Scientia Agricultura Sinica ›› 2020, Vol. 53 ›› Issue (17): 3597-3605.doi: 10.3864/j.issn.0578-1752.2020.17.016

• ANIMAL SCIENCE·VETERINARY SCIENCE·RESOURCE INSECT • Previous Articles     Next Articles

Screening and Analysis of Follicular Development Related Genes in Goat

ZHAO YuanYuan1(),LI PengFei2,XU QinZhi1,AN QingMing1,MENG JinZhu1()   

  1. 1Tongren University, Tongren 554300, Guizhou
    2College of Life Science, Shanxi Agricultural University, Taigu 030801, Shanxi
  • Received:2019-06-24 Accepted:2020-07-10 Online:2020-09-01 Published:2020-09-11
  • Contact: JinZhu MENG E-mail:84840293@163.com;mjz122021@126.com

Abstract:

【Background】 Dominant follicles (DF) and subordinate follicles (SF) were the most important two stages in the follicle development in the first follicle wave of goat. With further development of follicles, DF may eventually develop into mature follicles until ovulation, and SF will move towards atresia, while apoptosis of granulosa cells is the key factor leading to follicular atresia. However, the molecular mechanisms of promoting follicle dominance or causing its atresia are still unclear. 【Objective】This study was aimed to screen the key genes affecting follicular development and provide a theoretical basis for further exploring the regulation mechanism of follicular development by high-throughput sequencing of DF and SF granulosa cells in the first follicular wave of goat.【Method】Ten healthy Guizhou white goats were selected (1 year old), and the prostaglandin F were injected respectively for estrus synchronization. B-type ultrasonography was used to detect the follicle growth situation, and then all of the goats were slaughtered when estrus had appeared for three days. DF (4.5-6 mm in diameter) and SF (3-4.5 mm in diameter) were obtained, granulosa cells were separated in the first follicle wave, respectively. Total RNA were extracted, and libraries were constructed and sequenced by Illumina Hiseq 2500 platform. FastQC was used to evaluate the quality of raw reads sequenced and filter them to obtain clean reads with high quality. Trinity was used to assemble clean reads from scratch to obtain unigenes. mRNA was obtained by comparing unigenes with goat RefSeq database using CLC Genomics Workbench. DESeq2 software was used to analyze the differential expression of the obtained mRNA. The goseq and kobas software were used for GO analysis and KEGG signal pathway analysis. Finally, qRT-PCR was used to verify the selected key genes that might affect the follicle development. 【Result】After the raw reads which obtained by sequencing were filtered, 43 217 934 clean reads were obtained from DF granulosa cells, accounting for 95.19% of raw reads. 40 766 348 clean reads were obtained from SF granulosa cells, accounting for 95.35% of raw reads. When the unigenes were compared with the RefSeq database of goat, a total of 33 896 annotated transcripts were obtained. Setting FPKM>1 and q value<0.05, a total of 13 644 genes were obtained in two types of follicle granulosa cells. By setting parameters: FPKM≥1, SF-FPKM/DF-FPKM>1, P<0.05, 695 differentially expressed mRNAs were obtained, of which 233 were significantly up-regulated and 462 were down-regulated in SF granulosa cells. GO functional enrichment analysis was performed on 695 differentially expressed mRNAs and concentrated in 42 groups of three major categories: biological processes accounted for 47.6%, cellular components 47.6%, and molecular functions 4.8%. KEGG signaling pathway analysis revealed 20 pathways, among which ribosome pathway related genes were most significantly enriched. Six genes that might be closely related to follicle development in goats were screened out, among which PRLR, PTX3 and RGN were up-regulated in subordinate follicles granulosa cells. DKK3, ALDH1A2 and RARRES1 were down-regulated. qRT-PCR showed that the expression trend of PRLR, RGN, DKK3, ALDH1A2 and RARRES1 was consistent with high-throughput sequencing results, and the expression level of RGN in SF granulosa cells was significantly higher than that of dominant follicles (P<0.01). The expression levels of DKK3, ALDH1A2 and RARRES1 in dominant follicles granulosa cells were significantly higher than subordinate follicles (P<0.01).【Conclusion】DKK3, ALDH1A2, RARRES1 and RGN had extremely significant differences in the expression levels of dominant follicles and subordinate follicles. It was speculated that those genes might promote the predominance of follicles or lead to atresia during the follicle development of goat, which was of great significance to further explore the regulation mechanism of follicular development.

Key words: goat, high-throughput sequencing, follicle development, granulosa cells

Table 1

The list of gene primers of qRT-PCR"

基因名称
Gene name
引物(5′→3′)
Primer sequences (5′→3′)
产物大小
Size (bp)
PRLR F:ACCAGTTCCAGGGCCAAAAA
R:GCATCAGGTGTTGGTCCTCA
173
PTX3 F:CTCTCTGGTCTGCAGTGTCG
R:TGAAGAGCTTGTCCCACTCG
153
RGN F:GGGTCCCTGTACTCCCTCTT
R:TGCGGTTGGAGATCTTTCCC
178
DKK3 F:GTGGCCTCTTCCCAGTACAC
R:GACCAGTTTAGCAGCCCTGT
160
ALDH1A2 F:TGGAATCCCTCAATGGTGGC
R:AGCCCAGCCTGCATAATACC
95
RARRES1 F:TAAAAGCCCCTTGAACGCAG
R:ACGTAAGAGCTGCCCAGAAA
102
18S rRNA F:CCCACGGAATCGAGAAAGAG
R:TTGACGGAAGGGCACCA
117

Fig. 1

Classification of raw reads in DF and SF granulosa cells"

Table 2

Top 20 highly expressed genes in SF and DF follicle granulosa cells"

基因名称Gene name SF-FPKM DF-FPKM
Eukaryotic Translation Elongation Factor 1 Alpha 1(EEF1A1) 7 110.13 8 928.81
Tumor Protein, Translationally-Controlled 1(TPT1) 3 655.98 4 714.90
TIMP Metallopeptidase Inhibitor 1(TIMP1) 3 412.45 859.27
Ribosomal Protein L37(RPL37) 3 144.41 3 930.05
Cytochrome C Oxidase Assembly Factor COX1(COX1) 2 528.75 1 340.44
Ribosomal Protein S14(RPS14) 2 440.53 2 907.19
Ribosomal Protein S18(RPS18) 2 304.26 2 709.49
Ribosomal Protein S11(RPS11) 2 132.58 2 437.76
Ribosomal Protein Lateral Stalk Subunit P0(RPLP0) 2 129.79 2 196.60
Ribosomal Protein L21(RPL21) 2 054.80 2 740.17
Ribosomal Protein S19(RPS19) 2 054.79 2 494.83
Ribosomal Protein L19(RPL19) 2 045.15 2 609.62
Ribosomal Protein S23(RPS23) 2 005.29 2 171.69
Oxytocin/Neurophysin I Prepropeptide (OXT) 1 907.67 559.09
Ribosomal Protein Lateral Stalk Subunit P2(RPLP2) 1 869.02 2 217.82
Ribosomal Protein S27a (RPS27A) 1 862.11 2 363.39
Ribosomal Protein S17(RPS17) 1 841.97 2 247.31
Receptor For Activated C Kinase 1(RACK1) 1 805.27 1 950.70
Ribosomal Protein L26(RPL26) 1 738.19 2 382.36
Ribosomal Protein S16(RPS16) 1 737.56 2 035.36

Fig. 2

Volcano plot of differentially expressed genes in DF and SF granulosa cells"

Fig. 3

GO analysis of differentially expressed genes in DF and SF granulosa cells 1: RNA processing; 2: RNA splicing; 3: nucleic acid metabolic process; 4: nucleobase-containing compound metabolic process; 5: ribonucleoprotein complex biogenesis; 6: cellular macromolecule metabolic process; 7: ribosome biogenesis; 8: heterocycle metabolic process; 9: cellular aromatic compound metabolic process; 10: cellular nitrogen compound metabolic process; 11: gene expression; 12: RNA metabolic process; 13: ribosomal small subunit biogenesis; 14: rRNA processing; 15: macromolecule metabolic process; 16: nitrogen compound metabolic process; 17: ncRNA processing; 18: ncRNA metabolic process; 19: organic cyclic compound metabolic process; 20: rRNA metabolic process; 21: nuclear lumen; 22: nuclear part; 23: nucleus; 24: intracellular organelle; 25: organelle; 26: membrane-enclosed lumen; 27: intracellular organelle lumen; 28: organelle lumen; 29: intracellular organelle part; 30: intracellular; 31: intracellular part; 32: organelle part; 33: intracellular membrane-bounded organelle; 34: membrane-bounded organelle; 35: nucleoplasm; 36: ribonucleoprotein complex; 37: cytosolic small ribosomal subunit; 38: cytosolic ribosome; 39: nucleoplasm part; 40: macromolecular complex; 41: RNA binding; 42: mRNA binding"

Fig. 4

Scatter plot of enriched KEGG pathway in DF and SF granulosa cells"

Table 3

Candidate genes may associate with follicular development of goat"

基因名
Gene name
SF-FPKM DF-FPKM log2差异倍数
log2 fold change
基因功能
Gene function
Prolactin receptor (PRLR) 37.46 4.11 3.19 刺激卵泡LH受体生成
Stimulate the production of LH receptors in the follicle
Pentraxin 3 (PTX3) 39.92 6.16 2.70 影响雌性动物生殖能力
Affecting females’ reproductive ability
Regucalcin (RGN) 49.39 8.13 2.60 调节Ca2+信号传导,以及Ca2+依赖的细胞过程和酶活性
Regulates Ca2+ signaling, and Ca2+ dependent cellular processes and enzyme activity
Dickkopf WNT Signaling Pathway Inhibitor 3 (DKK3) 15.92 74.57 -2.23 影响胚胎发育
Affect embryonic development
Aldehyde Dehydrogenase 1 Family Member A2 (ALDH1A2) 3.80 34.23 -3.17 建立局部胚胎视黄酸水平
Establishing local embryonic retinoic acid levels
Retinoic Acid Receptor Responder 1 (RARRES1) 110.41 527.73 -2.26 调节管状酪氨酸循环
Regulating tubular tyrosine cycle

Fig. 5

Relative expression of candidate genes in DF and SF granulosa cells of goat (** indicates P<0.01)"

[1] QUAN Q, ZHENG Q, LING Y H, FANG F G, CHU M X, ZHANG X R, LIU Y, LI W Y. Comparative analysis of differentially expressed genes between the ovaries from pregnant and nonpregnant goats using RNA-Seq. Journal of Biological Research Thessaloniki, 2019,26(3):1-12.
doi: 10.1186/s40709-019-0093-y
[2] RO S, SONG R, PARK C, ZHENG H, SANDERS K M, YAN W. Cloning and expression profiling of small RNAs expressed in the mouse ovary. RNA, 2007,13(12):2366-2380.
doi: 10.1261/rna.754207 pmid: 17951331
[3] LI P F, MENG J Z, ZHU Z W, FOLGER J K, LYU L H. Detection of genes associated with follicle development through transcriptome analysis of bovine ovarian follicles GCs. Current Bioinformatics, 2018,13:127-140.
doi: 10.2174/1574893612666170403165746
[4] BAO B, GARVERICK H A, SMITH G W, SMITH M F, SALFEN B E, YOUNGQUIST R S. Changes in messenger ribonucleic acid encoding luteinizing hormone receptor, cytochrome P450-side chain cleavage, and aromatase are associated with recruitment and selection of bovine ovarian follicles. Biology of Reproduction, 1997,56(5):1158-1168.
doi: 10.1095/biolreprod56.5.1158 pmid: 9160714
[5] REVERCHON M, CORNUAU M, RAMÉ C, GUERIF F, ROYÈRE D, DUPONT J. Resistin decreases insulin-like growth factor I-induced steroid production and insulin-like growth factor I receptor signaling in human granulosa cells. Fertility and Sterility, 2013,100(1):247-255.
doi: 10.1016/j.fertnstert.2013.03.008 pmid: 23548939
[6] KHARITONENKOV A, DIMARCHI R. Fibroblast growth factor 21 night watch: advances and uncertainties in the field. Journal of Internal Medicine, 2016,281(3):233-246.
doi: 10.1111/joim.12580 pmid: 27878865
[7] DE CASTRO T, RUBIANES E, MENCHACA A, RIVERO A. Ovarian dynamics, serum estradiol and progesterone concentrations during the interovulatory interval in goats. Theriogenology, 1999,52(3):399-411.
doi: 10.1016/S0093-691X(99)00138-7 pmid: 10734375
[8] LI J, LI Z, LIU S, ZIA R, LIANG A, YANG L. Transcriptome studies of granulosa cells at different stages of ovarian follicular development in buffalo. Animal Reproduction Science, 2017,187:181-192.
doi: 10.1016/j.anireprosci.2017.11.004 pmid: 29126830
[9] TERENINA E, FABRE S, BONNET A, MONNIAUX D, ROBERT- GRANIÉ C, SANCRISTOBAL M, TOSSER-KLOPP G. Differentially expressed genes and gene networks involved in pig ovarian follicular atresia. Physiological Genomics, 2017,49(2):67-80.
doi: 10.1152/physiolgenomics.00069.2016 pmid: 27940565
[10] HUSSEIN M R. Apoptosis in the ovary: Molecular mechanisms. Human Reproduction Update, 2005,11(2):162-178.
doi: 10.1093/humupd/dmi001 pmid: 15705959
[11] FOLGER J K, JIMENEZ-KRASSEL F, IRELAND J J, LV L L, SMITH G W. Regulation of granulosa cell cocaine and amphetamine regulated transcript (CART) binding and effect of CART signaling inhibitor on granulosa cell estradiol production during dominant follicle selection in cattle. Biology of Reproduction, 2013,137:1-8.
[12] BROMFIELD J J, IACOVIDES S M. Evaluating lipopolysaccharide- induced oxidative stress in bovine granulosa cells. Journal of Assisted Reproduction and Genetics, 2017,34(12):1619-1626.
doi: 10.1007/s10815-017-1031-2 pmid: 28866821
[13] HAN P, XIN H Y, PENG J X, ZHANG L, SONG Y X, LI G, CAO B Y, AN X P. Identification and expression of X-linked inhibitor of apoptosis protein during follicular development in goat ovary. Theriogenology, 2017,98:30-35.
doi: 10.1016/j.theriogenology.2017.04.048 pmid: 28601152
[14] LI P F, MENG J Z, LIU W Z, SMITH G W, YAO J B, LYU L H. Transcriptome analysis of bovine ovarian follicles at predeviation and onset of deviation stages of a follicular wave. International Journal of Genomics, 2016,2016:1-9.
[15] ZI X D, CHEN D W, WANG H M. Molecular characterization, mRNA expression of prolactin receptor ( PRLR) gene during pregnancy, nonpregnancy in the yak( Bos grunniens). General and Comparative Endocrinology, 2012,175(3):384-388.
[16] RUAN W, CATANESE V, WIECZOREK R, FELDMAN M, KLEINBERG D L. Estradiol enhances the stimulatory effect of insulin-like growth factor-I (IGF-I) on mammary development and growth hormone-induced IGF-I messenger ribonucleic acid. Endocrinology, 1995,136(3):1296-1302.
doi: 10.1210/endo.136.3.7867584 pmid: 7867584
[17] PERKS C M, NEWCOMB P V, GROHMANN M, WRIGHT R J, MASON H D, HOLLY J M. Prolactin acts as a potent survival factor against C2-ceramide-induced apoptosis in human granulosa cells. Human Reproduction, 2003,18(12):2672-2677.
pmid: 14645190
[18] TISSIER P R, HODSON D J, MARTIN A O, ROMANÒ N, MOLLARD P. Plasticity of the prolactin (PRL) axis: Mechanisms underlying regulation of output in female mice. Advances in Experimental Medicine & Biology, 2015,846:139-162.
doi: 10.1007/978-3-319-12114-7_6 pmid: 25472537
[19] LI S H, LIN M H, HWU Y M, LU C H, YEH L Y, CHEN Y J, LEE R K. Correlation of cumulus gene expression of GJA1, PRSS35, PTX3, and SERPINE2 with oocyte maturation, fertilization, and embryo development. Reproductive Biology Endocrinology, 2015,13(1):93.
doi: 10.1186/s12958-015-0091-3
[20] ZHANG X Q, JAFARI N, BARNES R B, CONFINO E, MILAD M, KAZER R R. Studies of gene expression in human cumulus cells indicate pentraxin 3 as a possible marker for oocyte quality. Fertillty and Sterility, 2005,83(4):1169-1179.
[21] GARLANDA C, BOTTAZZI B, BASTONE A, MANTOVANI A. Pentraxins at the crossroads between innate immunity, inflammation, matrix deposition, and female fertility. Annual Review of Immunology, 2005,23(1):337-366.
[22] HATZIRODOS N, IRVING-RODGERS H F, HUMMITZSCH K, HARLAND M L, MORRIS S E, RODGERS R J. Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes. BMC Genomics, 2014,15(1):24.
[23] VOORHAM Q J, JANSSEN J, TIJSSEN M, SNELLENBERG S, MONGERA S, GRIEKEN N C, GRABSCH H, KLIMENT M, REMBACKEN B J, MULDER C J, ENGELAND M V, MEIJER G A, STEENBERGEN R D, CARVALHO B. Promoter methylation of Wnt-antagonists in polypoid and nonpolypoid colorectal adenomas. BMC Cancer, 2013,13(1):603.
doi: 10.1186/1471-2407-13-603
[24] BHATTACHARYYA S, FEFERMAN L, TOBACMAN J K. Chondroitin sulfatases differentially regulate Wnt signaling in prostate stem cells through effects on SHP2, phospho-ERK1/2, and Dickkopf Wnt signaling pathway inhibitor (DKK3). Oncotarget, 2017,59(8):100242-100260.
[25] WANG X S, SPERKOVA Z, NAPOLI J L. Analysis of mouse retinal dehydrogenase type 2 promoter and expression. Genomics, 2001,74(2):245-250.
doi: 10.1006/geno.2001.6546 pmid: 11386761
[26] LI X H, KAKKAD B, ONG D E. Estrogen directly induces expression of retinoic acid biosynthetic enzymes, compartmentalized between the epithelium and underlying stromal cells in rat uterus. Endocrinology, 2004,145(10):4756-4762.
doi: 10.1210/en.2004-0514 pmid: 15205379
[27] KUMAR S, CUNNINGHAM T J, DUESTER G. Resolving molecular events in the regulation of meiosis in male and female germ cells. Science Signaling, 2013,6(288): pe25.
[28] KRENTZ A D, MURPHY M W, SARVER A L, GRISWOLD M D, BARDWELL V J, ZARKOWER D. DMRT1 promotes oogenesis by transcriptional activation of Stra8 in the mammalian fetal ovary. Developmental Biology, 2011,356(1):63-70.
doi: 10.1016/j.ydbio.2011.05.658 pmid: 21621532
[29] NAILLAT F, PRUNSKAITE-HYYRYLAINEN R, PIETILA I, SORMUNEN R, JOKELA T, SHAN J D, VAINIO S J. Wnt4/5a signalling coordinates cell adhesion and entry into meiosis during presumptive ovarian follicle development. Human Molecular Genetics, 2010,19(8):1539-1550.
doi: 10.1093/hmg/ddq027 pmid: 20106871
[30] BOWLES J, FENG C W, SPILLER C, DAVIDSON T L, JACKSON A, KOOPMAN P. FGF9 suppresses meiosis and promotes male germ cell fate in mice. Developmental Cell, 2010,19(3):440-449.
doi: 10.1016/j.devcel.2010.08.010 pmid: 20833365
[1] LIU YuFang,CHEN YuLin,ZHOU ZuYang,CHU MingXing. miR-221-3p Regulates Ovarian Granulosa Cells Apoptosis by Targeting BCL2L11 in Small-Tail Han Sheep [J]. Scientia Agricultura Sinica, 2022, 55(9): 1868-1876.
[2] WU Yan,ZHANG Hao,LIANG ZhenHua,PAN AiLuan,SHEN Jie,PU YueJin,HUANG Tao,PI JinSong,DU JinPing. circ-13267 Regulates Egg Duck Granulosa Cells Apoptosis Through Let-7-19/ERBB4 Pathway [J]. Scientia Agricultura Sinica, 2022, 55(8): 1657-1666.
[3] WANG JiaMin,SHI JiaChen,MA FangFang,CAI Yong,QIAO ZiLin. Effects of Soy Isoflavones on the Proliferation and Apoptosis of Yak Ovarian Granulosa Cells [J]. Scientia Agricultura Sinica, 2022, 55(8): 1667-1675.
[4] LI Heng,ZI XiangDong,WANG Hui,XIONG Yan,LÜ MingJie,LIU Yu,JIANG XuDong. Screening of Key Regulatory Genes for Litter Size Trait Based on Whole Genome Re-Sequencing in Goats (Capra hircus) [J]. Scientia Agricultura Sinica, 2022, 55(23): 4753-4768.
[5] MA XueMeng,YU ChengMin,SAI XiaoLing,LIU Zhen,SANG HaiYang,CUI BaiMing. PSORA: A Strategy Based on High-Throughput Sequence for Analysis of T-DNA Insertion Sites [J]. Scientia Agricultura Sinica, 2022, 55(15): 2875-2882.
[6] LI LiYing,HE YingTing,ZHONG YuYi,ZHOU XiaoFeng,ZHANG Hao,YUAN XiaoLong,LI JiaQi,CHEN ZanMou. CTNNB1 Regulates the Function of Porcine Ovarian Granulosa Cells [J]. Scientia Agricultura Sinica, 2022, 55(15): 3050-3061.
[7] ZHANG Jing,ZHANG JiYue,YUE YongQi,ZHAO Dan,FAN YiLing,MA Yan,XIONG Yan,XIONG XianRong,ZI XiangDong,LI Jian,YANG LiXue. LKB1 Regulates Steroids Synthesis Related Genes Expression in Bovine Granulosa Cells [J]. Scientia Agricultura Sinica, 2022, 55(10): 2057-2066.
[8] DU Yu,WANG Yong,MENG QingYong,ZHU JiangJiang,LIN YaQiu. Knockdown Goat KLF12 to Promote Subcutaneous Adipocytes Differentiation [J]. Scientia Agricultura Sinica, 2022, 55(1): 184-196.
[9] ZHAO Le,YANG HaiLi,LI JiaLu,YANG YongHeng,ZHANG Rong,CHENG WenQiang,CHENG Lei,ZHAO YongJu. Expression Patterns of TETs and Programmed Cell Death Related Genes in Oviduct and Uterus of Early Pregnancy Goats [J]. Scientia Agricultura Sinica, 2021, 54(4): 845-854.
[10] DU Yu,ZHU ZhiWei,WANG Jie,WANG XiuNa,JIANG HaiBin,FAN YuanChan,FAN XiaoXue,CHEN HuaZhi,LONG Qi,CAI ZongBing,XIONG CuiLing,ZHENG YanZhen,FU ZhongMin,CHEN DaFu,GUO Rui. Construction and Annotation of Ascosphaera apis Full-Length Transcriptome Utilizing Nanopore Third-Generation Long-Read Sequencing Technology [J]. Scientia Agricultura Sinica, 2021, 54(4): 864-876.
[11] FENG YunKui,WANG Jian,MA JinLiang,ZHANG LiuMing,LI YongJun. Effects of miR-31-5p on the Proliferation and Apoptosis of Hair Follicle Stem Cells in Goat [J]. Scientia Agricultura Sinica, 2021, 54(23): 5132-5143.
[12] SHAO MeiQi,ZHAO WeiSong,SU ZhenHe,DONG LiHong,GUO QingGang,MA Ping. Effect of Bacillus subtilis NCD-2 on the Growth of Tomato and the Microbial Community Structure of Rhizosphere Soil Under Salt Stress [J]. Scientia Agricultura Sinica, 2021, 54(21): 4573-4584.
[13] CHEN Yuan,CAI He,LI Li,WANG LinJie,ZHONG Tao,ZHANG HongPing. Alternative Splicing of TNNT3 and Its Effect on the Differentiation of MuSCs in Goat [J]. Scientia Agricultura Sinica, 2021, 54(20): 4466-4477.
[14] HUANG ZiYue,LIU WenJun,QIN RenLiu,PANG ShiChan,XIAO Jian,YANG ShangDong. Endophytic Bacterial Community Composition and PICRUSt Gene Functions in Different Pumpkin Varieties [J]. Scientia Agricultura Sinica, 2021, 54(18): 4018-4032.
[15] DU Xing,ZENG Qiang,LIU Lu,LI QiQi,YANG Liu,PAN ZengXiang,LI QiFa. Identification of the Core Promoter of Linc-NORFA and Its Transcriptional Regulation in Erhualian Pig [J]. Scientia Agricultura Sinica, 2021, 54(15): 3331-3342.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!