleic Acid/isolation & purification , Stearoyl-CoA Desaturase/genetics , Up-Regulation/genetics
4.
Theriogenology ; 81(3): 459-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24290662

ABSTRACT

Somatic cell nuclear transfer (SCNT) is a promising technique to produce mammalian transgenic clones. Only a small proportion of manipulated embryos, however, can develop into viable offspring. The abnormal growth and development of cloned animals, furthermore, are accompanied by aberrant lung development. Our objective was to investigate molecular background of lung developmental problems in transgenic (random insertion of exogenous DNA) cloned goats. We examined expression of 15 genes involved in growth regulation, imprinting, and epigenetic transcription in lung tissue of deceased transgenic cloned and normal goats of various ages. Compared with normal goats of the same age from conventional reproduction, expression of 13 genes (BMP4, FGF10, GHR, HGFR, PDGFR, RABP, VEGF, H19, CDKNIC, PCAF, MeCP2, HDAC1, and Dnmt3b) decreased in transgenic cloned goats that died at or shortly after birth; Expression of eight genes (FGF10, PDGFR, RABP, VEGF, PCAF, HDAC1, MeCP2, and Dnmt3b) decreased in fetal death of transgenic cloned goats. Expression of two epigenetic transcription genes (PCAF and Dnmt3b) decreased in disease death of transgenic cloned goats (1-4 months old). Disruptions in gene expression might be associated with the high neonatal mortality in transgenic cloned animals. These findings have implications in understanding the low efficiency of transgenic cloning.


Subject(s)
Animals, Genetically Modified/growth & development , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genomic Imprinting , Goats/genetics , Lung/embryology , Animals , Goats/growth & development , Lung/pathology , Nuclear Transfer Techniques/veterinary
5.
Theriogenology ; 79(2): 225-33, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23174778

ABSTRACT

Myostatin, a member of the transforming growth factor-ß family, acts as a negative regulator of skeletal muscle mass. In this study, myostatin-targeted caprine fibroblasts were obtained and subjected to SCNT to determine whether myostatin-knockout goats could be created. Fibroblasts from a 2-mo-old goat were transfected with a myostatin-targeted vector to prepare transgenic donor cells for nuclear transfer. After serum-starvation (for synchronization of the cell cycle), the percentage of transgenic fibroblasts in the G(0)/G(1) phase increased (66.2% vs. 82.9%; P < 0.05) compared with that in the control group, whereas the apoptosis rate and mitochondrial membrane potential were unaffected (P > 0.05). There were no significant differences between in vivo- and in vitro-matured oocytes as recipient cytoplasts for rates of fusion (86.5% vs. 78.4%), pregnancy (21.6% vs. 16.7%), or kidding (2.7% vs. 0%). One female kid from an in vivo-matured oocyte was born, but died a few hours later. Microsatellite analysis and polymerase chain reaction identification confirmed that this kid was genetically identical to the donor cells. Based on Western blot analysis, myostatin of the cloned kid was not expressed compared with that of nontransgenic kids. In conclusion, SCNT using myostatin-targeted 2-mo-old goat fibroblasts as donors has potential as a method for producing myostatin-targeted goats.


Subject(s)
Animals, Genetically Modified/genetics , Gene Knockout Techniques/veterinary , Goats/genetics , Myostatin/genetics , Nuclear Transfer Techniques/veterinary , Animals , Embryo Transfer/veterinary , Female , Fibroblasts/ultrastructure , Genetic Vectors , Muscle, Skeletal/growth & development , Myostatin/physiology , Oocytes/physiology , Oocytes/ultrastructure , Pregnancy
6.
In Vitro Cell Dev Biol Anim ; 48(7): 418-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22806970

ABSTRACT

Bone marrow mesenchymal stem cells (MSCs) are adult pluripotent cells that are considered to be an attractive cell type for therapy models and for nuclear transfer transgenesis. To date, MSCs from various species have been studied, but only a limited amount of information regarding dairy goat MSCs (gMSCs) is available. The objectives of this study were to isolate, induce the multilineage mesenchymal differentiation, and investigate the gene modification efficiency of gMSCs, thereby initiating further research on these cells. The gMSCs isolated from bone marrow grew, attached to plastic with a typical fibroblast-like morphology, and expressed the mesenchymal surface marker CD44, CD29, CD90, and CD166, but not the hematopoietic marker CD45. Furthermore, the gMSCs expressed the transcription factors Oct-4 and Nanog, which have been shown to be critical for stem cell self-renewal and pluripotency. The multilineage differentiation potential of gMSCs was revealed by their ability to undergo adipogenic and osteogenic differentiation when exposed to specific inducing conditions. Transient transduction of gMSCs with a plasmid containing the GFP gene resulted in higher transfection rate compared with fetal fibroblasts (FFs). Furthermore, cell colonies with stable genetic modifications were obtained when gMSCs were transfected with a mammary-specific expression vector containing human lysosomal acid beta-glucosidase gene (hGCase). In conclusion, these results demonstrated that typical mesenchymal stem cells were isolated from dairy goat bone marrow, possessed the characteristics of pluripotent stem cells, and had the potential of specific genetic modifications for gene therapy and producing transgenic goats.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Goats , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Transduction, Genetic/methods , Animals , Antigens, Surface/metabolism , Cell Culture Techniques/methods , DNA Primers/genetics , Mesenchymal Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism
7.
Sheng Wu Gong Cheng Xue Bao ; 27(2): 253-61, 2011 Feb.
Article in Chinese | MEDLINE | ID: mdl-21650051

ABSTRACT

The aim of this study was to construct a mammary gland-specific expressional vector pBC1-hLF-Neo for Human Lactoferrin (hLF) gene and then investigate its expression in the mammary gland epithelium cells. The constructed vector contained the 6.2 kb long 5' flank regulation region including promoter, other elements and the 7.1 kb long 3' flank regulation region including transcriptional ending signal of a goat's beta-casein gene. A cassette of Neo gene was also inserted into the vector which gave a total length of 26.736 kb identified by restriction fragment analysis and partial DNA sequencing. The results revealed that the structure of the final constructed vector accords with the designed plasmid map. In order to analyze the bioactivity of the vector, we transfected the lined vector DNA into the dairy goat's mammary gland epithelium cells and C127 cells of a mouse's mammary epithelium by Lipofectamine. After selection with G418 for 8-10 days, G418-risistant clones were obtained. PCR analysis demonstrated that hLF gene cassette had been integrated into the genomic DNA of G418-risistant clones. After proliferation culture, the two kinds of transgenic cells were cultured in serum-free DMEM-F12 medium with prolactin, insulin and hydrocortisone- a medium capable of inducing recombinant hLF expression. RT-PCR, Western blotting and anti-bacteria bioactivity experiments demonstrated that the constructed mammary gland specific vector pBC1-hLF-Neo possessed the desirable bioactivity to efficiently express and could secrete hLF in both mammary gland cells and have the effect of E. coli proliferation inhibition. Paramount to everything, this study laid a firm foundation for preparing the hLF gene transgenic goat fetal-derived fibroblast cells.


Subject(s)
Caseins/genetics , Epithelial Cells/metabolism , Genetic Vectors/genetics , Lactoferrin/biosynthesis , Mammary Glands, Animal/metabolism , Animals , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Female , Goats , Humans , Lactoferrin/genetics , Mammary Glands, Animal/cytology , Mice , Molecular Sequence Data , Mutagenesis, Insertional , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...