Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Mol Genet Genomics ; 299(1): 41, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38551742

RESUMO

Primordial germ cells (PGCs) are the ancestors of female and male germ cells. Recent studies have shown that long non-coding RNA (lncRNA) and histone methylation are key epigenetic factors affecting PGC formation; however, their joint regulatory mechanisms have rarely been studied. Here, we explored the mechanism by which lncCPSET1 and H3K4me2 synergistically regulate the formation of chicken PGCs for the first time. Combined with chromatin immunoprecipitation (CHIP) sequencing and RNA-seq of PGCs transfected with the lncCPSET1 overexpression vector, GO annotation and KEGG enrichment analysis revealed that Wnt and TGF-ß signaling pathways were significantly enriched, and Fzd2, Id1, Id4, and Bmp4 were identified as candidate genes. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) showed that ASH2L, DPY30, WDR5, and RBBP5 overexpression significantly increased the expression of Bmp4, which was up-regulated after lncCPSET1 overexpression as well. It indicated that Bmp4 is a target gene co-regulated by lncCPSET1 and MLL2/COMPASS. Interestingly, co-immunoprecipitation results showed that ASH2L, DPY30 and WDR5 combined and RBBP5 weakly combined with DPY30 and WDR5. lncCPSET1 overexpression significantly increased Dpy30 expression and co-immunoprecipitation showed that interference/overexpression of lncCPSET1 did not affect the binding between the proteins in the complexes, but interference with lncCPSET1 inhibited DPY30 expression, which was confirmed by RNA immunoprecipitation that lncCPSET1 binds to DPY30. Additionally, CHIP-qPCR results showed that DPY30 enriched in the Bmp4 promoter region promoted its transcription, thus promoting the formation of PGCs. This study demonstrated that lncCPSET1 and H3K4me2 synergistically promote PGC formation, providing a reference for the study of the regulatory mechanisms between lncRNA and histone methylation, as well as a molecular basis for elucidating the formation mechanism of PGCs in chickens.


Assuntos
Galinhas , RNA Longo não Codificante , Masculino , Animais , Feminino , Galinhas/genética , Galinhas/metabolismo , Histonas/genética , Histonas/metabolismo , RNA Longo não Codificante/metabolismo , Metilação , Células Germinativas
2.
EMBO Rep ; 23(1): e52491, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34747116

RESUMO

The unique characteristics of chicken primordial germ cells (PGCs) provide potential strategies for transgenic animal generation; however, insufficient PGC availability has limited their application. Regulation of bone morphogenic protein 4 (BMP4), a crucial factor for PGCs formation, may provide new strategies for PGC generation. We here identify a long noncoding RNA (lncRNA) that targets BMP4 (LncBMP4). LncBMP4 has similar functions as BMP4, in that it facilitates the formation and migration of PGCs. LncBMP4 promotes BMP4 expression by adsorbing the miRNA gga-mir-12211, thus reducing its inhibitory effect on BMP4 expression. In addition, the small peptide EPC5 encoded by LncBMP4 promotes the transcription of BMP4. The competing endogenous RNA (ceRNA) effect of LncBMP4 requires N6-methyladenosine (m6A) modification, in a dose-dependent manner, and high levels of m6A modification hinder EPC5 translation. Understanding the molecular mechanisms through which LncBMP4 promotes BMP4 expression during PGC formation may provide new avenues for efficient PGC generation.


Assuntos
MicroRNAs , RNA Longo não Codificante , Animais , Galinhas/genética , Células Germinativas/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo
3.
J Cell Physiol ; 238(3): 610-630, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36745473

RESUMO

Currently, studies have analyzed the formation mechanism of primordial germ cell (PGC) at the transcriptional level, but few at the protein level, which made the mechanism study of PGC formation not systematic. Here, we screened differential expression proteins (DEPs) regulated PGC formation by label-free proteomics with a novel sampling strategy of embryonic stem cells and PGC. Analysis of DEPs showed that multiple key events were involved, such as the transition from glycolysis to oxidative phosphorylation, activation of autophagy, low DNA methylation ensured the normal formation of PGC, beyond that, protein ubiquitination also played an important role in PGC formation. Importantly, the progression of such events was attributed to the inconsistency between transcription and translation. Interestingly, MAPK, PPAR, Wnt, and JAK signaling pathways not only interact with each other but also interact with different events to participate in the formation of PGC, which formed the PGC regulatory network. According to the regulatory network, the efficiency of PGC formation in induction system can be significantly improved. In conclusion, our results indicate that chicken PGC formation is a complex process involving multiple events and signals, which provide technical support for the specific application in PGC research.


Assuntos
Galinhas , Células Germinativas , Proteoma , Proteômica , Animais , Diferenciação Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Fosforilação Oxidativa , Glicólise , Autofagia , Ubiquitinação , Transdução de Sinais , Proteoma/análise , Proteoma/biossíntese , Proteoma/metabolismo
4.
J Cell Sci ; 134(3)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33443086

RESUMO

The unique developmental characteristics of chicken primordial germ cells (PGCs) enable them to be used in recovery of endangered bird species, gene editing and the generation of transgenic birds, but the limited number of PGCs greatly limits their application. Studies have shown that the formation of mammalian PGCs is induced by BMP4 signal, but the mechanism underlying chicken PGC formation has not been determined. Here, we confirmed that Wnt signaling activated via BMP4 activates transcription of Lin28A by inducing ß-catenin to compete with LSD1 for binding to TCF7L2, causing LSD1 to dissociate from the Lin28A promoter and enhancing H3K4me2 methylation in this region. Lin28A promotes PGC formation by inhibiting gga-let7a-3p maturation to initiate Blimp1 expression. Interestingly, expression of Blimp1 helped sustain Wnt5A expression by preventing LSD1 binding to the Wnt5A promoter. We thus elucidated a positive feedback pathway involving Wnt-Lin28A-Blimp1-Wnt that ensures PGC formation. In summary, our data provide new insight into the development of PGCs in chickens.


Assuntos
Galinhas , Via de Sinalização Wnt , Animais , Células Germinativas
5.
Mol Reprod Dev ; 90(5): 275-286, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36966461

RESUMO

Meiosis, a key step in spermatogenesis, is affected by many factors. Current studies have shown that long noncoding RNAs (lncRNAs) are potential factors regulating meiosis, and their regulatory mechanisms have received much attention. However, little research has been done on its regulatory mechanism in the spermatogenesis of roosters. Here, we found that lncRNA involved in meiosis and spermatogenesis (lncRNA-IMS) was involved in the regulation of Stra8 by gga-miR-31-5p and hindered the inhibition of Stra8 by gga-miR-31-5p. The acquisition and loss of function experiments demonstrated that lncRNA-IMS was involved in meiosis and spermatogenesis. In addition, we predicted and determined the core promoter region of lncRNA-IMS. Prediction of transcription factors, deletion/overexpression of binding sites, knockdown/overexpression of Jun, and dual-luciferase reporter analysis confirmed that Jun positively activated transcription of lncRNA-IMS. Our findings further enrich the TF-lncRNA-miRNA-mRNA regulatory network during male meiosis and provide new ideas for studying the molecular mechanism of meiosis and spermatogenesis in chicken spermatogonial stem cells.


Assuntos
Células-Tronco Germinativas Adultas , Proteínas Aviárias , Meiose , MicroRNAs , RNA Longo não Codificante , Animais , Masculino , Células-Tronco Germinativas Adultas/metabolismo , Galinhas/genética , Galinhas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Aviárias/metabolismo
6.
Anim Biotechnol ; 34(1): 56-66, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34153202

RESUMO

Mono-Sex culturing is an important methodology for intensive livestock and poultry production. Here, Hintw was identified as a potential key gene in sex-determination process in chickens via RNA-seq. Then we developed an effective method to interfere or overexpress Hintw in chicken embryos through the intravascular injection. QRT-PCR, ELISA and H&E staining were used to detect the effects of Hintw on gonadal development of chicken embryos. Results showed that Hintw exhibited a female-biased expression pattern in the early stage of PGCs (primordial germ cells) in embryonic gonads. The qRT-PCR analysis showed that Foxl2, Cyp19a1 in females were upregulated under the overexpression of Hintw, while Sox9 and Dmrt1 were downregulated Hintw. Overexpression of Hintw can promote the development of gonadal cortex, while interference with Hintw show the opposite result. Additionally, we found that overexpression of the Hintw in male chicken embryos could inhibit androgen levels and increase estrogen levels. On the other hand, interfering with Hintw in female chicken embryos decreased estrogen levels and increased androgen levels. In conclusion, this work sets the basis for the understanding of the molecular regulatory network for the sex-determination process in chicken embryos as well as providing the theoretical basis for mono-sex culturing of poultry.


Assuntos
Proteínas Aviárias , Galinhas , Processos de Determinação Sexual , Animais , Embrião de Galinha , Feminino , Masculino , Androgênios/metabolismo , Galinhas/genética , Estrogênios/metabolismo , Gônadas/metabolismo , Diferenciação Sexual , Proteínas Aviárias/metabolismo
7.
Int J Mol Sci ; 24(15)2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37569891

RESUMO

The role of gga-miR-31 in chicken germ cell differentiation and spermatogenesis is of significant importance. The transcriptional properties of gga-miR-31 are crucial in establishing the foundation for the formation of chicken spermatogonia stem cells and spermatogenesis. In this study, a series of recombinant vectors including varying lengths of the gga-miR-31 promoter were predicted and constructed. Through the utilization of the dual luciferase reporting system, the upstream -2180~0 bp region of gga-miR-31 was identified as its promoter region. Furthermore, it was predicted and confirmed that the activity of the gga-miR-31 promoter is increased by retinoic acid (RA). The binding of RA to the gga-miR-31 and Stra8 promoter regions was found to be competitive. Through the deletion of C-jun binding sites and the manipulation of C-jun expression levels, it was determined that C-jun inhibits the activity of the gga-miR-31 promoter. Furthermore, the combined treatment of C-jun and RA demonstrated that the positive regulatory effect of RA on the gga-miR-31 promoter is attenuated in the presence of high levels of C-jun. Overall, this study establishes a foundation for further investigation into the regulatory mechanisms of gga-miR-31 action, and provides a new avenue for inducing chicken embryonic stem cells (ESC) to differentiate into spermatogonial stem cells (SSC), and sperm formation.


Assuntos
MicroRNAs , Tretinoína , Embrião de Galinha , Animais , Masculino , Tretinoína/farmacologia , Galinhas/genética , Galinhas/metabolismo , Sêmen/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas
8.
J Cell Physiol ; 237(9): 3640-3650, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35790000

RESUMO

Detailed analysis of the regulatory mechanism of spermatogonia stem cell (SSCs) genesis can provide a novel strategy for the application of SSCs in the fields of transgenic animal production and regenerative medicine. Previous studies in this study showed that WNT signaling can positively regulate the formation of SSCs, but the exact regulatory mechanism is not clear. Here, we predicted the target gene of the Wnt/TCF7L2 pathway, namely TDRD1, by bioinformatics analysis. Functional studies revealed that overexpression of TDRD1 during RA-induced SSCs formation in vitro significantly upregulated the expression of reproductive marker genes (Integrinß1 and Integrinα6), and further flow cytometric analysis also confirmed that the formation efficiency of SSCs was significantly increased after overexpression of TDRD1; while interference with TDRD1 showed the exact opposite result. The in vivo experiments were consistent with the results of the in vitro experiments. Interestingly, although Wnt/TCF7L2 can promote the formation of SSCs, its function must be dependent on the expression of TDRD1, which was also repeatedly demonstrated as a target gene of the Wnt/TCF7L2 signaling pathway. Mechanistically, we found a large number of CpG sites in the TDRD1 promoter, and BSP analysis also confirmed that DNA methylation modifications in the TDRD1 promoter were significantly higher in embryonic stem cells than in SSCs, and further dual-luciferase reporter system assays revealed that low DNA methylation modification levels could enhance TDRD1 promoter activity; although previous studies demonstrated that TCF7L2 could enrich in the TDRD1 promoter region, the binding of the two was dependent on low DNA methylation modification. Taken together, we confirmed that low DNA methylation mediates Wnt/TCF7L2 regulation of TDRD1 to promote the formation of SSCs, providing a basis for SSCs in improving animal productivity.


Assuntos
Células-Tronco Germinativas Adultas , Via de Sinalização Wnt , Células-Tronco Germinativas Adultas/metabolismo , Animais , DNA/metabolismo , Metilação de DNA/genética , Células-Tronco Embrionárias/metabolismo , Masculino , Espermatogônias/metabolismo , Via de Sinalização Wnt/genética
9.
Mol Genet Genomics ; 297(3): 903-910, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35347417

RESUMO

Tudor domain containing 1 (TDRD1) is a member of the TDRD family and plays an important role in embryogenesis and gametogenesis. A detailed study of the characteristics of chicken TDRD1 can lay a foundation for the study of chicken spermatogonia stem cell formation and spermatogenesis. We cloned 2117 bp upstream fragment of TDRD1 promoter and constructed a series of recombinant vectors with different length deletions. The dual-luciferase experiments reveal that the upstream region of - 161 to 0 bp was its core transcription promoter region. Bioinformatics analysis predicted the possible binding of Transcription Factor 7 Like 2 (TCF7L2) and Zinc Finger E-Box-Binding Homeobox 1(ZEB1) transcription factors in the core region. The transcriptional activity of TDRD1 was significantly decreased after mutation of TCF7L2-binding site, while that of TDRD1 was significantly increased after mutation of ZEB1-binding site. Further, ChIP experiments verified that TCF7L2 enriched in the TDRD1 core transcriptional initiation region, suggesting that TCF7L2 and ZEB1 play an important role in the regulation of TDRD1. In summary, the region from - 161 to 0 bp is the core promoter region of TDRD1; ZEB1 and TCF7L2 bind to the TDRD1 promoter region and TCF7L2 activates the transcription of TDRD1 gene.


Assuntos
Proteínas de Ciclo Celular/genética , Galinhas , Fatores de Transcrição , Animais , Sítios de Ligação/genética , Galinhas/genética , Masculino , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
10.
J Cell Physiol ; 236(2): 1391-1400, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32749682

RESUMO

The development of primordial germ cells (PGCs) undergoes epigenetic modifications. The study of histone methylation in regulating PGCs is beneficial to understand the development and differentiation mechanism of germ stem cells. Notably, it provides a theoretical basis for directed induction and mass acquisition in vitro. However, little is known about the regulation of PGC formation by histone methylation. Here, we found the high enrichment of H3K4me2 in the blastoderm, genital ridges, and testis. Chromatin immunoprecipitation sequencing was performed and the results revealed that genomic H3K4me2 is dynamic in embryonic stem cells, PGCs, and spermatogonial stem cells. This trend was consistent with the H3K4me2 enrichment in the gene promoter region. Additionally, narrow region triggered PGC-related genes (Bmp4, Wnt5a, and Tcf7l2) and signaling pathways (Wnt and transforming growth factor-ß). After knocking down histone methylase Mll2 in vitro and vivo, the level of H3K4me2 decreased, inhibiting Cvh and Blimp1 expression, then repressing the formation of PGCs. Taken together, our study revealed the whole genome map of H3K4me2 in the formation of PGCs, contributing to improve the epigenetic study in PGC formation and providing materials for bird gene editing and rescue of endangered birds.


Assuntos
Proteína Morfogenética Óssea 4/genética , Epigênese Genética/genética , Histona Metiltransferases/genética , Testículo/crescimento & desenvolvimento , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Blastoderma/crescimento & desenvolvimento , Diferenciação Celular/genética , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Genitália/crescimento & desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Masculino , Transdução de Sinais/genética , Testículo/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Proteína Wnt-5a/genética
11.
J Cell Physiol ; 235(12): 9895-9909, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32458486

RESUMO

Long noncoding RNAs (lncRNAs) participate in the formation of primordial germ cells (PGCs); however, the identity of the key lncRNAs and the molecular mechanisms responsible for the formation of PGCs remain unknown. Here, we identify a key candidate lncRNA (lncRNA PGC transcript-1, LncPGCAT-1) via RNA sequencing of embryonic stem cells, PGCs, and Spermatogonial stem cells (SSCs). Functional experiments confirmed that LncPGCAT-1 positively regulated the formation of PGCs by elevating the expression of Cvh and C-kit while downregulating the pluripotency(Nanog) in vitro and in vivo; PAS staining of genital ridges in vivo also showed that interference with LncPGCAT-1 can significantly reduce the number of PGCs in genital ridges, while overexpression of LncPGCAT-1 had the opposite result. The result of luciferase reporter assay combined with CHIP-qPCR showed that the expression of LncPGCAT-1 was promoted by the transcription factor P53 and high levels of H3K4me2. Mechanistically, the luciferase reporter assay confirmed that mitogen-activated protein kinase 1 (MAPK1) was the target gene of LncPGCAT-1 and gga-mir-1591. In the ceRNA system, high levels of N6 methylation of LncPGCAT-1 enhanced the adsorption capacity of LncPGCAT-1 for gga-mir-1591. Adsorption of gga-mir-1591 activated the MAPK1/ERK signaling cascade by relieving the gga-mir-1591-dependent inhibition of MAPK1 expression. Moreover, LncPGCAT-1 interacted with interleukin enhancer binding factor 3 (ILF3) to regulate the ubiquitination of P53 and phosphorylation of JNK. Interaction with ILF3 resulted in positive self-feedback regulation of LncPGCAT-1 and activation of JNK signaling, ultimately promoting PGC formation. Altogether, the study expands our knowledge of the function and molecular mechanisms of lncRNAs in PGC development.


Assuntos
Células Germinativas/crescimento & desenvolvimento , Histonas/genética , RNA Longo não Codificante/genética , Espermatogônias/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/genética , Animais , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Ovos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Germinativas/metabolismo , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Análise de Sequência de RNA , Transdução de Sinais/genética
12.
J Cell Biochem ; 120(1): 332-342, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30277598

RESUMO

To better understand the mechanisms in transcriptional regulation, we analyzed the promoters of the reprogramming key genes Sox2, c-Myc, and Oct4. Here, we cloned different 5' deletions of the goat Sox2, c-Myc, and Oct4 promoters, and evaluated their functions by green fluorescent protein reporter system and dual-luciferase reporter system. Site-directed mugagenesis and epigenetic modifiers were used to explore the influence of transcription binding sites and epigenetic status on the promoters. The results suggested that the basal promoters were located in the - 109 to 49, - 147 to 1, and - 96 to 30 bp regions of the Sox2, c-Myc, and Oct4 promoters. The transcription factors that identified to influence the Sox2, c-Myc, and Oct4 promoter activities were Elf-1 and activating protein 2 (AP-2), C/EBP and Sp1, and Mzf1 and Sp1, respectively. The epigenetic alternation of the Sox2, c-Myc, and Oct4 promoters by 5-aza-2'-deoxycytidine or/and trichostatin A significantly increased the promoter activities. In conclusion, the result determined the core promoter areas of the Sox2, c-Myc, and Oct4 genes, and identified the transcription factors that influence their promoter activities. We also verified that the Sox2, c-Myc, and Oct4 promoters were hypermethylated and hypoacetylated.


Assuntos
Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/genética , Acetilação , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Metilação de DNA/genética , Deleção de Genes , Regulação da Expressão Gênica , Cabras/embriologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Camundongos , Microscopia de Fluorescência , Filogenia , Plasmídeos , Regiões Promotoras Genéticas , Vírus 40 dos Símios/genética , Ativação Transcricional , Transfecção
13.
J Cell Biochem ; 120(2): 1376-1385, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30447017

RESUMO

In recent years, considerable attention has been paid to chicken embryonic stem cells (ESCs) studies in relation to extensive applications in gene therapy and regenerative medicine. However, the approaches used are still immature. In this study, we showed that the chicken ESCs clones with a clear border can express alkaline phosphatase and marker proteins such as SSEA-1, SOX2, and OCT4 stably. In addition, culture medium containing 10 µmol/L of vitamin C (VC) could significantly promote the proliferation of ESCs cells. Moreover, ESCs transfected with p:enhanced green fluorescent protein (pEGFP)-hTERT could be subcultured more than tenth generations in culture medium containing exogenous factors (mLIF + bFGF + hSCF) and VC, and these ESCs clone could still be regenerated following cryopreservation. Quantitative real-time polymerase chain reaction results showed that there was no significant difference between SSEA-1, SOX2, and OCT4 expression during ESCs immortalization and that the tenth generation of ESCs was still able to express marker proteins SSEA-1, SOX2, and OCT4. Our results showed that an immobilized system for ESCs was established, and the ESCs were cultured in vitro maintaining their pluripotency.

14.
J Cell Biochem ; 120(5): 8021-8031, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30485515

RESUMO

The acetyl CoA acyltransferase 2 (ACAA2) is a key enzyme of the fatty acid oxidation pathway, catalyzing the last step of the mitochondrial beta oxidation, thus playing an important role in the fatty acid metabolism. The purpose of this study was to investigate the effect of knocking out ACAA2 on the expression of genes lipoprteinlipase (LPL), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase, fat mass and obesity-associated gene, adipocyte fatty acid-binding protein (AP2) in precursor adipocytes and their differentiation into adipocytes. The knockout vector was constructed using CRISPR-Cas RNA-guided nuclease technology with an efficiency of 23.80%, and the vector was transfected into precursor adipocyte cells, while an overexpression vector of the ACAA2 gene was also transfected in another group of preadipocytes. Quantitative polymerase chain reaction showed that the expression of the PPAR-γ, LPL, and AP2 was significantly lower in the knockout compared with the overexpression group, while there was no difference in cell growth. After induction of adipocyte precursor cells into adipocytes using dexamethasone, insulin, and IBMX, oil red staining showed a significantly different number of lipid droplets in the knockout group. These results provide a preliminary indication for a possible involvement of the ACAA2 gene in adipocyte differentiation in vitro.

15.
J Cell Biochem ; 120(2): 1794-1806, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30242885

RESUMO

Myostatin (MSTN) is an important gene involved in the regulation of embryonic muscle cells and adult muscle development; it has a good application prospect in transgenic animal production by improving the yield of muscle. The purpose of this study is to construct MSTN gene knockout vector using clustered regularly interspaced short palindromic repeats ( CRISPR)/CRISPR-associated protein 9 ( Cas9). The knockout efficiency was evaluated in sheep ear fibroblasts (SEFs) by cleavage activity of transcription of guide RNA ( gRNA), luciferase-single-strand annealing assay, T7 endonuclease I assay (T7E1), and TA clone sequence (10/38); and above all, detection showed that the cleavage activity of CRISPR/Cas9-mediated MSTN reached 29%. MSTN-Cas9/gRNA4 was transfected into sheep skeletal muscle satellite cell (sSMSC) to confirm the function of MSTN in myotomes formation induced by starvation in low-serum medium. The results showed that myotubes formation efficiency were 11.2 ± 1.3% and 19.5 ± 2.1% in the control group and knockout group, respectively. The average length of myotomes was 22 ± 5.3 and 47 ± 3.6 µm, displaying that MSTN knockout can promote sSMSC differentiation in number and length. The unlabeled MSTN-Cas9/gRNA4 was transfected into SEFs and monoclonal positive cells was obtained after 48 hours transfection. The MSTN-positive cells were used as donor cells to perform somatic cell nuclear transplantation to produce transgenic sheep. A total of 20 embryos were transplanted into surrogate mothers, four of them normally produce offspring. The genomic DNA of surviving lambs were used as a template, three positive individuals were identified by T7E1 digestion. All the results demonstrated that the CRISPR/Cas9 system has the potential to become an important and applicable gene engineering tool in animal breeding.

16.
J Cell Biochem ; 120(5): 6952-6961, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30656739

RESUMO

Recently, the surface marker genes of spermatogonial stem cells (SSCs) were increasingly excavated and verified. However, few studies focused on the key genes involved in the regulation of SSCs differentiation. Our laboratory has screened the Lbc gene (GenBank accession number: XM_429585.3), which is specifically expressed on the SSCs. The aim of this study is to investigate the function of Lbc and its regulatory mechanism for SSCs. The indirect immunofluorescence assay (IFA) showed that Lbc was located in both nucleus and cytoplasm. Lbc was also overexpressed and knocked out both in vitro and in vivo to verify its function in SSCs, respectively. As a result, the overexpressed Lbc could promote the formation of spermatogonial stem cells like cells (SSCs-like), while the deficiency of Lbc blocked the formation of SSCs-like. We also identified the core region of Lbc promoter that located into the upstream of the transcription initiation site -247 to -2bp. Moreover, the activity of Lbc promoter could be increased by histone acetylation which is leading to the higher expression of Lbc. When we mutated the transcription factor HOXA5 and SOX10 that bound to the core region of Lbc promoter, HOXA5 could reduce the transcription activity of Lbc whereas the SOX10 was not. Currently, we found Lbc is a new specific marker of SSCs. This gene can be modified by histone acetylated and promote the formation of chicken SSCs via the transcription factor HOXA5. The present research will lay the foundation for further study on the regulatory mechanism of SSCs.

17.
J Cell Biochem ; 119(2): 2240-2247, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28914467

RESUMO

This study aimed to clone the Stearoyl-CoA desaturase 1 (SCD1) gene derived from Xuhuai goat (Capra hircus), and analyze the sub-cellular localization in cells and tissues. The cDNA was cloned by reverse transcription polymerase chain reaction (RT-PCR). pEGFP-SCD1 vector was constructed to detect sub-cellular localization and tissue distribution. pEGFP-SCD1 was transfected into NIH-3T3 cells using polyethylene imine (PEI) and observed under fluorescence inversion microscope system 48 h after transfection. The expression level of SCD1 was detected by RT-PCR. Testicular injection was used to produce transgenic mice with goat SCD1 gene. DNA and protein were extracted from the tail tissue of F1 mice. The expression of exogenous gene in the F1 generation was detected in both DNA and protein. The results showed that the coding sequence (CDS) fragments of C. hircus SCD1 gene was 1074 bp and encodes 360 amino acids. RT-PCR results showed that SCD1 could be expressed successfully in NIH-3T3 cells in vitro. Sub-cellular localization analysis showed that pEGFP-SCD1 fusion protein located in the cytoplasm. It can be concluded that transgenic mice with goat SCD1 expressed in sperm and tail tissue was successfully produced in the F1 mice generation.


Assuntos
Clonagem Molecular/métodos , Cabras/metabolismo , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica , Cabras/genética , Masculino , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Espermatozoides/metabolismo , Cauda/metabolismo
18.
J Cell Biochem ; 119(2): 1548-1557, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28815778

RESUMO

The study aims to analyze the key signaling pathways in regulating the process of embryonic stem cells (ESCs) differentiation into spermatogonial stem cells (SSCs). We explored the specific regulating mechanisms of C-Jun amino-terminal kinase (JNK) signaling in this process. Interference/overexpression of MAPK8 allows the JNK signaling pathway to be blocked/activated. In Retinoic acid (RA) induced in vitro differentiation assays, the expression of germ cell marker genes, cvh, c-kit, integrin α6 and integrin ß1, was observed to upregulate while activating JNK signaling significantly. Fluorescence Activated Cell Sorting (FACs) analysis showed that the proportion of cvh+ and integrin α6+ cells in the overexpression group was significantly higher than which in the RA + shRNA-MAPK8 group. In in vivo situations, shRNA-MAPK8 could stably express in chicken embryos and significantly down-regulate expression of MAPK8 and downstream genes in JNK signaling pathway. With PAS stain, we found that PGCs (primordial germ cells) was significantly decreased after inhibiting MAPK8. With real-time quantitative PCR (qRT-PCR) and Western Blot, we identified that reproductive related genes expression was significantly suppressed after inhibiting MAPK8 in vivo. We preliminarily concluded that knockdown/ overexpression of MAPK8 could affect differentiation of ESC by inhibiting/activating JNK signal.


Assuntos
Biomarcadores/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Espermatogônias/citologia , Tretinoína/farmacologia , Animais , Diferenciação Celular , Embrião de Galinha , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Sistema de Sinalização das MAP Quinases , Masculino , Espermatogônias/metabolismo , Regulação para Cima
19.
J Cell Biochem ; 119(1): 1083-1092, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28695988

RESUMO

The CREPT (cell cycle-related and expression elevated protein in tumor, also known as RPRD1B) and p15RS (p15INK4b -related sequence, also known as RPRD1A) have been shown to regulate cell proliferation and alter the cell cycle through Wnt/ß-catenin pathway downstream genes in human. Although several studies have revealed the mechanism by which CREPT and p15RS regulate cell proliferation in human and mammals, it is still unclear how these genes function in poultry. In order to determine the function of CREPT and p15RS in chicken, we examined the expression of CREPT and p15RS in a variety of chicken tissues and DF-1 cells. Then, we determined the effect of overexpression or depletion of CREPT or p15RS, by transiently transfecting chicken DF-1 cells with overexpression and short hairpin RNA (shRNA) vectors respectively, on the regulation of cell proliferation. The results showed that CREPT and p15RS had different expression patterns and opposite effects on the cell cycling and proliferation. Knockdown of p15RS expression or overexpression of CREPT facilitated cell proliferation by promoting the cell-cycle transition from G0/G1 to S-phase and G2/M, whereas knockdown of CREPT or overexpression of p15RS inhibited cell proliferation. Mechanistically, CREPT and p15RS control DF-1 cell proliferation by regulating the expression of Wnt/ß-catenin pathway downstream regulatory genes, including ß-catenin, TCF4, and Cyclin D1. In conclusion, CREPT and p15RS regulate cell proliferation and the cell-cycle transition in chicken DF-1 cells by regulating the transcription of Wnt/ß-catenin pathway downstream regulatory genes.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transcrição Gênica , Via de Sinalização Wnt , Animais , Ciclo Celular , Proteínas de Ciclo Celular/química , Linhagem Celular , Proliferação de Células , Galinhas , Humanos , Modelos Moleculares , Mapas de Interação de Proteínas , Distribuição Tecidual
20.
J Cell Biochem ; 119(6): 4435-4446, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29143989

RESUMO

Nanos2 is an evolutionarily conserved RNA-binding protein containing 2 CCHC-type zinc finger motives. Here, we report that Nanos2 is strongly expressed in the testis compared to other tissues in chicken (Gallus gallus). Overexpression and knockout plasmid vectors were constructed, and in-vitro Cas9/gRNA digestion and T7 endonuclease I (T7E1) assay indicated that Nanos2-g1 possessed the highest knockout activity. In vitro and in vivo, Nanos2 overexpression accelerated the production of embryoid bodies (EBs) and SSC-like cells and promoted cvh, c-kit, and integrin α6 expression. Immunofluorescence staining, periodic acid schiff (PAS) and flow cytometry (FCM) assay showed that primordial germ cells (PGCs) and spermatogonial stem cells (SSCs) formation were significantly promoted. On the contrary, Nanos2 knockout delayed the production of EBs and SSC-like cells and correspondingly reduced cvh, c-kit, and integrin α6 expression. Simultaneously, the quantity of PGCs and SSCs was blocked. Collectively, these results uncovered a novel function of Nanos2 involved in chicken male germ cell differentiation, where it acts as a facilitator.


Assuntos
Proteínas Aviárias/metabolismo , Diferenciação Celular , Galinhas/metabolismo , Células-Tronco Embrionárias/metabolismo , Células Germinativas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas Aviárias/genética , Galinhas/genética , Células-Tronco Embrionárias/citologia , Técnicas de Inativação de Genes , Células Germinativas/citologia , Masculino , Proteínas de Ligação a RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA