Skip to main content

Grade follicles transcriptional profiling analysis in different laying stages in chicken

Abstract

During follicular development, a series of key events such as follicular recruitment and selection are crucially governed by strict complex regulation. However, its molecular mechanisms remain obscure. To identify the dominant genes controlling chicken follicular development, the small white follicle (SWF), the small yellow follicle (SYF), and the large yellow follicle (LYF) in different laying stages (W22, W31, W51) were collected for RNA sequencing and bioinformatics analysis. There were 1866, 1211, and 1515 differentially expressed genes (DEGs) between SWF and SYF in W22, W31, and W51, respectively. 4021, 2295, and 2902 DEGs were respectively identified between SYF and LYF in W22, W31, and W51. 5618, 4016, and 4809 DEGs were respectively identified between SWF and LYF in W22, W31, and W51. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated that extracellular matrix, extracellular region, extracellular region part, ECM-receptor interaction, collagen extracellular matrix, and collagen trimer were significantly enriched (P < 0.05). Protein–protein interaction analysis revealed that COL4A2, COL1A2, COL4A1, COL5A2, COL12A1, ELN, ALB, and MMP10 might be key candidate genes for follicular development in chicken. The current study identified dominant genes and pathways contributing to our understanding of chicken follicular development. 

Peer Review reports

Introduction

In poultry breeds, high-efficiency follicular development means huge economic output for the egg industry. Follicles at different stages exist in the ovary of sexually mature hens, and a hierarchical system is formed in the ovary according to different functions and sizes: Pre-hierarchical follicle and hierarchical follicle (also known as pre-ovulatory follicle) [1]. Once ovulated, a new follicle is selected from the pre-hierarchal cohort to enter the hierarchical stage [2]. The development of follicles is crucially governed by strict intrinsic complex regulation [3]. During follicular development, a series of key events such as gene transcription and protein expression occur in series and are governed by specific gene expression, which is an intrinsic factor regulating follicular recruitment, selection, and apoptosis of follicles [4].

Over the past decade, RNA sequencing (RNA-seq) has become an indispensable tool for transcriptome analysis. RNA-seq is often used for analyzing differential expression genes [5]. RNA-seq has been widely applied to a variety of organisms such as Pigs [6], cattle [7], chickens [8], goats [9], deer [10], and mice [11]. Transcriptome studies have involved various traits such as egg quality [12], meat quality [13], genetic diversity [14], poultry disease screening [15], human aging [16], skeletal muscle development [17], growth [18].

The Nandan-Yao domestic chicken is a native breed in China. It has the characteristics of coarse food resistance, strong foraging ability, delicate and delicious meat, but its performance in egg production is low. In this study, RNA-seq and bioinformatics analysis were performed to identify the differentially expressed genes and pathways between different follicles in different laying stages to reveal the molecular mechanisms of follicular development.

Materials and methods

Ethics statement

All experimental and sample collection procedures were approved by the Institutional Animal Care and Use Committee (IACUC) of the College of Animal Science and Technology of Guangxi University (Guangxi, China), with approval number GXU2018-058.

Separation of follicles

Nandan-Yao domestic hens (Gallus gallus) purchased from Guangxi Guigang Gangfeng Agriculture and Husbandry Co. Ltd, laying continuously 3 eggs, were used in this study. Hens in early laying (22 weeks old, with a mean body weight of 1.67 ± 0.02 kg), peak laying (31 weeks old, with a mean body weight of 1.88 ± 0.06 kg), and late laying (51 weeks old, with a mean body weight of 2. 28 ± 0.08 kg) were selected for ovarian follicle sampling (n = 4). Follicles within the ovary were classified as small white follicles (SWF, 2–4 mm in diameter), large white follicles (LWF, 4–6 mm in diameter), small yellow follicles (SYF, 6–8 mm in diameter), and large yellow follicles (12–40 mm, named F5, F4, F3, F2, and F1, respectively) [19, 20]. SWFs, SYFs, and LYFs (F1, F2, F3) were collected for RNA extraction. The follicles were washed in PBS to remove the yolk.

Total RNA extraction

The total RNA was extracted from SWF, SYF, and LYF using TRIzol reagent (Invitrogen Life Technologies, USA) according to the manufacturer's instructions. RNA integrity was monitored on 1% agarose gels. RNA concentration was checked using the UV–Vis Spectrophotometer Q5000 (Quawell, USA).

RNA sequencing and quality control

The cDNA libraries were constructed and sequenced following the manufacturer’s standard procedures on an Illumina HiSeq 2500 (Illumina, San Diego, CA, USA) in Novogene Bioinformatics Technology Co., Ltd., Beijing, China. Raw reads of FASTQ format were processed with trim galore [21]. To obtain the clean reads, the sequence with low quality including adaptor sequences, quality score < 20, and N base rate of raw reads > 10% were removed. The Q20 scores, GC content, and sequence duplication levels of the clean data were calculated using FastQC [22].

RNA-Seq analysis

Reference genome and gene model annotation files were downloaded from the genome website (http://ftp.ensembl.org/pub/release-102/gtf/gallus_gallus/, http://ftp.ensembl.org/pub/release-102/fasta/gallus_gallus/dna/). The clean reads were mapped to the chicken reference genome using Hisat2v2.1.0 [23, 24]. The stringtiev2.1.1 was then used to annotate the transcripts [25]. The differential expressed genes between samples were identified using the DESeq2 R package (1.18.0) [26]. The P-value < 0.05 and |foldchange|> 2 were used as the criteria of significance. GO term and KEGG pathway analyses of coding genes were performed by the R package clusterProfiler 3.14.3 [27,28,29,30]. Both GO terms and KEGG pathways with corrected P-adjust < 0.05 were considered to be significantly enriched. The STRING (Franceschini et al., 2013) database was used to explore the interaction between DEGs. A confidence score > 0.9 was defined as valid.

Validation of RNA-Seq

RNA was reverse transcribed into cDNA using RT Reagent Kit (Takara, Dalian, China). Primer sequences of target and reference genes were shown in Supplemental table 1. QRT-PCR was carried out using SYBR Green Supermix kit (Takara, Dalian, China) in Bio-RAD CFX96 Real Time Detection system. The expression of β-actin was used to correct the gene expression data. The 2−ΔΔCT method was used to analyze the QRT-PCR data and calculate relative expression.

Results

Transcriptome data

As shown in Supplementary table 2, 18,911,563 to 34,680,085 clean reads per sample were obtained after quality control. The average GC content of all samples was 52.54%. The average mapped rate was 92.38% comparing clean reads with the reference genome. For all samples, at least 96.75% of the reads were equal to or exceeded Q20.

Analysis of differential expressed genes

At W22, 1866, 4021, and 5618 DEGs were respectively identified between SWF and SYF, SYF and LYF, SWF and LYF (Fig. 1A). At W31,1211, 2295 and 4016 DEGs were respectively identified between SWF and SYF, SYF and LYF, SWF and LYF (Fig. 1B). At W51, the number of DEGs between SWF and SYF, SYF and LYF, SWF and LYF were 1515, 2902, and 4809, respectively (Fig. 1C). As shown in Fig. 1a, 299, 440, and 267 DEGs were respectively obtained between W22 and W31, W22 and W51, W31 and W51 of SWF. In SYF, 295, 303, and 286 DEGs were respectively identified between W22 and W31, W22 and W51, W31 and W51 (Fig. 1b). In LYF, the number of DEGs between W22 and W31, W22 and W51, W31 and W51 were 488, 156, and 410, respectively (Fig. 1c).

Fig. 1
figure 1

Venn diagram of differentially expressed genes. (1) represents the Venn diagram of differentially expressed genes between different follicles at the same age. A, B, and C represent W21, W31, and W51, respectively. (2) represents the Venn diagram of differentially expressed genes in follicles of the same grade at different ages. a, b, and c represent SWF, SYF, and LYF, respectively. Abbreviations: SWF: small white follicle; SYF: small yellow follicle; LYF: large yellow follicle

GO and KEGG analysis for DEGs

Functional enrichment analysis was performed on DEGs of W51 intersection of SWF vs SYF, SYF vs LYF, SWF vs LYF (Fig. 2A, B). GO analysis indicated that differentially expressed genes were enriched in 8 items, including extracellular matrix, collagen-containing extracellular matrix, extracellular region part, extracellular region, collagen trimer, supramolecular complex, supramolecular polymer, supramolecular fiber (Supplementary table 3). KEGG analysis of differentially expressed mRNAs significantly enriched the ECM-receptor interaction and the Focal adhesion pathway (Supplementary table 4).

Fig. 2
figure 2

GO and KEGG enrichment analysis of DEGs. A and B represent GO enrichment and KEGG analysis of DEGs in the W51 intersection, respectively. C and D represent GO enrichment and KEGG analysis of DEGs in SWF intersection, respectively

Functional enrichment analysis was carried out for the intersection of DEGs of W22 vs W31, W22 vs W51, W31 vs W51 in SWF (Fig. 2C, D). The results showed that 17 items were significantly enriched in GO analysis (Supplementary table 5), such as extracellular region, extracellular region part, extracellular space, etc., and cytokine-cytokine receptor interaction was significantly enriched in KEGG analysis (Supplementary table 6).

Integration of PPI network

To reveal how these DEGs may interact, protein–protein interaction analyses were carried out based on the STRING database. The DEG network interaction analysis of W51 and SWF is shown in Fig. 3. The DEG network of W51 contains 13 genes, while the DEG network of SWF contains 37 genes. These genes may play an important regulatory role in the laying process.

Fig. 3
figure 3

The protein–protein interaction (PPI) network of DEGs. (A) PPI analysis of DEGs in the W51 intersection. (B) PPI analysis of DEGs in the SWF instersection. Line thickness indicates the strength of data support

Validation of RNA-seq

To verify our RNA-seq data, we selected 4 genes (CYP19A1, FOXL2, IGF1, SPP1) related to follicular development for QRT-PCR analysis (Fig. 4). The results showed that the differentially expressed genes had the same expression trends in QRT-PCR and RNA-seq, which validated their accuracy.

Fig. 4
figure 4

RNA-Seq validation using QRT-PCR. 4 DEGs (CYP19A1, FOXL2, IGF1, SPP1) were selected to test the accuracy of RNA sequencing. (A) Validation in the SWF and SYF at W22. (B) Validation in the SYF and LYF at W22. N = 6

Discussion

Follicular development is a complex physiological process, regulated by diverse genes and endocrine hormones [3]. Previous studies revealed the effect of GPR12 [20], GREM1 [31], BMP4 [32], RAC1 [33], FOXO3 [34], bFGF [35, 36], Melatonin [37, 38], CSAL1 and CSAL3 [39] on follicular development. RNA-seq has empowered many research areas and has led to new discoveries throughout the mRNA field [40]. In the present study, high-throughput transcriptome analyses were employed to study the differential gene expression profiles of three different follicles in different laying stages.

DEGs were significantly enriched in the extracellular matrix, extracellular region, extracellular region part, extracellular space, ECM receptor interaction, collagen containing extracellular matrix, and collagen trimer. The abilities of ECM to direct cell proliferation, differentiation, and function imply its remodeling in normal ovarian function [41]. The wall of the hen follicle is mainly composed of the extracellular matrix (ECM), which comprises collagenous fibers, dermatan sulfate, heparan sulfate, elastin, and hyaluronic acid [42].

Protein network interaction analyses of DEGs in W51 identified several genes associated with follicle development including COL4A2, COL1A2, COL4A1, COL5A2, COL12A1, ELN, FBN2, ALB, MMP10. COL4A2 (collagen type IV alpha 2 chain), COL1A2 (collagen type I alpha 2 chain), COL4A1 (collagen type IV alpha 1 chain), COL5A2 (collagen type V alpha 2 chain), COL12A1 (collagen type XII alpha 1chain) are five kinds of collagen. Type IV collagen is the main component of the basement membrane and constitutes its skeleton. It not only maintains the integrity of the basement membrane but also plays a key role in its formation. In normal conditions, the basement membrane is stable, dense, and continuous and can prevent macromolecules and cells from passing through [43]. The ELN gene encodes elastin. Fibrillin microfibrils are widely distributed components of extracellular matrices that function in the formation of elastin, serve structural roles and provide substrates for cell adhesion [44]. Albumin encoded by ALB may be a requirement for the control of follicle growth, which is attributable to albumin binding to specific cell-membrane components followed by the intracellular uptake of Alb-bound substances [45]. The MMP10 (matrix metallopeptidase 10) gene belongs to the matrix metallopeptidase family. A growing body of evidence suggests that MMPs play a relevant role in the ECM remodeling of ovarian tissues [46,47,48,49,50,51,52,53]. Many MMPs are produced in the mammalian ovary and participate in the regulation of ovarian functions [46, 49, 51, 53,54,55]. It indicates that increased collagen may support the structural integrity of follicles during growth.

Conclusions

The current study identified a series of key genes and signaling pathways associated with chicken follicular development by RNA-seq and bioinformatics analysis. These key genes (COL4A2, COL1A2, COL4A1, COL5A2, COL12A1, ELN, FBN2, ALB, MMP10) may regulate egg production by taking part in the extracellular matrix, extracellular region, extracellular region part, extracellular space, ECM-receptor interaction, collagen containing extracellular matrix and collagen trimer. The study constructed the transcriptional profiles of chicken growing follicles in different laying stages laying a foundation for further research on follicular development.

Availability of data and materials

The RNA sequencing data used and analyzed during the current study are available from the NCBI (accession number: PRJNA795703 and the link of the website: http://www.ncbi.nlm.nih.gov/bioproject/795703).

Abbreviations

RNA-Seq:

RNA sequencing

SWF:

Small white follicle

SYF:

Small yellow follicle

LYF:

Large yellow follicle

DEGs:

Differentially expressed genes

GO:

Gene Ontology

KEGG:

Kyoto Encyclopedia of Genes and Genomes

QRT-PCR:

Quantitative Real-time PCR

References

  1. Etches RJPJ. eptilian and avian follicular hierarchies: models for the study of ovarian development. R J Exp Zool Suppl. 1990;4:112–22.

    Article  CAS  Google Scholar 

  2. Onagbesan OBV, Decuypere EI. ntra-ovarian growth factors regulating ovarian function in avian species: a review. Anim Reprod Sci. 2009;111:121–40.

    Article  CAS  PubMed  Google Scholar 

  3. Zhou S, Ma Y, Zhao D, Mi Y, Zhang C. Transcriptome profiling analysis of underlying regulation of growing follicle development in the chicken. Poult Sci. 2020;99(6):2861–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Beg MA, Bergfelt DR, Kot K, Ginther OJ. Follicle selection in cattle: dynamics of follicular fluid factors during development of follicle dominance. Biol Reprod. 2002;66(1):120–6.

    Article  CAS  PubMed  Google Scholar 

  5. Stark R, Grzelak M, Hadfield J. RNA sequencing: the teenage years. Nat Rev Genet. 2019;20(11):631–56.

    Article  CAS  PubMed  Google Scholar 

  6. Jin L, Zhao L, Hu S, Long K, Liu P, Liu R, Zhou X, Wang Y, Huang Z, Lin X, et al. Transcriptional Differences of Coding and Non-Coding Genes Related to the Absence of Melanocyte in Skins of Bama Pig. Genes (Basel). 2019;11(1):47.

    Article  CAS  Google Scholar 

  7. Peng S, Song C, Li H, Cao X, Ma Y, Wang X, Huang Y, Lan X, Lei C, Chaogetu B, et al. Circular RNA SNX29 Sponges miR-744 to Regulate Proliferation and Differentiation of Myoblasts by Activating the Wnt5a/Ca(2+) Signaling Pathway. Mol Ther Nucleic Acids. 2019;16:481–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zou X, Wang J, Qu H, Lv XH, Shu DM, Wang Y, Ji J, He YH, Luo CL, Liu DW. Comprehensive analysis of miRNAs, lncRNAs, and mRNAs reveals potential players of sexually dimorphic and left-right asymmetry in chicken gonad during gonadal differentiation. Poult Sci. 2020;99(5):2696–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ren HWG, Chen L, Jiang J, Liu L, Li N, Zhao J, Sun X, Zhou P. Genome-wide analysis of long non-coding RNAs at early stage of skin pigmentation in goats (Capra hircus). BMC Genomics. 2016;17:67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Jia B, Liu Y, Li Q, Zhang J, Ge C, Wang G, Chen G, Liu D, Yang F. Altered miRNA and mRNA Expression in Sika Deer Skeletal Muscle with Age. Genes (Basel). 2020;11(2):172.

    Article  CAS  Google Scholar 

  11. Li B, Zhang K, Ye Y, Xing J, Wu Y, Ma Y, Li Y. Effects of Castration on miRNA, lncRNA, and mRNA Profiles in Mice Thymus. Genes (Basel). 2020;11(2):147.

    Article  CAS  Google Scholar 

  12. Zeng T, Zhang H, Liu J, Chen L, Tian Y, Shen J, Lu L. Genetic parameters of feed efficiency traits and their relationships with egg quality traits in laying period of ducks. Poult Sci. 2018;97(3):758–63.

    Article  CAS  PubMed  Google Scholar 

  13. Teng J, Gao N, Zhang H, Li X, Li J, Zhang H, Zhang X, Zhang Z. Performance of whole genome prediction for growth traits in a crossbred chicken population. Poult Sci. 2019;98(5):1968–75.

    Article  PubMed  Google Scholar 

  14. Li Q, Wang N, Du Z, Hu X, Chen L, Fei J, Wang Y, Li N. Gastrocnemius transcriptome analysis reveals domestication induced gene expression changes between wild and domestic chickens. Genomics. 2012;100:314–9.

    Article  CAS  PubMed  Google Scholar 

  15. Matulova M, Rajova J, Vlasatikova L, Volf J, Stepanova H, Havlickova H, Sisak F, Rychlik I. Characterization of chicken spleen transcriptome after infection with Salmonella enterica serovar Enteritidis. Plos One. 2012;7:e48101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Xiao FH, Chen XQ, Yu Q, Ye Y, Liu YW, Yan D, Yang LQ, Chen G, Lin R, Yang L, et al. Transcriptome evidence reveals enhanced autophagy-lysosomal function in centenarians. Genome Res. 2018;28(11):1601–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li T, Wang S, Wu R, Zhou X, Zhu D, Zhang Y. Identification of long non-protein coding RNAs in chicken skeletal muscle using next generation sequencing. Genomics. 2012;99:292–8.

    Article  CAS  PubMed  Google Scholar 

  18. Adapala NS, Kim HKW. A genome-wide transcriptomic analysis of articular cartilage during normal maturation in pigs. Gene. 2017;27:508–18.

    Article  CAS  Google Scholar 

  19. Johnson AL. Reproduction in the female. In: Scanes CG, editor. Sturkie's avian physiology . sixth ed. San Diego: Academic Press; 2015.p.635e65.

  20. Lin X, Ma Y, Qian T, Yao J, Mi Y, Zhang C. Basic fibroblast growth factor promotes prehierarchical follicle growth and yolk deposition in the chicken. Theriogenology. 2019;139:90–7.

    Article  CAS  PubMed  Google Scholar 

  21. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics. 2011;27(6):863–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. Transcript-level expression analysis of RNA-seq experiments with HISAT. StringTie and Ballgown Nature protocols. 2016;11(9):1650–67.

    Article  CAS  PubMed  Google Scholar 

  24. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33(3):290–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;5(16):284–7.

    Article  CAS  Google Scholar 

  28. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):1947–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–51.

    Article  CAS  PubMed  Google Scholar 

  31. Li Z, Jiang B, Cao B, Zhang Z, Zhang J, Li J, Huang Y, Wang Y. Characterization of Four Orphan Receptors (GPR3, GPR6, GPR12 and GPR12L) in Chickens and Ducks and Regulation of GPR12 Expression in Ovarian Granulosa Cells by Progesterone. Genes (Basel). 2021;12(4):489.

    Article  CAS  Google Scholar 

  32. Qin N, Tyasi TL, Sun X, Chen X, Zhu H, Zhao J, Xu R. Determination of the roles of GREM1 gene in granulosa cell proliferation and steroidogenesis of hen ovarian prehierarchical follicles. Theriogenology. 2020;151:28–40.

    Article  CAS  PubMed  Google Scholar 

  33. Yao J, Ma Y, Lin X, Zhou S, Mi Y, Zhang C. The Attenuating Effect of the Intraovarian Bone Morphogenetic Protein 4 on Age-Related Endoplasmic Reticulum Stress in Chicken Follicular Cells. Oxid Med Cell Longev. 2020;2020:4175613.

    PubMed  PubMed Central  Google Scholar 

  34. Tyasi TL, Sun X, Shan X, Liswaniso S, Chimbaka IM, Qin N, Xu R. Effects of RAC1 on Proliferation of Hen Ovarian Prehierarchical Follicle Granulosa Cells. Animals (Basel). 2020;10(9):1589.

    Article  Google Scholar 

  35. Cui C, Han SS, Yin HD, Luo B, Shen XX, Yang FL, Liu ZH, Zhu Q, Li DY, Wang Y. FOXO3 Is Expressed in Ovarian Tissues and Acts as an Apoptosis Initiator in Granulosa Cells of Chickens. Biomed Res Int. 2019;2019:6902906.

    PubMed  PubMed Central  Google Scholar 

  36. Guo CQ, Zhang GL, Lin X, Zhao D, Zhang CQ, Mi YL. Reciprocal stimulating effects of bFGF and FSH on chicken primordial follicle activation through AKT and ERK pathway. Theriogenology. 2019;132:27–35.

    Article  CAS  PubMed  Google Scholar 

  37. Hao EY, Wang DH, Chang LY, Huang CX, Chen H, Yue QX, Zhou RY, Huang RL. Melatonin regulates chicken granulosa cell proliferation and apoptosis by activating the mTOR signaling pathway via its receptors. Poult Sci. 2020;99(11):6147–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hao EY, Chen H, Wang DH, Huang CX, Tong YG, Chen YF, Zhou RY, Huang RL. Melatonin regulates the ovarian function and enhances follicle growth in aging laying hens via activating the mammalian target of rapamycin pathway. Poult Sci. 2020;99(4):2185–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhu HY, Qin N, Xu XX, Sun X, Chen XX, Zhao JH, Xu RF, Mishra B. Synergistic inhibition of csal1 and csal3 in granulosa cell proliferation and steroidogenesis of hen ovarian prehierarchical development. Biol Reprod. 2019;101(5):986–1000.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10(1):57–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Smith MFRW, Bakke LJ, Dow MP, Smith GW. Ovarian tissue remodeling: role of matrix metalloproteinases and their inhibitors. Mol Cell Endocrinol. 2002;191(1):45–56.

    Article  CAS  PubMed  Google Scholar 

  42. Hrabia A. Matrix Metalloproteinases (MMPs) and Inhibitors of MMPs in the Avian Reproductive System: An Overview. Int J Mol Sci. 2021;22(15):8056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Niemelä O, Risteli J, Blake JE, Risteli L, Compton KV, Orrego H. Markers of fibrogenesis and basement membrane formation in alcoholic liver disease. Gastroenterology. 1990;98:1612–9.

    Article  PubMed  Google Scholar 

  44. Burke RD, Wang D, Mark S, Martens G. Distribution of fibrillin I in extracellular matrix and epithelia during early development of avian embryos. Anat Embryol (Berl). 2000;201(4):317–26.

    Article  CAS  Google Scholar 

  45. Spanel-Borowski K. Immunocytochemical localization of albumin in ovarian follicles of fertile rats. Cell Tissue Res. 1987;248(3):699–702.

    Article  CAS  PubMed  Google Scholar 

  46. Curry TE Jr, Osteen KG. The matrix metalloproteinase system: changes, regulation, and impact throughout the ovarian and uterine reproductive cycle. Endocr Rev. 2003;24(4):428–65.

    Article  CAS  PubMed  Google Scholar 

  47. Curry TE Jr, Osteen KG. Cyclic changes in the matrix metalloproteinase system in the ovary and uterus. Biol Reprod. 2001;64(5):1285–96.

    Article  CAS  PubMed  Google Scholar 

  48. Peng J, Xin H, Han P, Gao K, Gao T, Lei Y, Ji S, An X, Cao B. Expression and regulative function of tissue inhibitor of metalloproteinase 3 in the goat ovary and its role in cultured granulosa cells. Mol Cell Endocrinol. 2015;412:104–15.

    Article  CAS  PubMed  Google Scholar 

  49. Hulboy DLR, Rudolph LA, Matrisian LM. Matrix metalloproteinases as mediators of reproductive function. Mol Hum Reprod. 1997;3:27–45.

    Article  CAS  PubMed  Google Scholar 

  50. Smith MF, Ricke WA, Bakke LJ, Dow MP. Smith GW : Ovarian tissue remodeling: Role of matrix metalloproteinases and their inhibitors. Mol Cell Endocrinol. 2002;191:45–56.

    Article  CAS  PubMed  Google Scholar 

  51. Ny T, Wahlberg P, Brandstrom IJ. Matrix remodeling in the ovary: regulation and functional role of the plasminogen activator and matrix metalloproteinase systems. Mol Cell Endocrinol. 2002;187(1–2):29–38.

    Article  CAS  PubMed  Google Scholar 

  52. Endo T, Kiya T, Goto T, Henmi H, Manase K, Honnma H, Baba T, Ishioka S, Hayashi T, Chida M, et al. Significance of matrix metalloproteinases in the pathophysiology of the ovary and uterus. Reprod Med Biol. 2006;5(4):235–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Fujihara M, Yamamizu K, Wildt DE, Songsasen N. Expression pattern of matrix metalloproteinases changes during folliculogenesis in the cat ovary. Reprod Domest Anim. 2016;51(5):717–25.

    Article  CAS  PubMed  Google Scholar 

  54. Goldman S, Shalev E. MMPS and TIMPS in ovarian physiology and pathophysiology. Front Biosci. 2004;9:2474–83.

    Article  CAS  PubMed  Google Scholar 

  55. Basini G, Bussolati S, Baioni L, Grasselli F. Gelatinases (MMP2 and MMP9) in swine antral follicle. BioFactors. 2011;37(2):117–20.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Science and Technology Key Project of Guangxi (GK AB21075010) and the Science and Technology Major Project of Guangxi (GK AA17204027).

Author information

Authors and Affiliations

Authors

Contributions

Xiurong Yang and Tiantian Sun designed the study. Tiantian Sun, Cong Xiao, Zhuliang Yang, Jixian Deng, Xiurong Yang performed the experiment, sample preparation, and bioinformatic analyses. Tiantian Sun wrote the manuscript. All authors approved the final manuscript. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Xiurong Yang.

Ethics declarations

Ethics approval and consent to participate

All experimental and sample collection procedures were approved by the Institutional Animal Care and Use Committee (IACUC) of the College of Animal Science and Technology of Guangxi University (Guangxi, China), with approval number GXU2018-058. All methods were carried out under relevant guidelines and regulations (declaration of helsinki). All methods are reported in accordance with ARRIVE guidelines for the reporting of animal experiments.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Supplementary Table 1. qPCR Primer sequences.

Additional file 2:

Supplementary Table 2. Summary of sequencing reads mapping to the reference genome and quality parameters.

Additional file 3:

Supplementary Table 3. Information of all enriched GO Terms based on DEGs of W51 intersection. Supplementary Table 4. Information of all enriched KEGG pathway based on DEGs in W51 intersection. Supplementary Table 5. Information of all enriched GO Terms based on DEGs in SWF intersection. Supplementary Table 6. Information of all enriched KEGG pathway based on DEGs in SWF intersection

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, T., Xiao, C., Yang, Z. et al. Grade follicles transcriptional profiling analysis in different laying stages in chicken. BMC Genomics 23, 492 (2022). https://doi.org/10.1186/s12864-022-08728-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-022-08728-w

Keywords