Please wait a minute...
Protein & Cell

ISSN 1674-800X

ISSN 1674-8018(Online)

CN 11-5886/Q

Postal Subscription Code 80-984

2018 Impact Factor: 7.575

Protein Cell    2019, Vol. 10 Issue (5) : 347-364    https://doi.org/10.1007/s13238-018-0603-y
RESEARCH ARTICLE
Loss-of-function of sox3 causes follicle development retardation and reduces fecundity in zebrafish
Qiang Hong, Cong Li, Ruhong Ying, Heming Lin, Jingqiu Li, Yu Zhao, Hanhua Cheng(), Rongjia Zhou()
Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
 Download: PDF(6439 KB)  
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Folliculogenesis is essential for production of female gametes in vertebrates. However, the molecular mechanisms underlying follicle development, particularly apoptosis regulation in ovary, remain elusive. Here, we generated sox3 knockout zebrafish lines using CRISPR/Cas9. sox3 knockout led to follicle development retardation and a reduced fecundity in females. Comparative analysis of transcriptome between sox3−/− and wild-type ovaries revealed that Sox3 was involved in pathways of ovarian steroidogenesis and apoptosis. Knockout of sox3 promoted follicle apoptosis and obvious apoptosis signals were detected in somatic cells of stages III and IV follicles of sox3−/− ovaries. Moreover, Sox3 can bind to and activate the promoter of cyp19a1a. Up-regulation of Cyp19a1a expression promoted 17β-estradiol synthesis, which inhibited apoptosis in follicle development. Thus, Sox3 functions as a regulator of Cyp19a1a expression, via 17β-E2 linking apoptosis suppression, which is implicated in improving female fecundity.

Keywords Sox3      follicle development      apoptosis      Cyp19a1a      zebrafish     
Corresponding Author(s): Hanhua Cheng,Rongjia Zhou   
Issue Date: 14 May 2019
 Cite this article:   
Qiang Hong,Cong Li,Ruhong Ying, et al. Loss-of-function of sox3 causes follicle development retardation and reduces fecundity in zebrafish[J]. Protein Cell, 2019, 10(5): 347-364.
 URL:  
https://academic.hep.com.cn/pac/EN/10.1007/s13238-018-0603-y
https://academic.hep.com.cn/pac/EN/Y2019/V10/I5/347
1 KEBaker, RParker (2004) Nonsense-mediated mRNA decay: terminating erroneous gene expression. Curr Opin Cell Biol 16:293–299
https://doi.org/10.1016/j.ceb.2004.03.003
2 MBauters, SGFrints, HVan Esch, LSpruijt, MMBaldewijns, CEMde Die-Smulders, JPFryns, PMarynen, GFroyen (2014) Evidence for increased SOX3 dosage as a risk factor for X-linked hypopituitarism and neural tube defects. Am J Med Genet Part A 164:1947–1952
https://doi.org/10.1002/ajmg.a.36580
3 FBrion, CRTyler, XPalazzi, BLaillet, JMPorcher, JGarric, PFlammarion (2004) Impacts of 17beta-estradiol, including environmentally relevant concentrations, on reproduction after exposure during embryo-larval-, juvenile- and adult-life stages in zebrafish (Danio rerio). Aquat Toxicol 68:193–217
https://doi.org/10.1016/j.aquatox.2004.01.022
4 MBylund, EAndersson, BGNovitch, JMuhr (2003) Vertebrate neurogenesis is counteracted by Sox1-3 activity. Nat Neurosci 6:1162–1168
https://doi.org/10.1038/nn1131
5 SChen, HZhang, FWang, WZhang, GPeng (2016) nr0b1 (DAX1) mutation in zebrafish causes female-to-male sex reversal through abnormal gonadal proliferation and differentiation. Mol Cell Endocrinol 433:105–116
https://doi.org/10.1016/j.mce.2016.06.005
6 CMCrowder, CSLassiter, DAGorelick (2018) Nuclear androgen receptor regulates testes organization and oocyte maturation in zebrafish. Endocrinology 159:980–993
https://doi.org/10.1210/en.2017-00617
7 CTDee, CSHirst, YHShih, VBTripathi, RKPatient, PJScotting (2008) Sox3 regulates both neural fate and differentiation in the zebrafish ectoderm. Dev Biol 320:289–301
https://doi.org/10.1016/j.ydbio.2008.05.542
8 DBDranow, KHu, AMBird, STLawry, MTAdams, ASanchez, JFAmatruda, BWDraper (2016) Bmp15 is an oocyte-produced signal required for maintenance of the adult female sexual phenotype in zebrafish. PLoS Genet 12:e1006323
https://doi.org/10.1371/journal.pgen.1006323
9 JJEppig (2001) Oocyte control of ovarian follicular development and function in mammals. Reproduction 122:829–838
https://doi.org/10.1530/rep.0.1220829
10 NFacchinello, TSkobo, GMeneghetti, EColletti, ADinarello, NTiso, RCosta, GGioacchini, OCarnevali, FArgentonet al. (2017) nr3c1 null mutant zebrafish are viable and reveal DNA-binding-independent activities of the glucocorticoid receptor. Sci Rep 7:4371
https://doi.org/10.1038/s41598-017-04535-6
11 JWFoster, JAMGraves (1994) An Sry-related sequence on the marsupial X-chromosome- implications for the evolution of the mammalian testis determining gene. Proc Natl Acad Sci USA 91:1927–1931
https://doi.org/10.1073/pnas.91.5.1927
12 XZFu, YBCheng, JYuan, CHHuang, HHCheng, RJZhou (2015) Loss-of-function mutation in the X-linked TBX22 promoter disrupts an ETS-1 binding site and leads to cleft palate. Hum Genet 134:147–158
https://doi.org/10.1007/s00439-014-1503-8
13 YGou, JGuo, KMaulding, BBRiley (2018a) Sox2 and Sox3 cooperate to regulate otic/epibranchial placode induction in zebrafish. Dev Biol 435:84–95
https://doi.org/10.1016/j.ydbio.2018.01.011
14 YGou, SVemaraju, EMSweet, HJKwon, BBRiley (2018b) Sox2 and Sox3 play unique roles in development of hair cells and neurons in the zebrafish inner ear. Dev Biol 435:73–83
https://doi.org/10.1016/j.ydbio.2018.01.010
15 YQGuo, HHCheng, XHuang, SGao, HSYu, RJZhou (2005) Gene structure, multiple alternative splicing, and expression in gonads of zebrafish Dmrt1. Biochem Bioph Res Co 330:950–957
https://doi.org/10.1016/j.bbrc.2005.03.066
16 BHaines, JHughes, MCorbett, MShaw, JInnes, LPatel, JGecz, JClayton-Smith, PThomas (2015) Interchromosomal insertional translocation at Xq26.3 alters SOX3 expression in an individual with XX male sex reversal. J Clin Endocr Metab 100:E815–E820
https://doi.org/10.1210/jc.2014-4383
17 YHou, JYuan, XZhou, XZFu, HHCheng, RJZhou (2012) DNA demethylation and USF regulate the meiosis-specific expression of the mouse miwi. PLoS Genet 8:e1002716
https://doi.org/10.1371/journal.pgen.1002716
18 SYHsu, RJLai, MFinegold, AJHsueh (1996) Targeted overexpression of Bcl-2 in ovaries of transgenic mice leads to decreased follicle apoptosis, enhanced folliculogenesis, and increased germ cell tumorigenesis. Endocrinology 137:4837–4843
https://doi.org/10.1210/endo.137.11.8895354
19 KJHutt (2015) The role of BH3-only proteins in apoptosis within the ovary. Reproduction 149:R81–R89
https://doi.org/10.1530/REP-14-0422
20 AMJelsig, BRDiness, SKreiborg, KMMain, VALarsen, HHove (2018) A complex phenotype in a family with a pathogenic SOX3 missense variant. Eur J Med Genet 61:168–172
https://doi.org/10.1016/j.ejmg.2017.11.012
21 SRJeng, GCWu, WSYueh, SFKuo, SDufour, CFChang (2018) Gonadal development and expression of sex-specific genes during sex differentiation in the Japanese eel. Gen Comp Endocrinol 257:74–85
https://doi.org/10.1016/j.ygcen.2017.07.031
22 RLJones, MEPepling (2013) Role of the antiapoptotic proteins BCL2 and MCL1 in the neonatal mouse ovary. Biol Reprod 88:1–8
https://doi.org/10.1095/biolreprod.112.103028
23 MKanehisa, MAraki, SGoto, MHattori, MHirakawa, MItoh, TKatayama, SKawashima, SOkuda, TTokimatsuet al. (2008) KEGG for linking genomes to life and the environment. Nucleic Acids Res 36:D480–D484
https://doi.org/10.1093/nar/gkm882
24 DKim, BLandmead, SLSalzberg (2015) HISAT: a fast spliced aligner with low memory requirements. Nat Methods 12:357–360
https://doi.org/10.1038/nmeth.3317
25 TKitano, KTakamune, YNagahama, SIAbe (2000) Aromatase inhibitor and 17alpha-methyltestosterone cause sex-reversal from genetical females to phenotypic males and suppression of P450 aromatase gene expression in Japanese flounder (Paralichthys olivaceus). Mol Reprod Dev 56:1–5
https://doi.org/10.1002/(SICI)1098-2795(200005)56:1<1::AID-MRD1>3.0.CO;2-3
26 TKobayashi, HKajiura-Kobayashi, YNagahama (2003) Induction of XY sex reversal by estrogen involves altered gene expression in a teleost, tilapia. Cytogenet Genome Res 101:289–294
https://doi.org/10.1159/000074351
27 PKoopman, JGubbay, NVivian, PGoodfellow, RLovell-Badge (1991) Male development of chromosomally female mice transgenic for Sry. Nature 351:117–121
https://doi.org/10.1038/351117a0
28 BLangmead, CTrapnell, MPop, SLSalzberg (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10:R25
https://doi.org/10.1186/gb-2009-10-3-r25
29 ESWLau, ZWZhang, MMQin, WGe (2016) Knockout of zebrafish ovarian aromatase gene (cyp19a1a) by TALEN and CRISPR/Cas9 leads to all-male offspring due to failed ovarian differentiation. Sci Rep 6:37357
https://doi.org/10.1038/srep37357
30 FLaumonnier, NRonce, BCJHamel, PThomas, JLespinasse, MRaynaud, CParingaux, Hvan Bokhoven, VKalscheuer, JPFrynset al. (2002) Transcription factor SOX3 is involved in X-linked mental retardation with growth hormone deficiency. Am J Hum Genet 71:1450–1455
https://doi.org/10.1086/344661
31 DMLeerberg, KSano, BWDraper (2017) Fibroblast growth factor signaling is required for early somatic gonad development in zebrafish. PLoS Genet 13:e1006993
https://doi.org/10.1371/journal.pgen.1006993
32 BLi, CNDewey (2011) RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform 12:323
https://doi.org/10.1186/1471-2105-12-323
33 EBLi, DTruong, SAHallett, KMukherjee, BCSchutte, ECLiao (2017) Rapid functional analysis of computationally complex rare human IRF6 gene variants using a novel zebrafish model. PLoS Genet 13:e1007009
https://doi.org/10.1371/journal.pgen.1007009
34 WCLiew, RBartfai, ZLim, RSreenivasan, KRSiegfried, LOrban (2012) Polygenic sex determination system in zebrafish. PLoS ONE 7:e34397
https://doi.org/10.1371/journal.pone.0034397
35 QLin, JMei, ZLi, XZhang, LZhou, JFGui (2017) Distinct and cooperative roles of amh and dmrt1 in self-renewal and differentiation of male germ cells in zebrafish. Genetics 207:1007–1022
https://doi.org/10.1534/genetics.117.300274
36 DLiu, ZWang, AXiao, YZhang, WLi, YZu, SYao, SLin, BZhang (2014a) Efficient gene targeting in zebrafish mediated by a zebrafish-codon-optimized Cas9 and evaluation of off-targeting effect. J Genet Genom 41:43–46
https://doi.org/10.1016/j.jgg.2013.11.004
37 JLiu, WYao, YYao, XDu, JZhou, BMa, HLiu, QLi, ZPan (2014b) MiR-92a inhibits porcine ovarian granulosa cell apoptosis by targeting Smad7 gene. FEBS Lett 588:4497–4503
https://doi.org/10.1016/j.febslet.2014.10.021
38 FMatsuda, NInoue, NManabe, SOhkura (2012) Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J Reprod Dev 58:44–50
https://doi.org/10.1262/jrd.2011-012
39 MDMichael, MWKilgore, KMorohashi, ERSimpson (1995) Ad4BP/SF-1 regulates cyclic AMP-induced transcription from the proximal promoter (PII) of the human aromatase P450 (CYP19) gene in the ovary. J Biol Chem 270:13561–13566
https://doi.org/10.1074/jbc.270.22.13561
40 WLMiller (2017) Steroidogenesis: unanswered questions. Trends Endocrinol Metab 28:771–793
https://doi.org/10.1016/j.tem.2017.09.002
41 CMiura, THigashino, TMiura (2007) A progestin and an estrogen regulate early stages of oogenesis in fish. Biol Reprod 77:822–828
https://doi.org/10.1095/biolreprod.107.061408
42 SMoalem, RBabul-Hirji, DJStavropolous, DWherrett, DJBagli, PThomas, DChitayat (2012) XX male sex reversal with genital abnormalities associated with a de novo SOX3 gene duplication. Am J Med Genet Part A 158A:1759–1764
https://doi.org/10.1002/ajmg.a.35390
43 YNagahama (1997) 17 alpha,20 beta-dihydroxy-4-pregnen-3-one, a maturation-inducing hormone in fish oocytes: mechanisms of synthesis and action. Steroids 62:190–196
https://doi.org/10.1016/S0039-128X(96)00180-8
44 YOkuda, EOgura, HKondoh, YKamachi (2010) B1 SOX coordinate cell specification with patterning and morphogenesis in the early zebrafish embryo. PLoS Genet 6:e1000936
https://doi.org/10.1371/journal.pgen.1000936
45 LOrban, RSreenivasan, PEOlsson (2009) Long and winding roads: testis differentiation in zebrafish. Mol Cell Endocrinol 312:35–41
https://doi.org/10.1016/j.mce.2009.04.014
46 YOshima, KNaruse, YNakamura, MNakamura (2009) Sox3: a transcription factor for Cyp19 expression in the frog Rana rugosa. Gene 445:38–48
https://doi.org/10.1016/j.gene.2009.05.011
47 MPannetier, SFabre, FBatista, AKocer, LRenault, GJolivet, BMandon-Pepin, CCotinot, RVeitia, EPailhoux (2006) FOXL2 activates P450 aromatase gene transcription: towards a better characterization of the early steps of mammalian ovarian development. J Mol Endocrinol 36:399–413
https://doi.org/10.1677/jme.1.01947
48 DPennisi, JBowles, ANagy, GMuscat, PKoopman (2000a) Mice null for sox18 are viable and display a mild coat defect. Mol Cell Biol 20:9331–9336
https://doi.org/10.1128/MCB.20.24.9331-9336.2000
49 DPennisi, JGardner, DChambers, BHosking, JPeters, GMuscat, CAbbott, PKoopman (2000b) Mutations in Sox18 underlie cardiovascular and hair follicle defects in ragged mice. Nat Genet 24:434–437
https://doi.org/10.1038/74301
50 GIPerez, RRobles, CMKnudson, JAFlaws, SJKorsmeyer, JLTilly (1999) Prolongation of ovarian lifespan into advanced chronological age by Bax-deficiency. Nat Genet 21:200–203
https://doi.org/10.1038/5985
51 MJPeters, SKParker, JGrim, CAHAllard, JLevin, HWDetrich3rd (2018) Divergent Hemogen genes of teleosts and mammals share conserved roles in erythropoiesis: analysis using transgenic and mutant zebrafish. Biol Open 7:bio035576
https://doi.org/10.1242/bio.035576
52 MWPopp, LEMaquat (2016) Leveraging rules of nonsensemediated mRNA decay for genome engineering and personalized medicine. Cell 165:1319–1322
https://doi.org/10.1016/j.cell.2016.05.053
53 MQiu, JLiu, CHan, BWu, ZYang, FSu, FQuan, YZhang (2014) The influence of ovarian stromal/theca cells during in vitro culture on steroidogenesis, proliferation and apoptosis of granulosa cells derived from the goat ovary. Reprod Domest Anim 49:170–176
https://doi.org/10.1111/rda.12256
54 SMQuirk, RGCowan, RMHarman (2006) The susceptibility of granulosa cells to apoptosis is influenced by oestradiol and the cell cycle. J Endocrinol 189:441–453
https://doi.org/10.1677/joe.1.06549
55 VSRatts, JAFlaws, RKolp, CMSorenson, JLTilly (1995) Ablation of bcl-2 gene expression decreases the numbers of oocytes and primordial follicles established in the post-natal female mouse gonad. Endocrinology 136:3665–3668
https://doi.org/10.1210/endo.136.8.7628407
56 SLPRegan, PGKnight, JLYovich, YLeung, FArfuso, ADharmarajan (2018) Granulosa cell apoptosis in the ovarian follicle-a changing view. Front Endocrinol 9:Article 61
https://doi.org/10.3389/fendo.2018.00061
57 KRizzoti, SBrunelli, DCarmignac, PQThomas, ICRobinson, RLovell-Badge (2004) SOX3 is required during the formation of the hypothalamo-pituitary axis. Nat Genet 36:247–255
https://doi.org/10.1038/ng1309
58 SHSadraie, HSaito, TKaneko, TSaito, MHiroi (2000) Effects of aging on ovarian fecundity in terms of the incidence of apoptotic granulosa cells. J Assist Reprod Genet 17:168–173
https://doi.org/10.1023/A:1009422323306
59 JMSantos, VGMenezes, RSBarberino, TJMacedo, TLLins, BBGouveia, VRBarros, LPSantos, RJGoncalves, MHMatos (2014) Immunohistochemical localization of fibroblast growth factor-2 in the sheep ovary and its effects on pre-antral follicle apoptosis and development in vitro. Reprod Domest Anim 49:522–528
https://doi.org/10.1111/rda.12322
60 MShen, ZLiu, BLi, YTeng, JZhang, YTang, SCSun, HLiu (2014) Involvement of FoxO1 in the effects of follicle-stimulating hormone on inhibition of apoptosis in mouse granulosa cells. Cell Death Dis 5:e1475
https://doi.org/10.1038/cddis.2014.400
61 CSifer, JLBenifla, AFBringuier, RPorcher, GBlanc-Layrac, PMadelenat, GFeldmann (2002) Could induced apoptosis of human granulosa cells predict in vitro fertilization-embryo transfer outcome? A preliminary study of 25 women. Eur J Obstet Gyn Reprod Biol 103:150–153
https://doi.org/10.1016/S0301-2115(02)00043-X
62 AHSinclair, PBerta, MSPalmer, JRHawkins, BLGriffiths, MJSmith, JWFoster, AMFrischauf, RLovell-Badge, PNGoodfellow (1990) A gene from the human sex-determining region encodes a protein with homology to a conserved DNA-binding motif. Nature 346:240–244
https://doi.org/10.1038/346240a0
63 YQSu, XMWu, MJO’Brien, FLPendola, JNDenegre, MMMatzuk, JJEppig (2004) Synergistic roles of BMP15 and GDF9 in the development and function of the oocyte-cumulus cell complex in mice: genetic evidence for an oocyte-granulosa cell regulatory loop. Dev Biol 276:64–73
https://doi.org/10.1016/j.ydbio.2004.08.020
64 YQSu, KSugiura, JJEppig (2009) Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med 27:32–42
https://doi.org/10.1055/s-0028-1108008
65 DSun, YZhang, CWang, XHua, XAZhang, JYan (2013) Sox9-related signaling controls zebrafish juvenile ovary-testis transformation. Cell Death Dis 4:e930
https://doi.org/10.1038/cddis.2013.456
66 ESutton, JHughes, SWhite, RSekido, JTan, VArboleda, NRogers, KKnower, LRowley, HEyreet al. (2011) Identification of SOX3 as an XX male sex reversal gene in mice and humans. J Clin Investig 121:328–341
https://doi.org/10.1172/JCI42580
67 HTakahashi (1977) Juvenile hermaphroditism in the zebrafish, Brachydanio rerio. Bull Fac Fish Hokkaido Univ 28:57–65
68 YTakehana, MMatsuda, TMyosho, MLSuster, KKawakami, ITShin, YKohara, YKuroki, AToyoda, AFujiyamaet al. (2014) Cooption of Sox3 as the male-determining factor on the Y chromosome in the fish Oryzias dancena. Nat Commun 5:4157
https://doi.org/10.1038/ncomms5157
69 RGThome, FFDomingos, HBSantos, PMMartinelli, YSato, ERizzo, NBazzoli (2012) Apoptosis, cell proliferation and vitellogenesis during the folliculogenesis and follicular growth in teleost fish. Tissue Cell 44:54–62
https://doi.org/10.1016/j.tice.2011.11.002
70 DUchida, MYamashita, TKitano, TIguchi (2002) Oocyte apoptosis during the transition from ovary-like tissue to testes during sex differentiation of juvenile zebrafish. J Exp Biol 205:711–718
71 DUchida, MYamashita, TKitano, TIguchi (2004) An aromatase inhibitor or high water temperature induce oocyte apoptosis and depletion of P450 aromatase activity in the gonads of genetic female zebrafish during sex-reversal. Comp Biochem Phys Part A 137:11–20
https://doi.org/10.1016/S1095-6433(03)00178-8
72 DSWang, TKobayashi, LYZhou, BPaul-Prasanth, SIjiri, FSakai, KOkubo, KMorohashi, YNagahama (2007a) Foxl2 up-regulates aromatase gene transcription in a female-specific manner by binding to the promoter as well as interacting with ad4 binding protein/steroidogenic factor 1. Mol Endocrinol 21:712–725
https://doi.org/10.1210/me.2006-0248
73 XGWang, RBartfai, ISleptsova-Freidrich, LOrban (2007b) The timing and extent of ‘juvenile ovary’ phase are highly variable during zebrafish testis differentiation. J Fish Biol 70:33–44
https://doi.org/10.1111/j.1095-8649.2007.01363.x
74 DSWang, LYZhou, TKobayashi, MMatsuda, YShibata, FSakai, YNagahama (2010) Doublesex- and Mab-3-related transcription factor-1 repression of aromatase transcription, a possible mechanism favoring the male pathway in tilapia. Endocrinology 151:1331–1340
https://doi.org/10.1210/en.2009-0999
75 MWatanabe, MTanaka, DKobayashi, YYoshiura, YOba, YNagahama (1999) Medaka (Oryzias latipes) FTZ-F1 potentially regulates the transcription of P-450 aromatase in ovarian follicles: cDNA cloning and functional characterization. Mol Cell Endocrinol 149:221–228
https://doi.org/10.1016/S0303-7207(99)00006-4
76 KAWebster, USchach, AOrdaz, JSSteinfeld, BWDraper, KRSiegfried (2017) Dmrt1 is necessary for male sexual development in zebrafish. Dev Biol 422:33–46
https://doi.org/10.1016/j.ydbio.2016.12.008
77 JWeiss, JJMeeks, LHurley, GRaverot, AFrassetto, JLJameson (2003) Sox3 is required for gonadal function, but not sex determination, in males and females. Mol Cell Biol 23:8084–8091
https://doi.org/10.1128/MCB.23.22.8084-8091.2003
78 CAWilson, SKHigh, BMMcCluskey, AAmores, YLYan, TATitus, JLAnderson, PBatzel, MJCarvan, MSchartlet al. (2014) Wild sex in zebrafish: loss of the natural sex determinant in domesticated strains. Genetics 198:1291–1308
https://doi.org/10.1534/genetics.114.169284
79 YXia, NPapalopulu, PKVogt, JLi (2000) The oncogenic potential of the high mobility group box protein Sox3. Cancer Res 60:6303–6306
80 HXia, CRZhong, XXWu, JChen, BBTao, XQXia, MJShi, ZYZhu, VLTrudeau, WHu (2018) Mettl3 mutation disrupts gamete maturation and reduces fertility in zebrafish. Genetics 208:729–743
https://doi.org/10.1534/genetics.117.300574
81 QYan, FWang, YMiao, XWu, MBai, XXi, YFeng (2016) Sexdetermining region Y-box3 (SOX3) functions as an oncogene in promoting epithelial ovarian cancer by targeting src kinase. Tumor Biol 37:12263–12271
https://doi.org/10.1007/s13277-016-5095-x
82 YJYang, YWang, ZLi, LZhou, JFGui (2017) Sequential, divergent, and cooperative requirements of Foxl2a and Foxl2b in ovary development and maintenance of zebrafish. Genetics 205:1551–1572
https://doi.org/10.1534/genetics.116.199133
83 BYao, LZhou, YWang, WXia, JFGui (2007) Differential expression and dynamic changes of SOX3 during gametogenesis and sex reversal in protogynous hermaphroditic fish. J Exp Zool Part A Ecol Genet Physiol 307:207–219
https://doi.org/10.1002/jez.361
84 JYe, LFang, HKZheng, YZhang, JChen, ZJZhang, JWang, STLi, RQLi, LBolundet al. (2006) WEGO: a web tool for plotting GO annotations. Nucleic Acids Res 34:W293–W297
https://doi.org/10.1093/nar/gkl031
85 YKYin, HPTang, YLiu, YChen, GFLi, XCLiu, HRLin (2017) Targeted disruption of aromatase reveals dual functions of cyp19a1a during sex differentiation in zebrafish. Endocrinology 158:3030–3041
https://doi.org/10.1210/en.2016-1865
86 JYuan, YZhang, YSheng, XZFu, HHCheng, RJZhou (2015) MYBL2 guides autophagy suppressor VDAC2 in the developing ovary to inhibit autophagy through a complex of VDAC2-BECN1-BCL2L1 in mammals. Autophagy 11:1081–1098
https://doi.org/10.1080/15548627.2015.1040970
[1] PAC-0347-18359-ZRJ_suppl_1 Download
[1] Chen Yin, Xiaoquan Li, Jiulin Du. Optic tectal superficial interneurons detect motion in larval zebrafish[J]. Protein Cell, 2019, 10(4): 238-248.
[2] Yuanlong Ge, Shu Wu, Zepeng Zhang, Xiaocui Li, Feng Li, Siyu Yan, Haiying Liu, Junjiu Huang, Yong Zhao. Inhibition of p53 and/or AKT as a new therapeutic approach specifically targeting ALT cancers[J]. Protein Cell, 2019, 10(11): 808-824.
[3] Ping Wang, Zunpeng Liu, Xiaoqian Zhang, Jingyi Li, Liang Sun, Zhenyu Ju, Jian Li, Piu Chan, Guang-Hui Liu, Weiqi Zhang, Moshi Song, Jing Qu. CRISPR/Cas9-mediated gene knockout reveals a guardian role of NF-κB/RelA in maintaining the homeostasis of human vascular cells[J]. Protein Cell, 2018, 9(11): 945-965.
[4] Haiyang Zhang,Jingjing Duan,Yanjun Qu,Ting Deng,Rui Liu,Le Zhang,Ming Bai,Jialu Li,Tao Ning,Shaohua Ge,Xia Wang,Zhenzhen Wang,Qian Fan,Hongli Li,Guoguang Ying,Dingzhi Huang,Yi Ba. Onco-miR-24 regulates cell growth and apoptosis by targeting BCL2L11 in gastric cancer[J]. Protein Cell, 2016, 7(2): 141-151.
[5] Qian Fan,Xiangrui Meng,Hongwei Liang,Huilai Zhang,Xianming Liu,Lanfang Li,Wei Li,Wu Sun,Haiyang Zhang,Ke Zen,Chen-Yu Zhang,Zhen Zhou,Xi Chen,Yi Ba. miR-10a inhibits cell proliferation and promotes cell apoptosis by targeting BCL6 in diffuse large B-cell lymphoma[J]. Protein Cell, 2016, 7(12): 899-912.
[6] Chao Lu,Yang Yang,Ran Zhao,Bingxuan Hua,Chen Xu,Zuoqin Yan,Ning Sun,Ruizhe Qian. Role of circadian gene Clock during differentiation of mouse pluripotent stem cells[J]. Protein Cell, 2016, 7(11): 820-832.
[7] Fan Chen,Jiebo Chen,Jiacheng Lin,Anton V. Cheltsov,Lin Xu,Ya Chen,Zhiping Zeng,Liqun Chen,Mingfeng Huang,Mengjie Hu,Xiaohong Ye,Yuqi Zhou,Guanghui Wang,Ying Su,Long Zhang,Fangfang Zhou,Xiao-kun Zhang,Hu Zhou. NSC-640358 acts as RXR&alpha; ligand to promote TNF&alpha;-mediated apoptosis of cancer cell[J]. Protein Cell, 2015, 6(9): 654-666.
[8] Xiangxuan Zhao,Yong Liu,Lei Du,Leya He,Biyun Ni,Junbo Hu,Dahai Zhu,Quan Chen. Threonine 32 (Thr32) of FoxO3 is critical for TGF-β-induced apoptosis via Bim in hepatocarcinoma cells[J]. Protein Cell, 2015, 6(2): 127-138.
[9] Anna Gortat,Mónica Sancho,Laura Mondragón,Àgel Messeguer,Enrique Pérez-Payá,Mar Orzáez. Apaf1 inhibition promotes cell recovery from apoptosis[J]. Protein Cell, 2015, 6(11): 833-843.
[10] Youguang Luo,Dengwen Li,Jie Ran,Bing Yan,Jie Chen,Xin Dong,Zhu Liu,Ruming Liu,Jun Zhou,Min Liu. End-binding protein 1 stimulates paclitaxel sensitivity in breast cancer by promoting its actions toward microtubule assembly and stability[J]. Protein Cell, 2014, 5(6): 469-479.
[11] Xiao-Xi Guo,Yang Li,Chao Sun,Dan Jiang,Ying-Jia Lin,Feng-Xie Jin,Seung-Ki Lee,Ying-Hua Jin. p53-dependent Fas expression is critical for Ginsenoside Rh2 triggered caspase-8 activation in HeLa cells[J]. Protein Cell, 2014, 5(3): 224-234.
[12] Guanghua Xu,Jing Wang,George Fu Gao,Cui Hua Liu. Insights into battles between Mycobacterium tuberculosis and macrophages[J]. Protein Cell, 2014, 5(10): 728-736.
[13] Yi Sun, Hua Li. Functional characterization of SAG/RBX2/ROC2/RNF7, an antioxidant protein and an E3 ubiquitin ligase[J]. Prot Cell, 2013, 4(2): 103-116.
[14] Shuang Sha, Honglin Jin, Xiao Li, Jie Yang, Ruiting Ai, Jinling Lu. Comparison of caspase-3 activation in tumor cells upon treatment of chemotherapeutic drugs using capillary electrophoresis[J]. Prot Cell, 2012, 3(5): 392-399.
[15] Yide Mei, Mian Wu. Multifaceted functions of Siva-1: more than an Indian God of Destruction[J]. Prot Cell, 2012, 3(2): 117-122.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed