Browse > Article
http://dx.doi.org/10.5713/ab.21.0259

Alterations of mRNA and lncRNA profiles associated with the extracellular matrix and spermatogenesis in goats  

Chen, Haolin (College of Animal Science and Technology, Gansu Agricultural University)
Miao Xiaomeng (Institute of Animal Husbandry and Veterinary, Guizhou Academy of Agricultural Sciences)
Xu, Jinge (Institute of Animal Husbandry and Veterinary, Guizhou Academy of Agricultural Sciences)
Pu, Ling (Institute of Animal Husbandry and Veterinary, Guizhou Academy of Agricultural Sciences)
Li, Liang (Institute of Animal Husbandry and Veterinary, Guizhou Academy of Agricultural Sciences)
Han, Yong (Institute of Animal Husbandry and Veterinary, Guizhou Academy of Agricultural Sciences)
Mao, Fengxian (Guizhou Province Livestock and Poultry Genetic Resources Management Station)
Ma, Youji (College of Animal Science and Technology, Gansu Agricultural University)
Publication Information
Animal Bioscience / v.35, no.4, 2022 , pp. 544-555 More about this Journal
Abstract
Objective: Spermatozoa are produced within the seminiferous tubules after sexual maturity. The expression levels of mRNAs and lncRNAs in testicular tissues are different at each stage of testicular development and are closely related to formation of the extracellular matrix (ECM) and spermatogenesis. Therefore, we set out to study the expression of lncRNAs and mRNAs during the different developmental stages of the goat testis. Methods: We constructed 12 RNA libraries using testicular tissues from goats aged 3, 6, and 12 months, and studied the functions of mRNAs and lncRNAs using the gene ontogeny (GO) and Kyoto encyclopedia of genes and genomes (KEGG) databases. Relationships between differentially expressed genes (DEGs) were analyzed by lncRNA-mRNA co-expression network and protein-protein interaction network (PPI). Finally, the protein expression levels of matrix metalloproteinase 2 (MMP2), insulin-like growth factor 2 (IGF2), and insulin-like growth factor-binding protein 6 (IGFBP6) were detected by western blotting. Results: We found 23, 8, and 135 differentially expressed lncRNAs and 161, 12, and 665 differentially expressed mRNAs that were identified between 3 vs 6, 6 vs 12, and 3 vs 12 months, respectively. GO, KEGG, and PPI analyses showed that the differential genes were mainly related to the ECM. Moreover, MMP2 was a hub gene and co-expressed with the lncRNA TCONS-0002139 and TCONS-00093342. The results of quantitative reverse-transcription polymerase chain reaction verification were consistent with those of RNA-seq sequencing. The expression trends of MMP2, IGF2, and IGFBP6 protein were the same as that of mRNA, which all decreased with age. IGF2 and MMP2 were significantly different in the 3 vs 6-month-old group (p<0.05). Conclusion: These results improve our understanding of the molecular mechanisms involved in sexual maturation of the goat testis.
Keywords
Goat; LncRNA; mRNA; Puberty; Testis;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Gao Y, Li S, Lai Z, et al. Analysis of long non-coding RNA and mRNA expression profiling in immature and mature bovine (Bos taurus) testes. Front Genet 2019;10:646. https://doi.org/10.3389/fgene.2019.00646   DOI
2 Siu MKY, Cheng CY. Extracellular matrix: recent advances on its role in junction dynamics in the seminiferous epithelium during spermatogenesis. Biol Reprod 2004;71:375-91. https://doi.org/10.1095/biolreprod.104.028225   DOI
3 Zhu Z, Li C, Yang S, et al. Dynamics of the transcriptome during human spermatogenesis: predicting the potential key genes regulating male gametes generation. Sci Rep 2016;6:19069. https://doi.org/10.1038/srep19069   DOI
4 Li C, Shen C, Shang X, et al. Two novel testis-specific long noncoding RNAs produced by 1700121C10Rik are dispensable for male fertility in mice. J Reprod Dev 2020;66:57-65. https://doi.org/10.1262/jrd.2019-104   DOI
5 Zeng F, Chen Y, Guo C, et al. Analysis of differentially abundant proteins related to boar fertility in seminal plasma using iTRAQ-based quantitative proteomics. J Proteomics 2021;236:104120. https://doi.org/10.1016/j.jprot.2021.104120   DOI
6 Mayerhofer A, Walenta L, Mayer C, Eubler K, Welter H. Human testicular peritubular cells, mast cells and testicular inflammation. Andrologia 2018;50:e13055. https://doi.org/10.1111/and.13055   DOI
7 Nosrati R, Kheirouri S, Ghodsi R, Ojaghi H. The effects of zinc treatment on matrix metalloproteinases: a systematic review. J Trace Elem Med Biol 2019;56:107-15. https://doi.org/10.1016/j.jtemb.2019.08.001   DOI
8 Cole JB, Durr JW, Nicolazzi EL. Invited review: The future of selection decisions and breeding programs: What are we breeding for, and who decides? J Dairy Sci 2021;104:5111-24. https://doi.org/10.3168/jds.2020-19777   DOI
9 Tan KS, Kulkeaw K, Nakanishi Y, Sugiyama D. Expression of cytokine and extracellular matrix mRNAs in fetal hepatic stellate cells. Genes Cells 2017;22:836-44. https://doi.org/10.1111/gtc.12517   DOI
10 van Dam S, Vosa U, van der Graaf A, Franke L, de Magalhaes JP. Gene co-expression analysis for functional classification and gene-disease predictions. Brief Bioinform 2018;19:575-92. https://doi.org/10.1093/bib/bbw139   DOI
11 Batista P, Chang H. Long noncoding RNAs: cellular address codes in development and disease. Cell 2013;152:1298-307. https://doi.org/10.1016/j.cell.2013.02.012   DOI
12 Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357-9. https://doi.org/10.1038/nmeth.1923   DOI
13 Liu K, Li T, Ton H, Mao X, Chen Y. Advances of long noncoding RNAs-mediated regulation in reproduction. Chin Med J 2018;131:226-34. https://doi.org/10.4103/0366-6999.222337   DOI
14 Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol 2013;14:R36. https://doi.org/10.1186/gb-2013-14-4-r36   DOI
15 Wathes D, Cheng Z, Fenwick MA, Fitzpatrick R, Patton J. Influence of energy balance on the somatotrophic axis and matrix metalloproteinase expression in the endometrium of the postpartum dairy cow. Reproduction (Cambridge, England) 2011;141:269-81. https://doi.org/10.1530/rep-10-0177   DOI
16 Voit-Ostricki L, Lovas S, Watts CR. Conformation and domain movement analysis of human matrix metalloproteinase-2: role of associated Zn2+ and Ca2+ ions. Int J Mol Sci 2019;20:4194. https://doi.org/10.3390/ijms20174194   DOI
17 Zhang C, Gao L, Xu EY. LncRNA, a new component of expanding RNA-protein regulatory network important for animal sperm development. Semin Cell Dev Biol 2016;59:110-7. https://doi.org/10.1016/j.semcdb.2016.06.013   DOI
18 Liu Y, Sun Y, Li Y, et al. Analyses of long non-coding RNA and mRNA profiling using RNA sequencing in chicken testis with extreme sperm motility. Sci Rep 2017;7:9055. https://doi.org/10.1038/s41598-017-08738-9   DOI
19 Zhang Y, Yang H, Han L, et al. Long noncoding RNA expression profile changes associated with dietary energy in the sheep testis during sexual maturation. Sci Rep 2017;7:5180. https://doi.org/10.1038/s41598-017-05443-5   DOI
20 Kong L, Zhang Y, Ye Z, et al. CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res 2007;35:W345-9. https://doi.org/10.1093/nar/gkm391   DOI
21 El-Gebali S, Mistry J, Bateman A, et al. The Pfam protein families database in 2019. Nucleic Acids Res 2019;47:D427-32. https://doi.org/10.1093/nar/gky995   DOI
22 Trapnell C, Williams B, Pertea G, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol 2010;28:511-5. https://doi.org/10.1038/nbt.1621   DOI
23 McMillen P, Holley SA. Integration of cell-cell and cell-ECM adhesion in vertebrate morphogenesis. Curr Opin Cell Biol 2015;36:48-53. https://doi.org/10.1016/j.ceb.2015.07.002   DOI
24 Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008;9:559. https://doi.org/10.1186/1471-2105-9-559   DOI
25 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 ΔΔ C T method. Methods 2001;25:402-8. https://doi.org/10.1006/meth.2001.1262   DOI
26 Young M, Wakefield M, Smyth G, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol 2010;11:R14. https://doi.org/10.1186/gb-2010-11-2-r14   DOI
27 Bao J, Wu J, Schuster A, Hennig G, Yan W. Expression profiling reveals developmentally regulated lncRNA repertoire in the mouse male germline. Biol Reprod 2013;89:107. https://doi.org/10.1095/biolreprod.113.113308   DOI
28 Joshi M, Rajender S. Long non-coding RNAs (lncRNAs) in spermatogenesis and male infertility. Reprod Biol Endocrinol 2020;18:103. https://doi.org/10.1186/s12958-020-00660-6   DOI
29 Yao Q, Chen T. LINC01128 regulates the development of osteosarcoma by sponging miR-299-3p to mediate MMP2 expression and activating Wnt/β-catenin signalling pathway. J Cell Mol Med 2020;24:14293-305. https://doi.org/10.1111/jcmm.16046   DOI
30 Matuszczak E, Komarowska M, Sankiewicz A, et al. Plasma concentration of MMP-1 and MMP-2 in boys with cryptorchidism and its lack of correlation with INSL3 and inhibin B. Scand J Clin Lab Invest 2019;79:412-8. https://doi.org/10.1080/00365513.2019.1637534   DOI
31 Clemmons DR. Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol 2018;61:T139-T69. https://doi.org/10.1530/jme-18-0016   DOI
32 Weng B, Ran M, Chen B, He C, Dong L, Peng F. Genome-wide analysis of long non-coding RNAs and their role in postnatal porcine testis development. Genomics 2017;109:446-56. https://doi.org/10.1016/j.ygeno.2017.07.001   DOI
33 Veneziano D, Nigita G, Ferro A. Computational approaches for the analysis of ncRNA through deep sequencing techniques. Front Bioeng Biotechnol 2015;3:77. https://doi.org/10.3389/fbioe.2015.00077   DOI
34 Sun L, Luo H, Bu D, et al. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res 2013;41:e166. https://doi.org/10.1093/nar/gkt646   DOI
35 Wang L, Park H, Dasari S, Wang S, Kocher JP, Li W. CPAT: Coding-potential assessment tool using an alignment-free logistic regression model. Nucleic Acids Res 2013;41:e74. https://doi.org/10.1093/nar/gkt006   DOI
36 Xie C, Mao X, Huang J, et al. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res 2011;39:W316-22. https://doi.org/10.1093/nar/gkr483   DOI
37 Safian D, Morais R, Bogerd J, Schulz R. Igf binding proteins protect undifferentiated spermatogonia in the zebrafish testis against excessive differentiation. Endocrinology 2016;157:4423-33. https://doi.org/10.1210/en.2016-1315   DOI
38 Yang H, Wang F, Li F, et al. Comprehensive analysis of long noncoding RNA and mRNA expression patterns in sheep testicular maturation. Biol Reprod 2018;99:650-61. https://doi.org/10.1093/biolre/ioy088   DOI
39 Bai M, Sun L, Zhao J, et al. Histological analysis and identification of spermatogenesis-related genes in 2-, 6-, and 12- month-old sheep testes. Naturwissenschaften 2017;104:84. https://doi.org/10.1007/s00114-017-1505-1   DOI
40 Huang H, Zhao W, Tang Z, et al. Characterization of porcine MMP-2 and its association with immune traits. Gene 2009;435:63-71. https://doi.org/10.1016/j.gene.2009.01.002   DOI
41 Su YY, Nishimoto T, Hoffman S, et al. Insulin-like growth factor binding protein-4 exerts antifibrotic activity by reducing levels of connective tissue growth factor and the C-X-C chemokine receptor 4. FASEB bioAdvances 2019;1:167-79. https://doi.org/10.1096/fba.2018-00015   DOI
42 Chen H, Fok K, Yu S, et al. CD147 is required for matrix metalloproteinases-2 production and germ cell migration during spermatogenesis. Mol Hum Reprod 2011;17:405-14. https://doi.org/10.1093/molehr/gar013   DOI
43 Finkielstain GP, Forcinito P, Lui JCK, et al. An extensive genetic program occurring during postnatal growth in multiple tissues. Endocrinology 2009;150:1791-800. https://doi.org/10.1210/en.2008-0868   DOI
44 Mayerhofer A. Human testicular peritubular cells: more than meets the eye. Reproduction (Cambridge, England) 2013; 145:R107-16. https://doi.org/10.1530/rep-12-0497   DOI
45 Schrade A, Kyronlahti A, Akinrinade O, et al. GATA4 regulates blood-testis barrier function and lactate metabolism in mouse sertoli cells. Endocrinology 2016;157:2416-31. https://doi.org/10.1210/en.2015-1927   DOI
46 Bo D, Jiang X, Liu G, Hu R, Chong Y. RNA-Seq implies divergent regulation patterns of lincRNA on spermatogenesis and testis growth in goats. Animals 2021;11:625. https://doi.org/10.3390/ani11030625   DOI