Please wait a minute...
Frontiers of Agricultural Science and Engineering

ISSN 2095-7505

ISSN 2095-977X(Online)

CN 10-1204/S

Postal Subscription Code 80-906

Front. Agr. Sci. Eng.    2019, Vol. 6 Issue (1) : 81-92    https://doi.org/10.15302/J-FASE-2017180
RESEARCH ARTICLE
SALL4 maintains self-renewal of porcine pluripotent stem cells through downregulation of OTX2
Ning WANG, Sile WANG, Yaxian WANG, Yuanxing CAI, Fan YANG, Huayan WANG()
Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
 Download: PDF(5248 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Sall4 as one of the spalt family members contains several alternative splicing variants, which are differentially expressed and has a key role in maintaining pluripotent stem cells. However, the molecular features and function of SALL4 have not been well elucidated in porcine induced pluripotent stem cells (piPSCs). In this study, we identified SALL4 splice variants and found two SALL4 splicing variants through analysis of the porcine transcriptome data derived from piPSCs. SALL4A was only detected in piPSCs but SALL4B was globally expressed in porcine tissues and piPSCs. The level of SALL4B was significantly reduced when piPSCs differen-tiation occurred, however, the expression of SALL4A was not affected, indicating that SALL4B may be essential for the maintenance of piPSCs self-renewal. Overexpression of SALL4A and SALL4B in PEF cells could significantly stimulated expression of endogenous pluripotent genes, when SALL4B significantly promoted OCT4 expression. Conversely, SALL4A significantly promoted KLF4 expression. Additionally, both SALL4A and SALL4B could repress OTX2 promoter activity in a dose-dependent manner. Conversely, OTX2 also negatively regulated SALL4 expression. These observations indicate that a negative feedback regulatory mechanism may exist between SALL4 and OTX2, which is useful for the maintenance of the self-renewal of piPSCs.

Keywords OTX2      pluripotency      pig      SALL4      transcription regulation     
Corresponding Author(s): Huayan WANG   
Just Accepted Date: 22 November 2017   Online First Date: 26 December 2017    Issue Date: 25 February 2019
 Cite this article:   
Ning WANG,Sile WANG,Yaxian WANG, et al. SALL4 maintains self-renewal of porcine pluripotent stem cells through downregulation of OTX2[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 81-92.
 URL:  
https://academic.hep.com.cn/fase/EN/10.15302/J-FASE-2017180
https://academic.hep.com.cn/fase/EN/Y2019/V6/I1/81
Fig.1  Identification and cloning of porcine SALL4 alternative splicing variants. (a) Transcriptome analysis of SALL4 in porcine tissues and pluripotent cells. Sequencing reads for SALL4A and SALL4B in piPS-F cells are given in the green box; (b) RT-PCR analysis of SALL4 splicing variants in piPSCs; (c) enzyme digestions (BamHI/XhoI) to confirm the constructs of pSALL4A and pSALL4B; (d) western blot analysis of fusion proteins, EGFP-SALL4A (140 kDa) and EGFP-SALL4B (95 kDa), and EGFP (27 kDa) in 293T cells; (e) vectors of pSALL4A, pSALL4B, and pEGFP-C1 were transfected into 293T cells for 48 h. EGFP-SALL4 fusion proteins were translocated into nuclei.
Fig.2  SALL4 expression in porcine tissues and pluripotent cells. (a) Dynamic DNA methylation profile of porcine SALL4. MeDIP-Seq data of SALL4 methylation in porcine longissimusdorsi muscle (LDM) and piPSCs were visualized in UCSC genome browser; (b) bisulfite genomic sequencing analysis of SALL4 in LDM and piPS-F cells. Open and filled circles represent unmethylated and methylated CpGs; (c) RT-PCR (upper) and densitometry (lower) analyses of SALL4 expression in porcine tissues and piPSCs; (d) alkaline phosphatase staining (upper) and immunofluorescence staining (lower) of SALL4, SOX2, and OCT4 in undifferentiated and differentiated piPSCs. Scale bar, 50 mm.
Fig.3  SALL4 regulates the expression of pluripotent genes. Quantitative RT-PCR analyses were applied to determine the expression of pluripotent genes in piPSCs and PEF cells. (a) Expression of SALL4 and pluripotent genes in the differentiated piPSCs (piPS+RA) that were treated by retinoic acid (RA) for various time points; (b) expression of SALL4A and SALL4B in piPS+RA; (c) overexpression of SALL4A (OE-4A) and SALL4B (OE-4B) in PEF cells for 48 h. Ctrl, cells were transfected by pEGFP-C1; (d) knockdown (KD) SALL4 expression by siRNAs affected the expression of pluripotent genes. Ctrl, cells were transfected with an unspecific siRNA. Data are presented as mean±SD, * P<0.05, ** P<0.01, n = 3; (e) morphology of piPSCs with (KD) and without (Ctrl) siRNA treatment. Scale bar, 100 mm.
Fig.4  SALL4 suppresses OTX2 expression. (a) Luciferase assay of OTX2 promoter activity. The pG-OTX2 only (left) and pG-OTX2 with pSALL4A and pSALL4B (right) were cotransfected into 293T cells for 36 h. Ctrl cells were cotransfected by pG-OTX2 and pGL3-basic; (b) diagram of pG-OTX2 and the truncated OTX2 promoter constructs with potential transcription binding sites (upper). Luciferase assay of OTX2 promoter activity in 293T cells (lower). Ctrl, cells were transfected by pGL3-basic; (c) SALL4A and SALL4B regulate OTX2 promoter activation in 293T cells. Ctrl, cells were transfected by pEGFP-C1; (d) luciferase assays. For time-dependent assay (left), pG-OTX2 with pSALL4 and pSALL4B were cotransfected into 293T cells for 48 h. For dose-dependent assay (right), pG-OTX2 and different amount of SALL4 constructs were cotransfected into 293T cells for 36 h. Ctrl, cells were transfected by pGL3-OTX2; (e) alkaline phosphatase staining of overexpression of OTX2 (pE-OTX2, OTX2+ ) and suppression of OTX2 (OTX2+ plus pSALL4A and pSALL4B) in piPSCs. Ctrl, cells were transfected by pEGFP-C1. Number of AP positive clones was counted in 36 h post-transfection. Scale bar, 50 mm. Data are presented as mean±SD, * P<0.05; ** P<0.01; n = 3.
Fig.5  OTX2 regulates SALL4 expression. (a) Morphology and AP staining of piPSCs that were transfected with pE-OTX2 (OTX2+ ). Ctrl, cells without transfection of pE-OTX2. Scale bar, 50 µm; (b, c) semiquantitative (B) and quantitative (C) RT-PCR analyses of SALL4A and SALL4B in piPSCs transfected by pE-OTX2 (OTX2+ ). Ctrl, cells were transfected by pEGFP-C1; (d) western blot analysis of SALL4 expression in piPSCs that were treated by OTX2 siRNAs. Ctrl, cells were treated by an unspecific siRNA; (e) constructs (left) and luciferase assay (right) of the full and truncated SALL4 promoter; (f) luciferase assay of SALL4 promoter activity. The SALL4 constructs with pE-OTX2 were cotransfected into 293T cells, respectively, for 36 h. Ctrl, cells without transfection of pE-OTX2. Data are presented as mean±SD. * P<0.05; ** P<0.01; n = 3.
1 P JTesar, J G Chenoweth, F A Brook, T J Davies, E P Evans, D L Mack, R L Gardner, R D G McKay. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature, 2007, 448(7150): 196–199
https://doi.org/10.1038/nature05972 pmid: 17597760
2 TEzashi, B P V L Telugu, A P Alexenko, S Sachdev, SSinha, R MRoberts. Derivation of induced pluripotent stem cells from pig somatic cells. Proceedings of the National Academy of Sciences of the United States of America, 2009, 106(27): 10993–10998
https://doi.org/10.1073/pnas.0905284106 pmid: 19541600
3 ZWu, J Chen, JRen, LBao, J Liao, CCui, LRao, H Li, YGu, HDai, H Zhu, XTeng, LCheng, LXiao. Generation of pig induced pluripotent stem cells with a drug-inducible system. Journal of Molecular Cell Biology, 2009, 1(1): 46–54
https://doi.org/10.1093/jmcb/mjp003 pmid: 19502222
4 DCheng, Y Guo, ZLi, YLiu, X Gao, YGao, XCheng, JHu, H Wang. Porcine induced pluripotent stem cells require LIF and maintain their developmental potential in early stage of embryos. PLoS One, 2012, 7(12): e51778
https://doi.org/10.1371/journal.pone.0051778 pmid: 23251622
5 S HFujishiro, KNakano, YMizukami, TAzami, YArai, H Matsunari, RIshino, TNishimura, MWatanabe, TAbe, Y Furukawa, KUmeyama, SYamanaka, MEma, H Nagashima, YHanazono. Generation of naive-like porcine-induced pluripotent stem cells capable of contributing to embryonic and fetal development. Stem Cells and Development, 2013, 22(3): 473–482
https://doi.org/10.1089/scd.2012.0173 pmid: 22889279
6 Y HLoh, Q Wu, J LChew, V BVega, WZhang, XChen, G Bourque, JGeorge, BLeong, JLiu, K Y Wong, K W Sung, C W Lee, X D Zhao, K P Chiu, L Lipovich, V AKuznetsov, PRobson, L WStanton, C LWei, YRuan, B Lim, H HNg. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nature Genetics, 2006, 38(4): 431–440
https://doi.org/10.1038/ng1760 pmid: 16518401
7 M HTan, K F Au, D E Leong, K Foygel, W HWong, M W MYao. An Oct4-Sall4-Nanog network controls developmental progression in the pre-implantation mouse embryo. Molecular Systems Biology, 2013, 9(1): 632
https://doi.org/10.1038/msb.2012.65 pmid: 23295861
8 UElling, C Klasen, TEisenberger, KAnlag, MTreier. Murine inner cell mass-derived lineages depend on Sall4 function. Proceedings of the National Academy of Sciences of the United States of America, 2006, 103(44): 16319–16324
https://doi.org/10.1073/pnas.0607884103 pmid: 17060609
9 JYang, T R Corsello, Y Ma. Stem cell gene SALL4 suppresses transcription through recruitment of DNA methyltransferases. Journal of Biological Chemistry, 2012, 287(3): 1996–2005
https://doi.org/10.1074/jbc.M111.308734 pmid: 22128185
10 JYang, C Gao, LChai, YMa. A novel SALL4/OCT4 transcriptional feedback network for pluripotency of embryonic stem cells. PLoS One, 2010, 5(5): e10766
https://doi.org/10.1371/journal.pone.0010766 pmid: 20505821
11 JYang, L Chai, T CFowles, ZAlipio, DXu, L M Fink, D C Ward, Y Ma. Genome-wide analysis reveals Sall4 to be a major regulator of pluripotency in murine-embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America, 2008, 105(50): 19756–19761
https://doi.org/10.1073/pnas.0809321105 pmid: 19060217
12 C YLim, W L Tam, J Zhang, H SAng, HJia, L Lipovich, H HNg, C LWei, W KSung, PRobson, HYang, B Lim. Sall4 regulates distinct transcription circuitries in different blastocyst-derived stem cell lineages. Cell Stem Cell, 2008, 3(5): 543–554
https://doi.org/10.1016/j.stem.2008.08.004 pmid: 18804426
13 HTatetsu, N R Kong, G Chong, GAmabile, D GTenen, LChai. SALL4, the missing link between stem cells, development and cancer. Gene, 2016, 584(2): 111–119
https://doi.org/10.1016/j.gene.2016.02.019 pmid: 26892498
14 XChen, H Xu, PYuan, FFang, M Huss, V BVega, EWong, Y L Orlov, W Zhang, JJiang, Y HLoh, H CYeo, Z XYeo, VNarang, K RGovindarajan, BLeong, AShahab, YRuan, G Bourque, W KSung, N DClarke, C LWei, H HNg. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell, 2008, 133(6): 1106–1117
https://doi.org/10.1016/j.cell.2008.04.043 pmid: 18555785
15 QWu, X Chen, JZhang, Y HLoh, T YLow, WZhang, WZhang, S KSze, BLim, H H Ng. Sall4 interacts with Nanog and co-occupies Nanog genomic sites in embryonic stem cells. Journal of Biological Chemistry, 2006, 281(34): 24090–24094
https://doi.org/10.1074/jbc.C600122200 pmid: 16840789
16 JZhang, W L Tam, G Q Tong, Q Wu, H YChan, B SSoh, YLou, J Yang, YMa, LChai, H H Ng, T Lufkin, PRobson, BLim. Sall4 modulates embryonic stem cell pluripotency and early embryonic development by the transcriptional regulation of Pou5f1. Nature Cell Biology, 2006, 8(10): 1114–1123
https://doi.org/10.1038/ncb1481 pmid: 16980957
17 YBuganim, S Markoulaki, Nvan Wietmarschen, HHoke, TWu, K Ganz, BAkhtar-Zaidi, YHe, B J Abraham, D Porubsky, EKulenkampff, D AFaddah, LShi, Q Gao, SSarkar, MCohen, JGoldmann, J RNery, M DSchultz, J REcker, AXiao, R A Young, P M Lansdorp, R Jaenisch. The developmental potential of iPSCs is greatly influenced by reprogramming factor selection. Cell Stem Cell, 2014, 15(3): 295–309
https://doi.org/10.1016/j.stem.2014.07.003 pmid: 25192464
18 JNichols, B Zevnik, KAnastassiadis, HNiwa, D Klewe-Nebenius, IChambers, HSchöler, ASmith. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell, 1998, 95(3): 379–391
https://doi.org/10.1016/S0092-8674(00)81769-9 pmid: 9814708
19 F DWest, S L Terlouw, D J Kwon, J L Mumaw, S K Dhara, K Hasneen, J RDobrinsky, S LStice. Porcine induced pluripotent stem cells produce chimeric offspring. Stem Cells and Development, 2010, 19(8): 1211–1220
https://doi.org/10.1089/scd.2009.0458 pmid: 20380514
20 FYang, J Zhang, YLiu, DCheng, HWang. Structure and functional evaluation of porcine NANOG that is a single-exon gene and has two pseudogenes. International Journal of Biochemistry & Cell Biology, 2015, 59: 142–152
https://doi.org/10.1016/j.biocel.2014.12.009 pmid: 25542179
21 FYang, Y Ren, HLi, HWang. ESRRB plays a crucial role in the promotion of porcine cell reprograming. Journal of Cellular Physiology, 2018, 233(2): 1601–1611
https://doi.org/10.1002/jcp.26063 pmid: 28636277
22 NWang, Y Wang, YXie, H.WangOTX2 impedes self-renewal of porcine iPS cells through downregulation of NANOG expression. Cell Death and Discovery, 2016, 2: 16090
23 CChen, H Ai, JRen, WLi, P Li, RQiao, JOuyang, MYang, J Ma, LHuang. A global view of porcine transcriptome in three tissues from a full-sib pair with extreme phenotypes in growth and fat deposition by paired-end RNA sequencing. BMC Genomics, 2011, 12(1): 448
https://doi.org/10.1186/1471-2164-12-448 pmid: 21906321
24 SXiao, D Xie, XCao, PYu, X Xing, C CChen, MMusselman, MXie, F D West, H A Lewin, T Wang, SZhong. Comparative epigenomic annotation of regulatory DNA. Cell, 2012, 149(6): 1381–1392
https://doi.org/10.1016/j.cell.2012.04.029 pmid: 22682255
25 BLangmead, C Trapnell, MPop, S LSalzberg. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biology, 2009, 10(3): R25
https://doi.org/10.1186/gb-2009-10-3-r25 pmid: 19261174
26 YMa, W Cui, JYang, JQu, C Di, H MAmin, RLai, J Ritz, D SKrause, LChai. SALL4, a novel oncogene, is constitutively expressed in human acute myeloid leukemia (AML) and induces AML in transgenic mice. Blood, 2006, 108(8): 2726–2735
https://doi.org/10.1182/blood-2006-02-001594 pmid: 16763212
27 KXu, X Chen, HYang, YXu, Y He, CWang, HHuang, BLiu, W Liu, JLi, XKou, Y Zhao, KZhao, LZhang, ZHou, H Wang, HWang, JLi, H Fan, FWang, YGao, Y Zhang, JChen, SGao. Maternal Sall4 Is indispensable for epigenetic maturation of mouse oocytes. Journal of Biological Chemistry, 2017, 292(5): 1798–1807
https://doi.org/10.1074/jbc.M116.767061 pmid: 28031467
28 SZhang, Y Guo, YCui, YLiu, T Yu, HWang. Generation of intermediate porcine iPS cells under culture condition favorable for mesenchymal-to-epithelial transition. Stem Cell Reviews and Reports, 2015, 11(1): 24–38
https://doi.org/10.1007/s12015-014-9552-x pmid: 25134796
29 K BLarsen, M Lutterodt, M FRath, MMøller. Expression of the homeobox genes PAX6, OTX2, and OTX1 in the early human fetal retina. International Journal of Developmental Neuroscience, 2009, 27(5): 485–492
https://doi.org/10.1016/j.ijdevneu.2009.04.004 pmid: 19414065
30 ASimeone, D Acampora, AMallamaci, AStornaiuolo, M RD’Apice, VNigro, EBoncinelli. A vertebrate gene related to orthodenticle contains a homeodomain of the bicoid class and demarcates anterior neuroectoderm in the gastrulating mouse embryo. EMBO Journal, 1993, 12(7): 2735–2747
pmid: 8101484
31 DDiaczok, C Romero, JZunich, IMarshall, SRadovick. A novel dominant negative mutation of OTX2 associated with combined pituitary hormone deficiency. Journal of Clinical Endocrinology and Metabolism, 2008, 93(11): 4351–4359
https://doi.org/10.1210/jc.2008-1189 pmid: 18728160
32 M HAsadi, K Khalifeh, S JMowla. OCT4 spliced variants are highly expressed in brain cancer tissues and inhibition of OCT4B1 causes G2/M arrest in brain cancer cells. Journal of Neuro-Oncology, 2016, 130(3): 455–463
https://doi.org/10.1007/s11060-016-2255-1 pmid: 27585657
33 J YHwang, J N Oh, D K Lee, K H Choi, C H Park, C K Lee. Identification and differential expression patterns of porcine OCT4 variants. Reproduction, 2015, 149(1): 55–66
https://doi.org/10.1530/REP-14-0403 pmid: 25342174
34 DLi, Z K Yang, J Y Bu, C Y Xu, H Sun, J BTang, PLin, W Cheng, NHuang, R JCui, X GYu, X LZheng. OCT4B modulates OCT4A expression as ceRNA in tumor cells. Oncology Reports, 2015, 33(5): 2622–2630
https://doi.org/10.3892/or.2015.3862 pmid: 25812694
35 M HTan, K F Au, D E Leong, K Foygel, W HWong, M WYao. An Oct4-Sall4-Nanog network controls developmental progression in the pre-implantation mouse embryo. Molecular Systems Biology, 2013, 9(1): 632
https://doi.org/10.1038/msb.2012.65 pmid: 23295861
36 L ABoyer, T I Lee, M F Cole, S E Johnstone, S S Levine, J P Zucker, M G Guenther, R M Kumar, H L Murray, R G Jenner, D K Gifford, D A Melton, R Jaenisch, R AYoung. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 2005, 122(6): 947–956
https://doi.org/10.1016/j.cell.2005.08.020 pmid: 16153702
37 XChen, H Xu, PYuan, FFang, M Huss, V BVega, EWong, Y L Orlov, W Zhang, JJiang, Y HLoh, H CYeo, Z XYeo, VNarang, K RGovindarajan, BLeong, AShahab, YRuan, G Bourque, W KSung, N DClarke, C LWei, H HNg. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell, 2008, 133(6): 1106–1117
https://doi.org/10.1016/j.cell.2008.04.043 pmid: 18555785
38 C YLim, W L Tam, J Zhang, H SAng, HJia, L Lipovich, H HNg, C LWei, W KSung, PRobson, HYang, B Lim. Sall4 regulates distinct transcription circuitries in different blastocyst-derived stem cell lineages. Cell Stem Cell, 2008, 3(5): 543–554
https://doi.org/10.1016/j.stem.2008.08.004 pmid: 18804426
39
40 SRao, S Zhen, SRoumiantsev, L TMcDonald, G CYuan, S HOrkin. Differential roles of Sall4 isoforms in embryonic stem cell pluripotency. Molecular and Cellular Biology, 2010, 30(22): 5364–5380
https://doi.org/10.1128/MCB.00419-10 pmid: 20837710
41 DAcampora, L G Di Giovannantonio, A Simeone. Otx2 is an intrinsic determinant of the embryonic stem cell state and is required for transition to a stable epiblast stem cell condition. Development, 2013, 140(1): 43–55
https://doi.org/10.1242/dev.085290 pmid: 23154415
42 CBuecker, R Srinivasan, ZWu, ECalo, D Acampora, TFaial, ASimeone, MTan, T Swigut, JWysocka. Reorganization of enhancer patterns in transition from naive to primed pluripotency. Cell Stem Cell, 2014, 14(6): 838–853
https://doi.org/10.1016/j.stem.2014.04.003 pmid: 24905168
43 S HYang, T Kalkan, CMorissroe, HMarks, HStunnenberg, ASmith, A DSharrocks. Otx2 and Oct4 drive early enhancer activation during embryonic stem cell transition from naive pluripotency. Cell Reports, 2014, 7(6): 1968–1981
https://doi.org/10.1016/j.celrep.2014.05.037 pmid: 24931607
[1] Ruigao SONG, Yu WANG, Yanfang WANG, Jianguo ZHAO. Base editing in pigs for precision breeding[J]. Front. Agr. Sci. Eng. , 2020, 7(2): 161-170.
[2] Chris PROUDFOOT, Gus MCFARLANE, Bruce WHITELAW, Simon LILLICO. Livestock breeding for the 21st century: the promise of the editing revolution[J]. Front. Agr. Sci. Eng. , 2020, 7(2): 129-135.
[3] Yong JIN, Manling ZHANG, Xinrong JU, Shuang LIANG, Qiang XIONG, Lihua ZHAO, Xiaowei NIE, Daorong HOU, Qiang LIU, Junzheng WANG, Chenyu WANG, Xiaokang LI, Lining ZHANG, Xiaorui LIU, Ying WANG, Haiyuan YANG, Yifan DAI, Rongfeng LI. Factors influencing the somatic cell nuclear transfer efficiency in pigs[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 73-80.
[4] Jianwen CHEN, Kaiyuan PAN, Zhen CHEN, Biao DING, Dandan SONG, Wenbin BAO, Yunhai ZHANG. Construction of multiple shRNA vectors targeting PEDV and TGEV and production of transgenic SCNT porcine embryos in vitro[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 66-72.
[5] Chengcheng ZHAO, Junsong SHI, Rong ZHOU, Ranbiao MAI, Lvhua LUO, Xiaoyan HE, Hongmei JI, Gengyuan CAI, Dewu LIU, Enqin ZHENG, Zhenfang WU, Zicong LI. Effects of enucleation method on in vitro and in vivo development rate of cloned pig embryos[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 61-65.
[6] Zheng AO, Chengfa ZHAO, Yanmin GAN, Xiao WU, Junsong SHI, Enqin ZHENG, Dewu LIU, Gengyuan CAI, Zhenfang WU, Zicong LI. Comparison of birth weight and umbilical and placental characteristics of cloned and artificial insemination-derived piglets[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 54-60.
[7] Jianyong HAN, Yi-Liang MIAO, Jinlian HUA, Yan LI, Xue ZHANG, Jilong ZHOU, Na LI, Ying ZHANG, Jinying ZHANG, Zhonghua LIU. Porcine pluripotent stem cells: progress, challenges and prospects[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 8-27.
[8] Qijing LEI, Qin PAN, Shuai YU, Na LI, Shulin CHEN, Kuldip SIDHU, Jinlian HUA. Reprogramming of the pig primordial germ cells into pluripotent stem cells: a brief review[J]. Front. Agr. Sci. Eng. , 2019, 6(1): 28-32.
[9] Lei CHEN, Shilin TIAN, Long JIN, Zongyi GUO, Dan ZHU, Lan JING, Tiandong CHE, Qianzi TANG, Siqing CHEN, Liang ZHANG, Tinghuan ZHANG, Zuohua LIU, Jinyong WANG, Mingzhou LI. Genome-wide analysis reveals selection for Chinese Rongchang pigs[J]. Front. Agr. Sci. Eng. , 2017, 4(3): 319-326.
[10] Haiyan WANG, Qiaoxia ZHANG, Lilin YIN, Xiangdong LIU, Shuhong ZHAO, Mengjin ZHU, Changchun LI. Transcriptomic basis of neutrophil ratio variation induced by poly I:C stimulation in porcine peripheral blood[J]. Front. Agr. Sci. Eng. , 2017, 4(3): 342-352.
[11] Longchao ZHANG, Jingwei YUE, Xin LIU, Jing LIANG, Kebin ZHAO, Hua YAN, Na LI, Lei PU, Yuebo ZHANG, Huibi SHI, Ligang WANG, Lixian WANG. Genome-wide search for candidate genes determining vertebrae number in pigs[J]. Front. Agr. Sci. Eng. , 2017, 4(3): 327-334.
[12] Xingwang WANG, Rongrong DING, Jianping QUAN, Linxue YANG, Ming YANG, Enqin ZHENG, Dewu LIU, Gengyuan CAI, Zhenfang WU, Jie YANG. Genome-wide association analysis reveals genetic loci and candidate genes associated with intramuscular fat in Duroc pigs[J]. Front. Agr. Sci. Eng. , 2017, 4(3): 335-341.
[13] Wen LUO, Bahareldin A. ABDALLA, Qinghua NIE, Xiquan ZHANG. The genetic regulation of skeletal muscle development: insights from chicken studies[J]. Front. Agr. Sci. Eng. , 2017, 4(3): 295-304.
[14] Shaohua WANG,Kun ZHANG,Yunping DAI. Advances in genetic engineering of domestic animals[J]. Front. Agr. Sci. Eng. , 2016, 3(1): 1-10.
[15] Weiya ZHANG,Wei WEI,Yuanyuan ZHAO,Shuhong ZHAO,Xinyun LI. The microRNA, miR-29c, participates in muscle development through targeting the YY1 gene and is associated with postmortem muscle pH in pigs[J]. Front. Agr. Sci. Eng. , 2015, 2(4): 311-317.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed