RNA sequencing-based identification of microRNAs in the antler cartilage of Gansu red deer (Cervus elaphus kansuensis)

View article
Zoological Science

Introduction

Velvet antlers, the bone-free and furry horns in male deer, develop through a highly complicated and genetically programmed process facilitating the growth of bone, skin, blood vessels and nerves. The proliferation and differentiation of cells in growing antler tips are regulated by several intracellular and extracellular factors and signaling pathways (Hu et al., 2019). The growth process of velvet antlers is very similar to endochondral ossification of limbs, which drives cartilage formation through mesenchymal cell differentiation. Therefore, velvet antlers are often used as a model to study cartilage and bone tissue regeneration and injury repair (Price & Allen, 2004).

During their rapid growth phase, antlers can exceed 2 cm/day. This is a hot topic in the field of biology (Price, Faucheux & Allen, 2005; Chu et al., 2017). Combining data on growth rate, shedding time and age, the fastest growing period of velvet antlers was ∼60 days after shedding, indicating that this is a critical stage during antler development. However, the underlying mechanisms involved in the rapid growth stage (RGS) of antler development remains unknown. Deer antler growth is mainly controlled by the growth center located at antler tips, comprising velvet skin, mesenchyme and cartilage tissue (Li et al., 2002). The cartilage zone dominates a large part of the antler growth center, and hence, plays a key role during the RGS of antler development (Ba et al., 2019).

MicroRNAs (miRNAs) are a class of small non-coding RNAs (ncRNAs), usually 17–22 nucleotides long, which are critical in post-transcriptional gene regulation via target messenger RNA (mRNA) degradation or via translation inhibition. MicroRNAs function through pairing with complementary sequences in the 3′-untranslated region (3′-UTR) of their target mRNAs (Mohr & Mott, 2015). These small regulatory molecules are ubiquitous in the genomes of animals, plants and even some viruses (Lu & Rothenberg, 2018). Previous studies have provided a considerable amount of information on the miRNA-mediated regulation of various biological processes, including organ development (Zhang et al., 2016a; Zhang et al., 2016b), phase transition (Wójcik & Gaj, 2016; Zhang et al., 2016a; Zhang et al., 2016b) and stress response (Candar-Cakir, Arican & Zhang, 2016; Li et al., 2016). Furthermore, recent studies have reported that specific miRNAs participate in the regulation of cartilage development, degradation and integrity (Zhang et al., 2019; Liu et al., 2020; Tu et al., 2020).

Deep sequencing methods now provide a fast and convenient way to identify and profile small RNA populations in various tissues and at different developmental stages (Fahlgren et al., 2007). Recent advances in RNA sequencing (RNA-seq) have also provided new opportunities to integrate changes in protein-coding mRNAs with regulatory signals, such as those driven by ncRNAs and alternative RNA splicing events (Ackerman 4th et al., 2018). RNA-seq has been widely used to study miRNA expression in humans, other animals and plants (Chiba et al., 2021; Hao et al., 2021; Wang et al., 2021; Yang et al., 2021).

To elucidate miRNA function during the RGS of antler development, we investigated miRNA expression profiles in the cartilage of antler growth centers at three developmental stages, specifically at 30, 60, and 90 days (d). During the proliferation stage, antlers grow at an extremely rapid rate without any carcinogenesis, which is unique among mammals. This growth characteristic makes deer antlers an ideal medical model for studying cancer treatment. The longitudinal growth of velvet antler is achieved through osteogenesis in the cartilage of each branch, very similar to the cartilage formation process in mammals. Thus, the velvet antler is a model of cartilage damage repair. Our findings may contribute new insights into the roles of miRNAs and their target mRNAs in the complex regulatory networks affecting antler development.

Materials & Methods

Ethics approval statement

All experimental protocols were approved by the Institutional Animal Care and Use Committee of Qinghai University (Xining, China), and all methods were carried out in accordance with approved guidelines and regulations (code: SL-2022024). All procedures involving animals were conducted in accordance with the US National Institutes of Health Guide for the Care and Use of Laboratory Animals (National Academies Council, 2011). This study was carried out in compliance with the ARRIVE guidelines for reporting animal research (Percie du Sert et al., 2020).

Animals and samples collection

Samples were collected from a C. elaphus kansuensis population of 200 individuals, raised in a semi-wild setting and fed under the same conditions (Shandan County, Gansu Province, China). Cartilage tissues in the antler tips at different growth stages (30, 60, and 90 d) were collected from three healthy adult individuals (4–5 years old, male) for RNA extraction. Three antler samples from three different individuals were prepared for RNA-seq and qRT-PCR. Antlers were collected after anaesthetising deer with special Mian Naining (anaesthetic, No.9812, People’s Liberation Army (PLA) Military Supplies University Research Institute, China), and hemostasis measures were taken immediately after velvet cutting. After recovering from anaesthesia, deer were returned to the herd.

RNA extraction, library preparation, and sequencing

Total RNA was extracted using TRIzol™ Reagent (Thermo Fisher Scientific, Waltham, MA, USA) following the manufacturer’s protocol. Extracted RNA was treated with DNase I (Promega, Madison, WI, USA) to remove contamination. To ensure that the RNA samples were qualified for sequencing, concentration and integrity were assessed using a NanoDrop 2000 Spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA) and RNA Nano 6000 Assay Kit with the Bioanalyzer 2100 System (Agilent Technologies, Santa Clara, CA, USA), respectively.

In this study, sample collection, high-throughput sequencing and data collection were conducted as previously described in Chen et al. (2022). Specifically, a total of 2.5 ng RNA per sample was used as input material for library preparation. Sequencing libraries were generated using NEBNext® Ultra™ RNA Library Prep Kit for Illumina® (New England Biolab, Ipswich, MA, USA) following manufacturer’s protocol, and index codes were added to attribute sequences. Library quality was assessed using Bioanalyzer 2100 (Agilent Technologies, Santa Clara, CA, USA).

Pooled miRNA libraries were sequenced for 50 cycles using HiSeq® Rapid SBS Kit v2 and HiSeq® 2500 platform (Illumina, San Diego, CA, USA). Sequencing data are available at the National Center for Biotechnology Information (NCBI) Sequence Read Archive (SRA) (BioProject: PRJNA731682).

Data analysis

Raw reads in FASTQ format were preprocessed using in-house Perl scripts. In this step, clean reads were obtained via trimming adapters and removing reads containing poly(A) tails, low-quality bases, and sequences <18 or >30 nucleotides long. Additionally, Q20 and Q30 scores, GC content (%), and sequence duplication level of cleaned data were determined.

Second, cleaned reads were aligned against the Silva (Quast et al., 2013) (https://www.arb-silva.de/), Genomic tRNA (Chan & Lowe, 2016) (GtRNAdb; http://lowelab.ucsc.edu/GtRNAdb/), Rfam (Kalvari et al., 2018) (http://rfam.xfam.org/), and Repbase (Bao, Kojima & Kohany, 2015) (http://www.girinst.org/repbase) databases using Bowtie (Langmead et al., 2009) to remove sequences that matched to rRNAs, tRNAs, snRNAs, snoRNAs, other ncRNAs, and repeats. Then clean small RNAs with a length range from 18 to 35 nucleotides were further analyzed. Third, Randfold tools (Bonnet et al., 2004) (http://www.aquafold.com) was used for predicting novel miRNA secondary structure. Fourth, the miRBase v21.0 database (Kozomara & Griffiths-Jones, 2014) (http://www.mirbase.org/) with 0 or 1 mismatch and miRDeep2 (Friedländer et al., 2012) with default parameters were used to identify known miRNAs and predict novel miRNAs. The miRNA expression profiles were determined based on TPM values using the normalized formula (Zhao, Ye & Stanton, 2020): TPM = 106 ∗ [Reads mapped to transcript/Transcript length]/[Sum (Reads mapped to transcript/Transcript length)]. (Note: “Reads mapped to the transcript” refers to read number of one miRNA, “Transcript length” refers to length of that one miRNA).

Based on the normalization counts, the significantly differentially expressed miRNAs in three stages were determined by their fold change. Based on a power analysis using the IDEG6 (Romualdi et al., 2003), we determined that our design had over 99% power to detect differentially expressed miRNAs (DEMs) at | log2(FC) | > = 1 and FDR < = 0.01 (Hou et al., 2019). We then screened for DEMs in the 30 d vs. 60 d and 60 d vs. 90 d pairwise comparisons.

MiRNA target prediction and functional analysis of candidate target genes

We put more focus on the 25 selected DEMs and the 30 specifically expressed miRNAs in 60 d library. Potential target genes of novel miRNAs and specifically expressed miRNAs were predicted using Miranda v3.3a (https://anaconda.org/bioconda/miranda) and TargetScan 7.2 (http://www.targetscan.org/vert_72/) with default parameters. Genes simultaneously predicted by both methods were considered as candidate targets of selected miRNAs. Functional annotation of these candidates were performed using GO enrichment (Ashburner et al., 2000) and KEGG pathway (Kanehisa & Goto, 2000) analyses.

Verification of miRNA expression using qRT-PCR

Stem-loop qRT-PCR was used to verify the accuracy of the sequencing data. Twelve of the selected 25 DEMs had similar names (except species), sequences and expression levels, and the remaining 13 DEMs were used for qRT-PCR. Total RNA was extracted as previously described. Approximately 1 µg of total RNA was reverse transcribed using Revert Aid First Strand cDNA Synthesis Kit (Fermentas, Thermo Fisher Scientific, Waltham, MA, USA). Reactions were incubated at 42 °C for 15 min and then terminated at 80 °C for 5 s. The qRT-PCR assay was performed using TB Green® Premix Ex Taq™ II (Tli RNaseH Plus) Kit (Takara Bio, Shiga, Japan) and CFX96 Real-time PCR Detection System (Bio-Rad Laboratories, Hercules, CA, USA). The miRNA expression level was detected using stem-loop qRT-PCR and miRNA-specific stem-loop primers. Gene-specific forward primers and universal reverse primers (Sangon Biotech, Shanghai, China) were used for qRT-PCR, with U6 RNA as internal control. Cycling profiles were as follows: 95 °C for 30 s, followed by 40 cycles at 95 °C for 5 s, and 60 °C for 30 s. Table 1 lists all primers used in this study. Melting curve analysis was also performed to determine product specificity. All statistical analyses were performed in triplicate. All data are presented as the mean ± standard deviation. Relative gene expression was subsequently analyzed using the comparative 2−ΔΔCt method (Livak & Schmittgen, 2001; Rao et al., 2013).

Table 1:
Primers uesd in qPCR.
Primers used for reverse transcription and stem-loop real-time qPCR.
Gene ID miRNA sequence Primers Sequence
U6 RNA RTP AACGCTTCACGAATTTGCGT
Forward primer CTCGCTTCGGCAGCACA
Reverse primer AACGCTTCACGAATTTGCG
aga-miR-184 TGGACGGAGAACTGATAAGGG RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCCCCTTA
Forward primer CTTATCAGTTCTCCGTCCATGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
bta-miR-146b TGAGAACTGAATTCCATAGGCTGT RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCACAGCC
Forward primer GCCTATGGAATTCAGTTCTCAGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
bta-miR-187 TCGTGTCTTGTGTTGCAGCCGG RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCCCGGCT
Forward primer TGCAACACAAGACACGAGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
bta-miR-27a-5p AGGGCTTAGCTGCTTGTGAGCA RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCTGCTCA
Forward primer CAAGCAGCTAAGCCCTTGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
bta-miR-504 AGACCCTGGTCTGCACTCTGTC RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCGACAGA
Forward primer CAGAGTGCAGACCAGGGTCTTCATG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
cfa-miR-10a TACCCTGTAGATCCGAATTTGT RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCACAAAT
Forward primer ACAAATTCGGATCTACAGGGTATGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
chi-miR-411b-3p TATGTCACATGGTCCACTAAT RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCATTAGT
Forward primer ATTAGTGGACCATGTGACATAGGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
efu-miR-181a AACCATCGACCGTTGATTGTACC RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCGGTACA
Forward primer ATCAACGGTCGATGGTTTGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
oar-miR-10a TACCCTGTAGATCCGAATTTG RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCCAAATT
Forward primer CAAATTCGGATCTACAGGGTATGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
ssa-miR-144-5p GGATATCATCATATACTGTAAGTT RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCAACTTA
Forward primer CAGTATATGATGATATCCGGCAACGCG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
unconservative_CM008019.1_137729 CCCAGGGATGTAGCTCCTAGTGC RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCGCACTA
Forward primer AGCTACATCCCTGGGTTATGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
unconservative_CM008022.1_159134 CAAATTCGTGAAGCGTTCCATATTT RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCAAATAT
Forward primer GAACGCTTCACGAATTTGTGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
unconservative_CM008039.1_315920 CGGATCAGCTCAGTGCCGGGC RTP GGCCAAGCAGTGGTATCAACGCAGAGTGGCCGCCCGG
Forward primer CACTGAGCTGATCCGAATTGCGTG
Reverse primer AAGCAGTGGTATCAACGCAGAGT
DOI: 10.7717/peerj.13947/table-1

Notes:

RTP

Primer used in reverse transcription

Results

MiRNA sequencing

To determine miRNA function during velvet antler RGS, we constructed three small RNA libraries for antler growth centers at 30, 60, and 90 d, generating 14,971,388, 16,985,192, and 15,732,412 raw reads, respectively.

After data preprocessing, we obtained 9,836,383, 13,092,918, and 10,769,127 clean reads for 30-, 60-, and 90-d antler tips, respectively. We then aligned clean reads with Silva, GtRNAdb, Rfam, and Repbase databases using Bowtie (Langmead et al., 2009), a comparison software for short sequences, especially reads generated from highlight sequencing. After filtering out sequences that matched ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), other ncRNAs, and repeats, we obtained 8,646,311 (30 d), 11,699,869 (60 d), and 9,461,208 (90 d) unannotated reads that only contained miRNAs.

For miRNA prediction, the remaining reads were aligned to the deer genome (https://www.ncbi.nlm.nih.gov/genome/?term=red+deer), with no mismatch allowed between unannotated reads and the reference sequence. Respectively, 5,430,521 (62.81%), 7,081,757 (60.52%), and 5,621,148 (59.41%) reads from 30-, 60-, and 90-d antler cartilage tissues were successfully mapped and annotated (Table 2).

Table 2:
Data of miRNA-seq.
Statistic for deep-sequencing results generated from antler cartilage tissues of Cervus elaphus kansuensis. miRNA: microRNA.
30 d 60 d 90 d
Raw_reads 14971388 16985192 15732412
Clean_reads 9836383 13092918 10769127
rRNA 854250 996335 932966
scRNA 0 0 0
snRNA 3 4 4
snoRNA 21446 18939 15955
tRNA 188055 239173 237900
Repbase 126318 138598 121094
Unannotated Reads 8646311 11699869 9461208
Mapped_Reads 5430521 7081757 5621148
Known miRNAs 1003 1052 1021
Novel miRNAs 184 190 183
DOI: 10.7717/peerj.13947/table-2

Identification of known and novel miRNAs in velvet antler

To identify miRNAs in the three libraries, all mapped reads that were 18–35 nucleotides long were compared with mature animal miRNAs in the miRBase (v21.0) database. Perfectly matched sequences were considered as known miRNAs, while the remaining sequences were designated as novel miRNAs. The analysis identified 1,073 known miRNAs (1,003, 1,052, and 1,021 from 30-, 60-, and 90-d antler tips) and 204 novel miRNAs (184, 190, and 183 from 30-, 60-, and 90-d antler tips). The size distribution of identified miRNAs was similar in all samples, and most of them changed from 20 to 24 nt, which was consistent with the typical size range of miRNAs (Fig. 1; Table S1). The most abundant size class was 22 nt, which accounted for approximately 38.92%, 39.29% and 39.53% in the three libraries. All miRNAs were classified into 188 miRNA families, with the mir-2284 family being the most represented (Table S2).

miRNAs length distribution.

Figure 1: miRNAs length distribution.

The expression levels of each miRNA were normalized to the expression of tags per million (TPM) (Table S3). The top 20 known miRNAs with the highest number of reads (Table 3) accounted for 82.36%, 74.23%, and 76.69% of total reads from 30-, 60-, and 90-d antler cartilage tissues, respectively. The top 20 novel miRNAs also gathered the most reads (Table 4), accounting for 69.36%, 70.27%, and 79.65% of total reads from 30-, 60-, and 90-d antler cartilage tissues, respectively. Novel miRNAs had considerably lower sequencing frequencies than known miRNAs (Table S3). The same pattern has also been reported in other species, suggesting that novel miRNAs are typically weakly expressed and known miRNAs are highly expressed in different organisms.

Table 3:
Top 20 known miRNAs.
The 20 most abundant known miNRAs in antler cartilage of Cervus elaphus kansuensis.
30 d 60 d 90 d
miRNA Count TPM miRNA Count TPM miRNA Count TPM
ggo-miR-148a 755406 220514.4 ggo-miR-148a 781385 181034.9 ggo-miR-148a 704227 206292
aca-miR-148a-3p 750540 219093.9 aca-miR-148a-3p 778239 180306 aca-miR-148a-3p 700427 205178.9
bta-miR-21-5p 215228 62828.29 bta-miR-21-5p 198752 46047.78 bta-miR-21-5p 164868 48295.45
chi-miR-21-5p 215227 62828 chi-miR-21-5p 198752 46047.78 chi-miR-21-5p 164866 48294.86
sha-miR-21 215110 62793.85 sha-miR-21 198623 46017.9 sha-miR-21 164771 48267.03
aca-miR-21-5p 215106 62792.68 aca-miR-21-5p 198623 46017.9 aca-miR-21-5p 164766 48265.57
ccr-let-7i 65670 19170.06 ccr-let-7i 114404 26505.65 ccr-let-7i 74011 21680.34
aca-let-7i-5p 65670 19170.06 aca-let-7i-5p 114404 26505.65 aca-let-7i-5p 74011 21680.34
ssc-let-7i 65670 19170.06 ssc-let-7i 114404 26505.65 ssc-let-7i 74010 21680.05
gga-let-7g-5p 38050 11107.37 gga-let-7g-5p 55889 12948.62 aca-miR-27b-3p 41660 12203.63
aca-let-7g 37827 11042.27 aca-let-7g 55593 12880.04 bta-miR-27b 41660 12203.63
aca-let-7f-5p 26320 7683.204 aca-miR-27b-3p 53525 12400.92 gga-let-7g-5p 39633 11609.85
ggo-let-7f 26320 7683.204 bta-miR-27b 53525 12400.92 aca-let-7g 39451 11556.54
aca-miR-27b-3p 25840 7543.085 aca-miR-99a-5p 49497 11467.69 aca-let-7f-5p 28763 8425.661
bta-miR-27b 25840 7543.085 bta-miR-99a-5p 49497 11467.69 ggo-let-7f 28762 8425.368
aca-let-7a-5p 16504 4817.766 oar-miR-99a 49497 11467.69 aca-miR-99a-5p 22479 6584.864
prd-let-7-5p 16495 4815.139 aca-let-7f-5p 39435 9136.484 bta-miR-99a-5p 22479 6584.864
bta-miR-199b 15746 4596.495 ggo-let-7f 39435 9136.484 oar-miR-99a 22479 6584.864
ssc-miR-199b-5p 15745 4596.203 aca-let-7a-5p 30258 7010.314 aca-let-7a-5p 22389 6558.5
aca-miR-99a-5p 13211 3856.49 prd-let-7-5p 30254 7009.387 prd-let-7-5p 22382 6556.449
DOI: 10.7717/peerj.13947/table-3
Table 4:
Top 20 novel miRNAs.
The 20 most abundant novel miRNAs in antler cartilage of Cervus elaphus kansuensis.
30 d 60 d 90 d
miRNA Count TPM miRNA Count TPM miRNA Count TPM
CM008037.1_299589 1225 357.596 CM008037.1_299589 1537 356.0993 CM008037.1_299589 1135 332.4801
CM008025.1_188962 504 147.1252 CM008025.1_188962 597 138.3157 CM008012.1_55433 632 185.1343
CM008041.1_334512 326 95.16431 CM008022.1_160011 417 96.61249 CM008041.1_329491 631 184.8414
CM008022.1_160011 220 64.22131 CM008027.1_215935 283 65.56675 CM008037.1_302513 631 184.8414
CM008008.1_9244 137 39.99236 CM008041.1_334512 275 63.71327 CM008025.1_188962 541 158.4773
CM008021.1_154122 121 35.32172 CM008021.1_154122 265 61.39643 CM008022.1_160011 316 92.56715
CM008029.1_241215 118 34.44598 CM008021.1_149700 182 42.1666 CM008034.1_284828 287 84.07206
CM008021.1_149700 113 32.9864 CM008029.1_241215 164 37.99628 CM008041.1_334512 264 77.33458
CM008027.1_215935 104 30.35917 CM008008.1_9244 120 27.80216 CM008027.1_215935 154 45.11184
CM008020.1_143075 93 27.1481 CM008026.1_198184 97 22.47341 CM008029.1_241215 139 40.71783
CM008027.1_214244 86 25.1047 CM008041.1_329491 85 19.69319 CM008008.1_9244 125 36.61675
CM008041.1_332283 82 23.93704 CM008037.1_302513 85 19.69319 CM008021.1_149700 119 34.85915
CM008022.1_159134 71 20.72597 CM008012.1_55433 85 19.69319 CM008021.1_154122 84 24.60646
CM008025.1_184228 71 20.72597 CM008041.1_332283 82 18.99814 CM008021.1_147678 66 19.33365
CM008012.1_63603 60 17.5149 CM008019.1_131447 77 17.83972 CM008020.1_143075 65 19.04071
CM008026.1_198184 53 15.4715 CM008027.1_214244 71 16.44961 CM008018.1_117444 63 18.45484
CM008020.1_147456 51 14.88767 CM008012.1_63603 66 15.29119 CM008012.1_63603 60 17.57604
CM008016.1_98716 50 14.59575 CM008016.1_98716 65 15.0595 CM008025.1_184228 58 16.99017
CM008021.1_151843 43 12.55235 CM008036.1_292916 60 13.90108 CM008022.1_159134 57 16.69724
CM008021.1_150571 43 12.55235 CM008020.1_147456 58 13.43771 CM008027.1_214244 51 14.93964
DOI: 10.7717/peerj.13947/table-4

There were 1,128 unique miRNAs co-expressed in 30- (95.03%), 60- (90.82%), and 90-d (93.69%) antler cartilage tissues (Fig. 2). The most abundant miRNA was ggo-miR-148a, then aca-miR-148a-3p, bta-miR-21-5p, chi-miR-21-5p, and sha-miR-21 (Table 3). In addition, eight (0.67%), 30 (2.42%), and 11 (0.91%) miRNAs were specifically expressed (SEMs) in each sample (Fig. 2; Table 5).

Venn diagram of all miRNAs.

Figure 2: Venn diagram of all miRNAs.

Table 5:
Specifically expressed miRNAs.
The specifically expressed miNRAs in antler cartilage of Cervus elaphus kansuensis.
30 d 60 d 90 d
miRNA Count TPM miRNA Count TPM miRNA Count TPM
aja-miR-3120 2 0.6102 aga-miR-10 3 0.7211 bta-miR-105a 1 0.2779
bta-miR-3154 1 0.3051 bma-miR-92 1 0.2524 cgr-miR-128-5p 1 0.2779
cgr-miR-23a-5p 1 0.2912 bta-miR-154a 1 0.2294 dre-miR-181b-3p 1 0.2906
cgr-miR-29b-5p 2 0.5339 bta-miR-202 1 0.2404 eca-miR-302d 1 0.2779
eca-miR-421 1 0.3051 bta-miR-2366 1 0.2294 hsa-miR-181b-2-3p 1 0.3196
CM008010.1_21055 6 1.8306 bta-miR-490 3 0.6883 mdo-miR-218-2-3p 1 0.3044
CM008025.1_186565 2 0.5825 bta-miR-544a 1 0.2294 mml-miR-7178-5p 1 0.2906
CM008038.1_307839 8 2.2285 bta-miR-767 1 0.2195 mmu-miR-128-1-5p 1 0.3044
bta-miR-885 5 1.1472 CM008015.1_78022 10 3.1962
cfa-miR-377 1 0.2294 CM008020.1_145882 4 1.1117
cgr-miR-103-5p 1 0.2103 CM008027.1_215433 5 1.4528
cgr-miR-505-5p 2 0.4389
chi-miR-324-3p 1 0.2404
chi-miR-33b-3p 1 0.2404
chi-miR-411b-3p 9 2.1633
chi-miR-491-3p 1 0.2294
gga-miR-103-2-5p 1 0.2195
hsa-miR-324-3p 1 0.2524
hsa-miR-34a-3p 1 0.2294
mdo-miR-34a-3p 1 0.2294
mml-miR-3059-5p 1 0.2294
mmu-miR-324-3p 1 0.2524
oan-miR-145-5p 1 0.2657
oar-miR-544-3p 1 0.2404
ssa-miR-144-3p 2 0.5048
CM008012.1_49180 2 0.4807
CM008012.1_53957 2 0.4589
CM008022.1_167031 2 0.4807
CM008025.1_186278 2 0.4807
CM008039.1_315920 13 3.1248
DOI: 10.7717/peerj.13947/table-5

Differential expression analysis of miRNAs

Differential expression analysis (| log2 fold change [FC] values | ≥ 1 and P ≤ 0.05) revealed 243 differentially expressed miRNAs (DEMs) across the three growth stages in this study (Tables S4S5). Pairwise comparisons of 30 d vs. 60 d, 30 d vs. 90 d, and 60 d vs. 90 d revealed 177 (136 upregulated and 41 downregulated), 116 (97 upregulated and 19 downregulated), and 85 (45 upregulated and 40 downregulated) DEMs, respectively. Venn diagrams of DEMs in the three pairwise comparisons are presented in Fig. 3.

Venn diagram of differentially expressed miRNAs.

Figure 3: Venn diagram of differentially expressed miRNAs.

To identify key miRNAs controlling rapid antler growth, we selected 25 DEMs that had different expression patterns in the 30 d vs. 60 d and 60 d vs. 90 d comparisons only (Table 6). These are the miRNAs that likely play important roles in antler development during RGS at day 60. Among the 25 DEMs, cfa-miR-146b has the highest expression in 60 d, followed by cgr-miR-146b-5p, and bta-miR-146b, bta-miR-27a-5p, and cgr-miR-181a-3p. Two DEMs were specifically expressed in 60 d antler cartilage tissue, namely chi-miR-411b-3p and CM008039.1_315920. We were more interested in the potential biological function of novel miRNA CM008039.1_315920.

MiRNA target prediction and functional annotation of candidate target genes

MiRNAs usually act via translational repression and/or mRNA cleavage, but some evidence shows that they can also upregulate translation through diverse mechanisms. The rapid growth stage is a special period in the growth process of antler velvet; thus, we pay more attention to miRNAs in 60d. In this study, the target genes of 25 DEMs, 30 miRNAs specifically expressed in 60 d and novel miRNA CM008039.1_315920 were predicted. In particular, 726, 1,759 and 139 targets were predicted for 25 selected DEMs, specifically expressed miRNAs in 60 d and CM008039.1_315920, respectively. We then functionally annotated candidate target genes by using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses.

The GO analysis showed that 726 target genes of the 25 DEMs were classified into 52 functional subcategories. “cellular process” (431 genes), “cell part” (487 genes) and “cell” (486 genes), and “binding” (397 genes) were the top enriched GO terms under the biological process (BP), cellular component (CC), and molecular function (MF) categories, respectively (Fig. 4A). For the specifically expressed miRNAs in 60 d, 1,759 target genes were classified into 53 functional subcategories. Similarly, “cellular process” (1,028 genes) “cell” (1,169 genes) and “binding” (943 genes) were the top enriched GO terms under BP, CC, and MF (Fig. 4B). For novel miRNA CM008039.1_315920, 139 target genes were classified into 44 functional subcategories. “cellular process” (89 genes), “cell part” (98 genes) and “binding” (81 genes) were also the top enriched GO terms under BP, CC, and MF (Fig. 4C).

The KEGG classification showed that target genes of the 25 DEMs were mainly associated with the “Metabolic pathways”, followed by pathways for “Pathways in cancer”, and “PI3K-Akt signaling pathway” (Fig. 5A). Similarly, most target genes of the specifically expressed miRNAs in 60 d were related to the “Metabolic pathways”, followed by pathways for “PI3K-Akt signaling pathway” and “Pathways in cancer” (Fig. 5B). For novel miRNA CM008039.1_315920, most targets were classified to “Metabolic pathways”, as well as pathways of “Gap junction”, “Oocyte meiosis”, “PI3K-Akt signaling pathway”, “Rap1 signaling pathway”, “Ras signaling pathway”, “Proteoglycans in cancer” and “Progesterone-mediated oocyte maturation” (Fig. 5C). KEGG enrichment of miRNA target genes was also analyzed (Fig. S1). Notably, among predicted targets of the 25 DEMs, we discovered that CM008039.1_315920 miRNA was highly enriched in “NF-kappa B signaling pathway” (Fig. S1C), which was mainly associated with apoptosis and cell proliferation.

Table 6:
Selected 25 DEMs that may participate in antler rapid growth.
#ID miRNA_Seq Length 30 d 60 d 90 d 30 d_vs_60 d 30 d_vs_90 d 60 d_vs_90 d
1 aca-miR-10a-5p UACCCUGUAGAUCCGAAUUUGUG 23 6.12849554 17.99627 4.724809 Up Normal Down
2 aca-miR-144-5p GGAUAUCAUCAUAUACUGUAA 21 1.83058958 12.97989 5.174791 Up Normal Down
3 aca-miR-146a-5p UGAGAACUGAAUUCCAUAGGC 21 1.52549131 7.932154 1.521997 Up Normal Down
4 aga-miR-184 UGGACGGAGAACUGAUAAGGG 21 0.30509826 5.047734 0.608799 Up Normal Down
5 bta-miR-146b UGAGAACUGAAUUCCAUAGGCUGU 24 17.8863856 41.85413 12.78478 Up Normal Down
6 bta-miR-187 UCGUGUCUUGUGUUGCAGCCGG 22 1.4561508 5.277177 1.452816 Up Normal Down
7 bta-miR-27a-5p AGGGCUUAGCUGCUUGUGAGCA 22 54.75127 26.15644 68.5729 Down Normal Up
8 bta-miR-504 AGACCCUGGUCUGCACUCUGUC 22 0.29123016 3.67108 0.581126 Up Normal Down
9 cfa-miR-10a UACCCUGUAGAUCCGAAUUUGU 22 6.40706351 18.81428 4.939573 Up Normal Down
10 cfa-miR-146b UGAGAACUGAAUUCCAUAGGCU 22 19.5124207 45.65905 13.94703 Up Normal Down
11 cgr-miR-146b-5p UGAGAACUGAAUUCCAUAGGCUG 23 18.6640546 43.67388 13.34064 Up Normal Down
12 cgr-miR-181a-3p ACCAUCGACCGUUGAUUGUACC 22 41.0634525 20.19094 42.13165 Down Normal Up
13 cgr-miR-187 UCGUGUCUUGUGUUGCAGCCG 21 1.52549131 5.528471 1.521997 Up Normal Down
14 chi-miR-187 UCGUGUCUUGUGUUGCAGCC 20 1.60176588 5.804894 1.598097 Up Normal Down
15 chi-miR-411b-3p UAUGUCACAUGGUCCACUAAU 21 0 2.163315 0 Up Down
16 chi-miR-504 AGACCCUGGUCUGCACUCUGU 21 0.30509826 3.845893 0.608799 Up Normal Down
17 efu-miR-181a AACCAUCGACCGUUGAUUGUACC 23 39.278085 19.31307 40.29984 Down Normal Up
18 mdo-miR-181a-1-3p CCAUCGACCGUUGAUUGUACC 21 42.7137568 20.67167 43.22472 Down Normal Up
19 oar-miR-10a UACCCUGUAGAUCCGAAUUUG 21 7.01726004 19.95057 5.174791 Up Normal Down
20 sha-miR-181a-3p ACCAUCGACCGUUGAUUGU 19 47.5471556 23.37898 48.78402 Down Normal Up
21 ssa-miR-144-5p GGAUAUCAUCAUAUACUGUAAGUU 24 1.60176588 11.3574 4.527942 Up Normal Down
22 CM008019.1_137729 CCCAGGGAUGUAGCUCCUAGUGC 23 1.39283989 4.389334 1.11172 Up Normal Down
23 CM008022.1_159134 CAAAUUCGUGAAGCGUUCCAUAUUU 25 18.1960604 6.259191 14.57465 Down Normal Up
24 CM008025.1_184228 CAAAUUCGUGAAGCGUUCCAUAUUU 25 18.1960604 6.259191 14.83034 Down Normal Up
25 CM008039.1_315920 CGGAUCAGCUCAGUGCCGGGC 21 0 3.124788 0 Up Down
DOI: 10.7717/peerj.13947/table-6

Quantitative real-time PCR (qRT-PCR) verification

To validate DEM expression profiles, we randomly selected 13 miRNAs, including 10 known and three novel miRNAs, for stem-loop qRT-PCR. We discovered that qRT-PCR results and high-throughput sequencing data were correlated (Fig. 6), validating DEM expression patterns and confirming the reliability and accuracy of our miRNA-seq data.

Discussion

The velvet antler is capable of regeneration and rapid growth, processes that are inseparable from the self-renewal and differentiation ability of mesenchymal tissue in the antler growth center. These characteristics differentiate the antler from normal cartilage or bone tissue (Li et al., 2014). The growth center (mainly comprising velvet skin, mesenchyme, and cartilage) controls velvet antler regeneration and development (Li et al., 2002; Ba et al., 2019). Antler growth centers are rich in regulatory factors, including growth and transcription factors related to cartilage growth and ossification (Yao et al., 2018). Multiple studies have reported that let-7a and let-7f (Hu et al., 2014a), miR-18a (Hu et al., 2014b), miR-19a/b (Yan et al., 2020), and miR-15a/b (Liu et al., 2018) participate in the regulation of antler chondrocyte proliferation and regeneration. Therefore, discovering novel regulators and their associated pathways will help us understand the mechanisms involved in velvet antler RGS.

Here, we investigated and compared miRNA profiles of antler cartilage tissues at three growth stages (30, 60, and 90 d), revealing several DEMs and candidate target genes that are associated with the rapid growth of velvet antlers.

This study predicted 1,277 miRNAs, including 1,073 known and 204 novel miRNAs. Among these, 243 were differentially expressed across the three growth stages. We screened 25 DEMs from the 30 d vs. 60 d and 60 d vs 90 d comparisons, including 21 known and four novel miRNAs; 18 of these were upregulated and seven were downregulated in 60 d antler cartilage tissue. MicroRNA (miRNA) is evolutionarily conserved non-coding small RNA molecule between species. Since no miRNA sequence data of C. elaphus kansuensis was available in the miRBase v21.0 database, the miRNAs in this study were extrapolated from other species. This method was also used in other studies (Ba, Wang & Li, 2016).

GO classification for target genes of 25 DEMs.

Figure 4: GO classification for target genes of 25 DEMs.

For target genes of 25 DEMs, specifically expressed miRNAs in 60 d and CM008039.1_315920, the GO terms of “cellular process,” “cell part,” “cell,” and “binding” were clustered the most target genes. (A) GO classification of target genes for 25 DEMs; (B) GO classification of target genes for specifically expressed miRNAs in 60 d; (C) GO classification of target genes for novel miRNA CM008039.1_315920.
KEGG classification for target genes of 25 DEMs.

Figure 5: KEGG classification for target genes of 25 DEMs.

The ordinate represents the KEGG pathways, and the abscissa represents the number and proportion of target genes annotated to the pathway. For target genes of 25 DEMs, specifically expressed miRNAs in 60 d and CM008039.1_315920, most target genes annotated to pathways of “Metabolic pathways”, “PI3K-Akt signaling pathway” and “Proteoglycans in cancer”. (A) KEGG classification for target genes of all 243 DEMs. (B) KEGG classification for target genes of specifically expressed miRNAs in 60 d; (C) KEGG classification for target genes of CM008039.1_315920.
qPCR of DEMs.

Figure 6: qPCR of DEMs.

Two DEMs (CM008039.1_315920 and chi-miR-411b-3p) with the lowest expression levels among 25 DEMs were specifically expressed in 60 d antler cartilage. Through suppressing target gene expression, miRNAs are known to alter expression patterns in specific tissues at specific times (Mohr & Mott, 2015). However, no studies have investigated the role of mir-411b in chondrogenesis. Thus, further research on these two miRNAs is required to determine their biological functions during the RGS of antler development.

Bioinformatics analysis revealed that the predicted target genes of 25 DEMs and specifically expressed miRNAs in 60d were widely classified into multiple GO terms, indicating that these genes may participate in many important biological processes. The GO enrichment results showed that most target genes were highly enriched in the “cellular process”, “cell”, “cell part”, and “binding” terms, implying involvement in antler development. The velvet antler may grow at a speed of two cm/d during RGS, a period of rapid cell proliferation, differentiation, and aggregation. The amount of ATP, nucleotides, proteins, and other factors involved in velvet antler growth may be able to support vigorous cell activity of antler, such as proliferation and differentiation.

Unlike the GO analysis, the KEGG pathway analysis yielded more direct and detailed results. The pathways represent protein-protein interactions and any change in each pathway reflects changes in a specific protein’s expression or activity. KEGG pathway analysis revealed that most target genes of 25 DEMs, specifically expressed miRNAs in 60d and CM008039.1_315920 were mainly associated with “Metabolic pathways”, “PI3K-Akt signaling pathway” and “Proteoglycans in cancer”. Metabolic pathways have extensive contents, such as glycolysis, fatty acid oxidation, and amino acid metabolism (Boroughs & DeBerardinis, 2015). These pathways play important roles in cells fate and functions (Boroughs & DeBerardinis, 2015; Kim & DeBerardinis, 2019) and determine vasculature formation (Li, Kumar & Carmeliet, 2019). Antler cartilage is rich in nerves and blood vessels, and there is a definite link between the abundant blood vessels and the rapid growth and regeneration of antlers (Clark et al., 2006; Hu et al., 2019). In summer, velvet antler grows rapidly, and a large number of blood vessels in antler can transport nutrients to and excrete metabolites from the antler. It has been reported that there may be a close relationship between metabolites and the size of velvet antlers (Su et al., 2020). Thus, metabolic pathways play important roles in rapid growth and regeneration of velvet antler.

PI3K-Akt pathway can regulate multiple cellular events including cell apoptosis, proliferation and differentiation. Moreover, PI3K-Akt pathway is frequently involved in cancers (Jiang et al., 2020). It can be activated by Wnt signaling pathway and there is complex crosstalk between PI3K-Akt and Wnt/β-catenin pathways (Dong et al., 2020). These two signaling pathways can function synergistically (Evangelisti et al., 2020; Prossomariti et al., 2020). PI3K-Akt pathway also participates in glucose and glutamine metabolism within the hierarchy of pathways altered in cancer (Boroughs & DeBerardinis, 2015).

Proteoglycans are a group of molecules, which play significant roles in cancer development, progression, invasion and metastasis (Wei et al., 2020). Whether the pathway of “Proteoglycans in cancer” participates in antler rapid growth and regeneration needs further investigation.

Target genes associated with the above signaling pathways may also be involved with the well-known Wnt signaling pathway, widely distributed in invertebrates and vertebrates. This pathway mediates important cellular responses, including cancer development, body axis development, and morphogenesis. Wnt signaling also plays a crucial role in the early development of animal embryos, organ formation, tissue regeneration, and other normal physiological processes (Leucht, Lee & Yim, 2019). Previous studies have reported that the Wnt signaling pathway participates in the regulation of antler growth and development (Mount et al., 2006; Li et al., 2012; Zhang et al., 2018). Some researchers believe that antler growth is a tumor-like development due to their similar growth rates (Goss, 1990; Kierdorf & Kierdorf, 2011; Wang et al., 2017), suggesting a possible mechanism connecting the miRNAs detected in this study and antler development. Indeed, numerous genes are involved in both cancer and antler development. However, further studies are required to identify the specific genes acting on antler growth and development during RGS.

KEGG pathway analysis also revealed that CM008039.1_315920 miRNA was abundant in the NF- κB pathway. NF- κB is a ubiquitous transcription factor that regulates the expression of genes involved in multiple cell functions; it is activated by various extracellular stimuli (Caviedes et al., 2017). NF- κB plays an important role during immune response and inflammation. However, growing evidence suggests that NF- κB also has a major role in oncogenesis through regulating the expression of genes involved in cancer development and progression, such as tumor cell proliferation, migration, and apoptosis (Dolcet et al., 2005). Therefore, further investigation is required to confirm the function of this novel miRNA in velvet antler development.

We screened 25 DEMs that are potentially important for antler development from the 30 d vs. 60 d and 60 d vs. 90 d comparisons. Through qRT-PCR, we confirmed that the expression levels of 13 randomly selected miRNAs were consistent with the bioinformatics analysis.

Taken together, our findings provide new insights into the possible mechanisms involved in antler development during the RGS. Further research on the 25 DEMs and specifically expressed miRNAs in 60 d identified here, especially the novel miRNA CM008039.1_315920, will be helpful in elucidating mechanisms underlying the rapid growth of deer antlers and may also contribute novel therapeutic targets for cancer treatment.

Conclusions

In this study, the novel miRNA CM008039.1_315920 was differentially expressed in the 30 d vs. 60 d and 60 d vs. 90 d comparisons, but not in the 30 d vs. 90 d comparison. This miRNA was also a specifically expressed miRNA in deer antler cartilage that grew for about 60 d. Moreover, the target genes of this novel miRNA were annotated to NF- κB pathway, indicating that CM008039.1_315920 may possess important biological functions in rapid antler growth.

Supplemental Information

Author Checklist - Full

DOI: 10.7717/peerj.13947/supp-1

MiRNAs length distribution

DOI: 10.7717/peerj.13947/supp-2

MiRNA family clusters

DOI: 10.7717/peerj.13947/supp-3

Log2fold changes of all miRNAs

DOI: 10.7717/peerj.13947/supp-5

Raw data for qRT-PCR

DOI: 10.7717/peerj.13947/supp-7

KEGG enrichment for target genes of 25 DEMs

DOI: 10.7717/peerj.13947/supp-8

KEGG enrichment for target genes of SEMs in 60 d

DOI: 10.7717/peerj.13947/supp-9

KEGG enrichment for target genes of selected novel miRNAs

DOI: 10.7717/peerj.13947/supp-10
1 Citation   Views   Downloads