Genome-wide identification and characterization of long non-coding RNAs in Longissimus dorsi skeletal muscle of Shandong black cattle and Luxi cattle

DOI: https://doi.org/10.21203/rs.3.rs-120327/v1

Abstract

Background

There is increasing understanding of the possible regulatory role of long non-coding RNAs (LncRNA). Studies on livestock have mainly focused on the regulation of cell differentiation, fat synthesis, and embryonic development. However, there has been little study of skeletal muscle of domestic animals and the potential role of lncRNA.

Results

RNA samples were collected from longissimus dorsi muscle samples of Shandong black cattle and Luxi cattle and libraries were constructed and sequenced. A total of 1415 transcripts (of which 480 were LncRNAs) were differentially expressed (P < 0.05) in the different breeds, and fourteen of these RNAs were randomly selected and validated by qPCR. We found that the most differentially expressed LncRNAs were found on chromosome 9, with 1164 within 50 kb of a protein-coding gene. In addition, Pearson's correlation coefficients of co-expression levels indicated a potential trans regulatory relationship between the differentially expressed LncRNAs and 43844 mRNAs (r > 0.9). The identified co-expressed mRNAs (MYORG, Dll1, EFNB2, SOX6, MYOCD, and MYLK3) are related to the formation of muscle structure, and enriched in muscle system process, strained muscle cell differentiation, muscle cell development, striated muscle tissue development, calcium signaling, and AMPK signaling. Additionally, we also found that some LncRNAs (LOC112444238, LOC101903367, LOC104975788, LOC112441863, LOC112449549, and LOC101907194) may interact with miRNAs related to cattle muscle growth and development. Based on this, we constructed a LncRNAs-miRNA-mRNA interaction network as the putative basis for biological regulation in cattle skeletal muscle. Interestingly, a candidate differential LncRNA (LOC104975788) and a protein-coding gene (Pax7) contain miR-133a binding sites and binding was confirmed by luciferase reporter assay. LOC104975788 may bind miR-133a competitively with Pax7, thus relieving the inhibitory effect of miR-133a on Pax7 to regulate skeletal muscle development. These results will provide the theoretical basis for further study of LncRNA regulation and activity in different cattle breeds.

Conclusions

The data obtained in this study were used to predict muscle-related LncRNAs-miRNA-mRNA interaction networks, which can help elucidate the molecular mechanism of cattle muscle development. These results can be used to facilitate livestock breeding and improve livestock production.

Background

As an important economic trait that affects the production efficiency of beef cattle, meat production traits are a research focus in the field of beef cattle genetics and breeding. The analysis of the molecular regulation mechanisms of muscle growth can facilitate cattle breeding. Local cattle breeds in China have good meat quality but slow growth is slow, while imported cattle typically grow more quickly, but the meat quality is poor. These differences may be related to differences in skeletal muscle development among different breeds. The growth and development of beef cattle skeletal muscle can be affected by a variety of regulatory factors. Previous work mainly focused on contributions from DNA, mRNA, and miRNA, but regulatory effects of long non-coding RNA (LncRNA) on the growth and development of beef skeletal muscle remain poorly understood.

Previous studies reported contributions of LncRNA in the process of skeletal muscle proliferation and differentiation1,2. Liu identified, mapped, and determined the global and skeletal muscle expression patterns of 7188 LncRNAs, finding that these LncRNAs had similar open reading frames and expression levels as those of other mammalian LncRNAs. Subsequently, Yue identified a highly expressed LncRNA in muscle tissue, LncRNA-YYW. Microarray analysis showed that LncRNA-YYW positively regulated the expression of growth hormone-1 and its downstream genes AKT1 and PIK3CD in bovine myoblasts, and promoted myoblast proliferation 3. RNA samples from the longissimus pectoralis muscle of Limousin beef cattle were subjected to paired-end RNA sequencing, and some of the identified expressed LncRNAs mapped to quantitative trait loci of meat quality traits4. These results suggest that LncRNA may regulate the growth and development of beef skeletal muscle and meat quality. Although LncRNAs lack coding capacity, many LncRNAs act in various biological processes, serving as an additional level of genomic regulation.

The first embryo transfer calf in China was obtained from a vitrified frozen somatic cell cloned embryo, which combined Japan black cattle, Luxi cattle, and Bohai black cattle. The new generation core breeding group of this new breed, Shandong black cattle, was established by filial generation. The new generation core breeding group hybridized with Shandong black cattle, and the second generation group of Japan black cattle (3/4) was crossed with luxi cattle (1/4) to produce the new generation (3/4) × bohai black cattle (1/4). Ideal black cattle and Shandong black cattle were selected from the second-generation group for cross-cross fixation. In this stage, several good lines were cross-bred with distant relatives, and then selected and the excellent lines were preserved. After four generations, this line was finally bred into a new Shandong black cattle variety matching line and used as a bull. In 2015, the Shandong black cattle variety was approved by the National Animal and Poultry Genetic Resources Committee as a new population and successfully established as a new cultivated variety in China 5,6.

In recent years, there has been growing research of LncRNA, but most work has mainly focused on the effects of neurologic diseases, tumors, embryonic development, and cell differentiation in human and mouse, with little research on domestic animals. The growth and development of beef cattle skeletal muscle are highly regulated processes. Although the contributions of DNA, mRNA, and miRNA to this regulation have been studied, the extent to which LncRNA regulates beef skeletal muscle growth and development remains poorly understood. To investigate the potential contribution of LncRNA, RNA samples were prepared from longissimus dorsi muscle tissues of Shandong black cattle (hybrid offspring) and Luxi Cattle (the first maternal generation). RNA-seq technology was used to identify LncRNA transcripts and their genomic characteristics, and LncRNAs differentially expressed in skeletal muscle tissues in different breeds of beef cattle were determined. Using these data, we constructed an interaction network of LncRNA-miRNA-mRNA and muscle development which can guide future breeding efforts.

Methods

  1. Ethics statement

The methods used in this study were performed in accordance with the guidelines of Good Experimental Practices adopted by the Institute of Animal Science (Qingdao

Agricultural University, Qingdao, China). All surgical procedures involving cattle were performed according to the approved protocols of the Biological Studies Animal Care and Use Committee, Shandong Province, China.

  1. Animal and tissue preparation

Shandong black cattle and Luxi cattle were used in this study and were purchased from Shandong Black Cattle Technology Co., Ltd.. Cattle were fed three times and received approximately equal amounts of green coarse feed and concentrate feed according to standard NY5127—2002 pollution-free feeding management of beef cattle. Cattle were supplied sufficient drinking water. Six 18-month-old healthy beef cattle were randomly selected for this study. The selected cattle had no scratches, scars, and scabs on their bodies, with no fat deposits in the internal organs or abdomen. No disease was found during examination, and all physiological and biochemical indexes were normal. Longissimus dorsi muscle samples were collected and immediately frozen in liquid nitrogen for RNA extraction. The cattle after the experiment were euthanized. First, a 1-3% sodium pentobarbital solution was prepared with physiological saline, and then intravenously injected. The injection dose is 90~135mg/kg.

  1. Hematoxylin and eosin (HE) staining of muscle tissue and fast/slow muscle fiber fluorescence staining

Paraffin sections were prepared from muscle tissue fixed with 4% paraformaldehyde. The HE staining protocol was performed as described previously7. Briefly, dewaxing, covering with water, haematoxylin staining, washing with water, 5% acetic acid differentiation, eosin staining, dehydration, natural drying, sealing, and image acquisition were performed.

Tissue sections were placed in a box filled with EDTA antigen repair buffer(Purchased from Shanghai Beyotime Biotechnology Co., Ltd) (ph 8.0) for antigen repair. After natural cooling, the slides were washed in PBS (pH7.4) with three ashes of five minutes each. BSA was added for blocking, and then the antibody was added, followed by DAPI to stain the nucleus. An autofluorescence quenching agent was added to the slices for 5 min and the samples were then washed with running water for 10 min. After natural drying, the film was sealed with an anti-fluorescence quenching sealing agent. Finally, the slices were observed under the fluorescence microscope and images were collected.

Image-Pro Plus software was used to count the images and measure the surface area. SPASS software was used for statistical analysis to determine significant differences.

  1. RNA extraction, library construction, and sequencing

Total RNA samples were isolated using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. RNA degradation and contamination were monitored on 1 % agarose gels. RNA purity was checked using a NanoPhotometer® spectrophotometer (IMPLEN, Los Angeles, CA, USA). RNA concentration was measured using a Qubit® RNA Assay Kit in a Qubit® 2.0 Fluorometer (Life Technologies, Carlsbad, CA, USA). RNA integrity was assessed using a RNA Nano 6000 Assay Kit in a Bioanalyzer 2100 system (Agilent Technologies, Santa Clara, CA, USA). Only samples with RNA Integrity Number (RIN) scores > 8 were used for sequencing. A total of 3 μg RNA per sample was used as the input material for RNA library preparation.

  1. Transcriptome assembly

The original sequencing data were processed by Fastp software with parameters “‘-Q 20 -P90”’ with disjointing sequence and low-quality sequence. Clean data were obtained by removing reads containing adapters, reads containing over 10 % of poly(N), and low-quality reads (>50 % of the bases had Phred quality scores ≤ 10) from the raw data. The clean data were then calculated. All downstream analyses were based on the high quality clean data. The Bos taurus genome reference genome and gene model annotation files were downloaded from the NCBI database (CHIR_1.0, NCBI) [60]. An index of the reference genome was built using Bowtie v2.0.6 [61, 62] and paired-end clean reads were aligned to the reference genome using HISAT v2-2.1.0 [63]. The mapped reads from each library were assembled with Cufflinks v2.2.1 [64], using Cufflinks with ‘min-frags-per-transfrag = 0’ and‘–library-typefr-firststrand’, and other parameters set as default.

  1. Screening of LncRNA

Stringtie was used to sort reads into different classes and then generate a mosaic map for each class. Based on the length of LncRNA, and the characteristics of non-protein coding sequences, we established strict screening conditions to screen LncRNA as follows:

(1) Transcripts equal to or longer than 200 bp in length, and containing two or more exons;

(2) Transcript read coverage of at least five reads;

(3) No transcripts of known mRNA or other specific non-coding RNAs (rRNA, tRNA, snoRNA, or snRNA). This screening was based on gffcompare (http://ccb.jhu.edu/software/stringtie/gff.shtml) using the same species annotation file;

(4) According to the class in the comparison result_ Code information ("U", "I", "X") was used to screen potential lincrna and intronic LncRNA、anti-sense LncRNA.

Coding potential prediction screening:

To further assess if the identified transcripts are LncRNAs, a variety of coding potential analysis software was integrated, including CNCI analysis, CPC analysis, Pfam protein domain analysis, and CPAT analysis (only for animals)8. The transcripts identified as non-coding by several methods were the final Novell LncRNA dataset.

  1. Differential expression analysis

Comparison of FPKM9 (fragments per kilobase of transcript per million mapped reads) for different genes is a very effective tool for quantitative estimation of gene expression based on RNA-Seq data. This method can eliminate the influence of gene length and sequencing quantity on the determination of gene expression levels. The calculated gene expression values allow direct comparison of gene expression differences among different varieties. Here, we used DEseq10to analyze the differential expression between the treatment group with the reference group, and selected |Log2ratio| ≥1 and Q < 0.05 genes as indicative of significant differential expression. The numbers of up-regulated and down-regulated genes were also obtained.

  1. Prediction and analysis of target genes of differential LncRNA

LncRNA can act on target genes by Cis or Trans. Cis effect refers to LncRNA acting on adjacent target genes11. To predict the cis regulatory genes of LncRNA, we screened the 50 kb of sequence upstream and downstream of LncRNA and looked for potential target genes. The coexpression relationship between LncRNA and mRNA was described by Trans. According to the correlation coefficient between LncRNA and mRNA expression (correlation coefficient cor>= 0.9);  WGCNA(Weighted correlation network analysis was used to predict the target genes of lncRNA for cluster analysis and functional enrichment analysis of LncRNA target genes. P-value < 0.05 was set as the significance threshold.

  1. Analysis of enrichment pathway of GO and KEGG

GO (gene ontology) can be used for enrichment analysis of target genes with differential expression and Goseq12was used to analyze the target genes of differentially expressed LncRNA. A value of P < 0.05 is considered as significant enrichment of differentially expressed genes.

The KEGG database describes advanced functions and utility of biological systems such as cells, organisms, and ecosystems (http://www.genome.jp/kegg/). We used KOBAS V3.0 software to detect the enrichment of LncRNA target genes differentially expressed in KEGG pathway.

  1. Prediction of LncRNA-miRNA-mRNA interaction

As competing endogenous RNAs (ceRNA), LncRNA can act as a sponge to adsorb miRNA and affect mRNA expression. Therefore, to further understand the function of LncRNA in the growth and development of skeletal muscle, we used TargetScan(http://www.targetscan.org/vert_71/) and miRanda((http://www.microrna.org/microrna/home.do) software to predict the differentially expressed miRNAs adsorbed by LncRNA and downstream-regulated mRNA. We then constructed an LncRNA-miRNA-mRNA interaction network map using Cytoscape version 3.5.1.

  1. Luciferase reporter assay

Cells were seeded in 96-well plates at a density of 5×103 cells (HEK-293T) per well, 24 h before transfection. The cells were co-transfected with a mixture of 50 ng Firefly luciferase(FL) reporter vectors, 5 ng Renilla luciferase (RL) reporter vectors (pRL-TK), and miRNA mimics at the indicated concentration. The miRNA mimics were obtained from Life Technologies. After 48 h, the luciferase activity was measured with a dual luciferase reporter assay system using the psiCHECK-2 vector (Promega, Madison, WI). The LOC104975788 and LOC536229 (Pax7) sequences were separately cloned into the reporter gene vector (psiCHECK-2) to synthesize the predicted miRNA mimics and control. The potential binding target of each miRNA was cloned into the 3'UTR region of r-luciferase (hrluc), and then co-transfected with the miRNA to determine the activity of R-Luciferase. F-Luciferase (hluc +) was used as an internal reference to correct for differences in transfection efficiency between different samples. The miRNA mimics and psicheck-LOC104975788 or psicheck-LOC536229 (Pax7) were co-transfected into 293T cells. The expression level of reporter genes was detected using a multi-functional enzyme labeling instrument, and the miRNAs that exhibited down-regulated reporter gene expression were further screened.

  1. Validation of sequencing data by quantitative reverse transcription PCR (qRT-PCR)

The reaction system (20 μL) consisted of the following: 1 μL of template cDNA, 10 μL each of the upstream and downstream primers, and 5 ml (5×1 mL vials) of RNase-free water. The thermal cycling procedure was as follows: 94 ℃ for 10 min, 94 ℃ for 30 s, 60 ℃ for 30 s, and 72 ℃ for 40 s, with 40 cycles12.The expression of GAPDH was calculated by the 2-△△CT method. Primers used for qRT-PCR as shown in Supplementary documents Table S1.

Results

To investigate the potential regulatory effects of LncRNA on the growth and development of beef skeletal muscle, we analyzed samples from muscles of different breeds of beef cattle.

  1. Apparent differences in muscle fibres in different breeds of beef cattle

First, we obtained samples from Shandong black cattle and Luxi cattle and performed fluorescence staining sections of fast and slow muscle fibers (Fig. 1a) and HE staining (Fig. 1b). The results showed that significant differences in muscle fibers of longissimus dorsi muscles between Shandong black cattle and Luxi cattle. IPP software analysis showed that the average area of muscle fibres of Shandong black cattle was significantly larger than that of Luxi cattle (P<0.05), with significant differences in the muscle fibre density and the ratio of fast-twitch fibers to slow-twitch fibers and slow-twitch fibers to muscle fiber area (P<0.05), but not in other muscle fibre properties (P>0.05) (Table 1).

Table 1 Comparison of muscle fibre characteristics and growth characteristics of different breeds of cattle

Characteristics

Shandong black cattle

Luxi cattle

Area (μm2)

5490.222±184.649*

4869.008±69.596

Diameter (μm)

106.837±12.537

120.491±4.324

Length (μm)

174.220±7.395

142.435±0.968

Density (Number of muscle fibres/ Muscle fiber area, EA/μm2)

4887.848±373.586**

7172.966±319.501

Number of muscle fibres(EA)

57.667±4.333

39±1.732

Fast-twitch fibers/Slow-twitch fibers

0.412±0.096**

3.280±1.082

Fast-twitch fibers/Muscle fiber area

0.200±0.0342

0.652±0.110

Slow-twitch fibers/Muscle fiber area

0.508±0.046*

0.246±0.072

Weight (kg)

509.667±2.026

489.333±1.764

Note: In the table, * indicates a significant difference (P < 0.05)** indicates a extremely significant difference(P < 0.01).

  1. Overview of sequencing data of longissimus dorsi in beef cattle

RNA was extracted from longissimus dorsi muscle samples of Shandong black cattle and Luxi cattle and sequenced. Totals of 105345396 and 126735736 reads were obtained from Shandong black cattle and Luxi cattle, respectively, with 101145946 and 122463284 mapped reads. In the data for Luxi cattle longissimus dorsi muscle, the percentages of exons, introns, and genes were 43.26%, 41.34%, and 15.4%, respectively, with 38.16%, 48.3%, and 13.54%, respectively, for Shandong black cattle longissimus dorsi muscle tissue, as shown in Fig.2.

  1. Analysis of differential mRNA expression

A total of 20104 transcripts were obtained from the two libraries, including 18859 and 18976 mRNAs for Luxi cattle and Shandong black cattle, respectively. According to the screening criteria of |FC|≥2 and P < 0.05, 1415 differentially expressed mRNA (Table S2) were found, with 939 mRNAs significantly up-regulated and 476 down-regulated in Luxi cattle. The 92 mRNAs were specifically expressed in cattle , with 30 and 62 mRNAs specifically expressed in Shandong black cattle and Luxi Cattle, respectively (Table S2). The top 40 genes with significant differential expression in the two breeds in cattle are listed in Table S3, and suggest genes that could be used for the functional comparison and analysis of differences among varieties.

  1. Functional analysis of differential mRNA

Gene Ontology (GO) is an international standardized classification system of gene function that includes three categories: biological process (BP), molecular function (MF), and cell composition (CC). We performed GO enrichment analysis on 1415 differentially expressed genes (Fig. 3a and b), and the results showed 2662 items (P < 0.05), with 388 were enriched in cell composition (CC), 361 in molecular function (MF), and 1913 in biological process (BP) in up-regulated genes. The enrichment results of down-regulated genes showed 1883 items in total (P < 0.05), with 251 enriched in cell composition (CC), 247 in molecular function (MF), and 1385 in biological process (BP). In the muscle regulation system, muscle contraction is enriched in the first 50 GO terms, indicating that some differential genes may participate in muscle development and function and other life activities. Muscle-related GO terms are shown in Table 2.

Different genes coordinate with each other to perform their biological functions in organisms. We next performed KEGG pathway analysis of the differentially expressed genes to identify the related signal transduction and metabolic pathways (Fig. 3c and d). The results showed enrichment of 939 up-regulated genes in 243 pathways (P<0.05), and enrichment of 476 down-regulated genes in 239 pathways, including metabolism, disease, cell communication and endocrine regulation (Table S4). The enriched pathways (P<0.05) included some signaling pathways related to skeletal muscle growth, development, and function (Table 3), such as the Wnt signaling pathway, the calcium signal transduction pathway, gap junction, the Hippo signaling pathway, the AMPK signaling pathway, and the thyroid hormone synthesis pathway.

  1. Screening of differential LncRNAs

A total of 8427 LncRNAs were found in longissimus dorsi muscle of Shandong black cattle and Luxi cattle. Of these, 3498 LncRNAs were annotated in non-coding regions and the remaining 4929 LncRNAs were in regions that are not annotated. According to the screening criteria of |FC|≥2 and P<0.05, 480 differentially expressed LncRNAs were found, with 245 and 235 LncRNAs significantly up-regulated and down regulated in Luxi Cattle compared to the levels in Shandong black cattle (Table S5). A total of 92 LncRNAs were only expressed in one cattle breed, with 30 specific LncRNAs in Shandong black cattle and 62 specific LncRNAs in Luxi cattle (Table S5). These breed-specific longissimus dorsi muscle LncRNAs included eight annotated LncRNAs in Shandong black cattle and 13 annotated LncRNAs in Luxi cattle, as shown in Table 4. The LncRNAs specifically expressed in Shandong black cattle and Luxi cattle, the first 40 unexplained LncRNAs and annotated LncRNAs are listed in Table S6.

Table 4 LncRNA specially expressed in Shandong Black cattle and Luxi cattle

Breed

Genes

Genomic position

P-value

q-value

Luxi cattle

LOC112447810

chrNC_037335.1:59843881-59851000

0.002587626

0.03702486

LOC112448752

chrNC_037338.1:13047319-13056419

3.14E-06

0.000158716

LOC101905635

chrNC_037340.1:76723459-76739979

0.000910843

0.016809347

LOC112449550

chrNC_037341.1:76478493-76487345

5.40E-06

0.000252129

LOC104974324

chrNC_037342.1:75777823-75780010

0.000204918

0.005162159

LOC104974692

chrNC_037344.1:67130432-67132302

2.33E-05

0.000858982

LOC112442200

chrNC_037345.1:45609576-45616675

0.001584248

0.025977108

LOC112442383

chrNC_037345.1:62159119-62160399

0.000214833

0.005332407

LOC112443129

chrNC_037348.1:25091115-25128295

0.003878832

0.049737964

LOC101903141

chrNC_037353.1:47669900-47687837

7.72E-13

1.95E-10

LOC112444607

chrNC_037354.1:36287211-36298269

9.39E-07

5.76E-05

LOC112446394

chrNC_037331.1:68871351-68873779

1.62E-08

1.64E-06

LOC101907194

chrNC_037332.1:26047494-26049444

3.59E-12

8.06E-10

Shandong Black cattle

LOC112447868

chrNC_037335.1:410273-419534

4.43E-06

0.000212223

LOC112449245

chrNC_037340.1:3165193-3220562

7.90E-15

2.68E-12

LOC112441643

chrNC_037342.1:54862373-54875026

4.23E-05

0.001397951

LOC112441659

chrNC_037342.1:78782537-78785977

0.000658605

0.01318643

LOC104975164

chrNC_037346.1:61277209-61278623

6.05E-05

0.001918059

LOC112442997

chrNC_037347.1:23104372-23114203

0.000324072

0.007462557

LOC100847412

chrNC_037348.1:32211463-32219891

1.13E-06

6.79E-05

LOC100847415

chrNC_037357.1:18645452-18649324

2.19E-06

0.000118565

We constructed a Circos diagram to visually display the distribution of differential LncRNAs and differential mRNAs (Fig. 4a). The results showed that most differentially expressed LncRNAs mapped to chromosome 9, with others mapping to chromosomes 3, 1, 11, and 7. Pearson correlation coefficient analysis of differentially expressed LncRNA and differential mRNA genes (Fig. 4b) showed 43844 pairs of co-expressed LncRNAs and differential mRNA genes with correlation coefficient >0.8 (Table S7). Based on genomic location, 387 differentially expressed LncRNAs were selected for further study.

  1. Prediction of differentially expressed LncRNAs target genes

LncRNAs can regulate the expression of adjacent mRNAs. We analyzed the LncRNAs and protein-coding genes by analyzing mRNAs within 50 kb of LncRNAs. The results showed that 387 of the differentially expressed LncRNAs targeted 1164 target genes. In these predictions, one LncRNA targeted multiple mRNA and many LncRNAs targeted the same mRNA (Table S8). Of these, LOC112448071, LOC112444635, LOC112445963, LOC104975064, LOC101903261, LOC535280, and LOC521224 can target genes related to muscle development, including MYORG, Wnt4, PAK1, and ADCY7.

The potential target genes of LncRNA were next subjected to GO and KEGG analysis (Fig. 5). We found 130 significantly enriched GO items (P<0.05), with 20 related to the regulation of muscle development (Table 5). The 40 most enriched terms include multicellular organization development, single multicellular organization process, and multicellular organic process (Table S9). The GO terms related to muscle development are listed in Table 5. KEGG analysis showed enrichment of 1164 potential target genes in 299 pathways, with the 40 most significantly enriched pathways listed in Table S9. Some of these are related to muscle development (Table 6), including the calcium signaling pathway, the AMPK signaling pathway, the cGMP-PKG signaling pathway, and the PPAR signaling pathway.

  1. LncRNA-miRNA-mRNA network interaction analysis

We used Targetscan, Miranda, and PITA qtar software13 to predict the target gene relationships between mRNA and miRNA, and used Miranda and PITA qtar to predict the target gene relationships between miRNA and LncRNA. The top 5% most related pairs were visualized using the average degree method and the Fruchterman- Reingold layout method in Gephi(Fig. 6a). LncRNA can function as ceRNA to regulate the expression of downstream genes at the post-transcriptional level by binding miRNAs. We selected several miRNAs related to muscle development (including miR-1, miR-133, miR-206, miR-181, miR-21, and mir-23a) to predict and screen downstream-regulated mRNA using Targetscan and Miranda software. We then constructed an LncRNA-miRNA-mRNA interaction network diagram using Cytoscape version 3.5.1 (Fig. 6b). LncRNAs that may be involved in the development of beef cattle muscle are summarized in Table 7.

  1. Luciferase reporter assay

We next looked for potential miRNA binding sites of LOC104975788 and found potential miR-133a and miR-103 binding sites. Online miRNA binding site prediction software (RNA22 and RNAhybird) both predicted potential interaction of miR-133a with LOC104975788 and LOC536229(Pax7). The results showed that both LOC104975788 and LOC536229 (Pax7) contained miR-133a binding sites (Fig. 7a). Luciferase reporter assay showed significantly lower luciferase activity of miR-133a co transfected with the psi-LOC112448162 vector compared to the control group (P<0.01). The luciferase activity of miR-133a co-transfected with psi-LOC536229 (Pax7) vector was also significantly decreased relative to the control group (P<0.01)(Fig. 7b and 7C). These results indicate that both LOC104975788 and LOC536229 (Pax7) contain miR-133a binding sites.

  1. Verification of sequencing results

Nine differentially expressed LncRNAs and five differentially expressed mRNAs were verified by qRT-PCR. The qRT-PCR results (Fig. 7d and 7e) confirmed differential expression of these LncRNAs and mRNAs.

Discussion

Muscle fiber is the basic unit of muscle, and the type of muscle fiber greatly shapes muscle characteristics14. There were obvious differences in appearance in the muscle fibres isolated from longissimus dorsi muscles of Shandong black cattle and Luxi cattle. The average muscle fiber area of Shandong black cattle was significantly larger than that of Luxi cattle (P < 0.05), and the muscle fiber diameter was smaller than that of Luxi cattle. There was a significant positive correlation between muscle fiber area and carcass traits. Muscle fiber diameter is closely related to meat quality and taste and thicker muscle fiber diameter corresponds to decreased muscle tenderness. There is a negative correlation between muscle fiber diameter and muscle fiber density15. The amount of slow muscle fiber affects sarcomere length and has an important impact on meat quality16. There was a higher proportion of slow muscle fibers in longissimus dorsi muscle samples from Shandong black cattle than that of Luxi cattle. There were also significant differences in the muscle fiber density, the ratio of fast-twitch fibers/slow-twitch fibers, and weight (P < 0.05) in this study. These differences may be key factors leading to the differences in meat production performance and meat quality of the two breeds of cattle after birth, which was also the research basis of this study to explore the underlying molecular regulatory mechanism.

With low conservation of LncRNA sequences among species, bioinformatics methods are required to screen and identify LncRNAs. This type of analysis is based on transcript length, the number of exons, and the coding potential. In this study, transcriptome data of skeletal muscle of Shandong black cattle and Luxi cattled were generated and analyzed. According to the screening criteria of |FC|≥2 and P < 0.05, 1415 transcripts were found to be differentially expressed, with 939 and 476 transcripts were significantly up-regulated and down regulated in Luxi cattle, respectively. Further, 19 GO items (Table 2) and 14 regulatory pathways (Table 3) related to muscle development were screened by hierarchical GO and KEGG cluster analysis. Many genes involved in the regulation of muscle development were identified, including myosin protein family genes (MYH1, MYH3, MYH4, MYH8, and MYL3), myogenic regulatory factor family genes (MYO10 and MYO5B), Homeobox family genes (HOXC10, HOXC11, and HOXD9), Troponin T family genes (TNNT2 and TNNT3) and some regulatory transcription factors (WNT4 and TNNT2). It is worth noting that three LncRNAs (LOC107133268, LOC112445823, LOC104969184) are directly enriched into muscle regulation items. Further target gene prediction analysis shows that LncRNAs may participate in the regulation of muscle development through bta-miR-2892 (LOC107133268), bta-miR-2360 and bta-miR-2449 (LOC112445823). The specific regulatory mechanism needs to be further verified and analyzed.

Compared with studies in human17 and other model organisms18, there has been limited identification and characterization of beef LncRNAs, especially ones related to skeletal muscle development, with most studies of cattly focused on the identification of genes and miRNAs 19,20. In this study, we identified 8427 multiple exon LncRNAs in beef skeletal muscle, and 480 differentially expressed LncRNAs were identified. More LncRNAs were detected in this study than previously reported in goats21,22. Fifteen randomly selected differentially expressed transcripts were verified by qPCR, and the results were consistent with the results of RNA sequencing. In conclusion, these results confirm the reliability of the identification of LncRNAs23.

Although LncRNAs can act on gene sites far from their chromosomal location24, genes in close proximity on a chromosome may participate in the same cellular metabolic pathways and have similar biological functions25. Therefore, the distribution of differentially expressed LncRNAs on chromosomes and the linkage differential expression of nearby genes may have biological significance that can help us to determine the function of a gene. The most differentially expressed LncRNAs were found on chromosome 9, followed by chromosomes 3, 1, 11, and 7. In co-expression analysis, we detected 387 differentially expressed LncRNA transcripts related to protein-coding genes according to the expression correlation coefficient values (r > 0.9), and predicted 1164 target genes. GO and KEGG analysis showed significant enrichment of 130 GO items (P < 0.05), with 20 of them related to the regulation of genes involved in muscle development. KEGG analysis showed enrichment of 1164 potential target genes in 299 pathways, with some related to muscle development (Table 6), such as the calcium signaling pathway, the AMPK signaling pathway, the cGMP - PKG signaling pathway, and the PPAR signaling pathway. Interestingly, we also found MYORG, Dll1, EFNB2, SOX6, PROX1, MYOCD, NEBL and MYLK3 genes, annotated as related to muscle development (Table 5). Overall, LncRNAs may play a regulatory role in skeletal muscle biological development through cis or trans mechanisms.

At the post transcriptional level, LncRNA binds miRNA competitively with mRNA for protein-coding genes, thus relieving the inhibitory effect of miRNA on protein-coding genes to promote expression of these genes26. We analyzed the expression patterns of LncRNAs differentially expressed among different breeds, and constructed an LncRNA-miRNA-mRNA interaction network related to skeletal muscle development. A total of 52 LncRNAs related to muscle development were identified, with nine up-regulated and five down-regulated. All of these LncRNAs contain one or more putative miRNA binding sites with 48 LncRNAs predicted to interact with 1–2 miRNAs related to muscle development, and other LncRNAs predicted to interact with more than three miRNAs, such as LOC525506, LOC540051, LOC514189. With the miRNA seed sequences, LncRNAs can bind to miRNA to act as a sponge, preventing miRNA from binding to its target mRNA. As a classic example, M. Cesana27 confirmed that linc-MD1, a long non-coding RNA specifically expressed during myoblast differentiation, regulates the expression of muscle specific transcription regulators MAML1 and MEF2C through miR-133 and miR-135. In particular, LOC525506 may be involved in the development of beef skeletal muscle by regulating miR-1, miR-23a, miR-378, let-7, miR-483, and miR-21. This is the most predicted LncRNA, but little is known about its expression and function. Both LOC112447073 and LOC104975788 are involved in the skeletal muscle development of beef cattle by interaction with miR-103. More interestingly, some target genes of LOC112447073, LOC104975788, and LOC101903367 directly regulate the development of muscle fiber and maintain the stability and development of muscle, such as miRna-103, miR-133a, miR-145, MEF2C, myocardin, and Pax728,29. Choi identified 11 LncRNAs in bovine transcripts by studying skeletal muscle and adipose tissue of Korean cattle, with four related to muscle function30. These data provide new insight into the role of LncRNA in muscle development and enrich the existing LncRNA mammalian skeletal muscle resources.

The second to eighth bases of the sequence at the 5 'end of miRNA is called the seed sequence. Complete complementarity of this sequence and that of the target gene indicates the potential for binding of the target gene by miRNA31,32. We predicted the miRNA binding sites of functional LncRNAs that may be related to skeletal muscle development. Using a double luciferase test, we found a recognition site of bta-mir-133a in the sequence of LOC104975788 and a binding site for bta-mir-133a in the downstream target gene Pax7. We speculate that LOC104975788 may be involved in the regulation of skeletal muscle development by competitively inhibiting the expression of the target gene Pax7 through binding of bta-miR-133a. Future work should test this proposed regulatory mechanism.

Conclusion

The expression patterns of LncRNAs in skeletal muscle of Shandong black cattle and Luxi cattle were elucidated by RNA sequence analysis, and the LncRNAs that may be involved in the skeletal muscle development of beef cattle were identified. The results allowed construction of interaction networks of LncRNAs-miRNA-mRNA regulated by muscle biology, which can provide insight into the regulatory mechanisms of LncRNAs involved in skeletal muscle development. Interactions among mRNA, miRNA, and LncRNA may regulate muscle development. This work provides a framework to explore the molecular mechanisms used by LncRNA to regulate important economic traits of muscle development, and should help to accelerate the livestock breeding process and improve animal production performance.

Abbreviations

DEGs:differentially expressed genes; KEGG:Kyoto Encyclopedia of Genes and Genomes

GO:The Gene Ontology; LncRNA:Long noncoding RNA; miRNA: MicroRNA (miRNA) is a class of noncoding single stranded RNA molecules with a length of about 22 nucleotides encoded by endogenous genes; mRNA: Messenger RNA(mRNA) that encodes proteins.

Declarations

Ethics approval and consent to participate

All experimental design and procedures of this study was performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of Qingdao Agricultural University IACUC (Institutional Animal Care and Use Committee) .

Consent for publication

Not applicable.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Data availability

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request. We have stored the RNA sequence data in the public domain GEO NCBI to obtain the GEO accession numbers: GSM4904154, GSM4904155, GSM4904156, GSM4904157, GSM4904158, GSM4904159.

Competing interests

The authors declare that they have no competing interests.

Funding

This work was supported by Shandong modern agricultural industry technology system cattle industry innovation team (668-2216030). The team participated in the collection and analysis of data.

Authors’ contributions

YJD, XJB, CZX and RLL designed the study. RLL,XXL and KY conducted the experiment. RLL, XXL and KY performed and collected the data. RLL analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgements

The authors would like to thank Shanghai Sangon Biotechnology Co., Ltd. and Beijing AnnoroadGene Co., Ltd. for their assistance in the original data processing and related bioinformatics analysis for RNA- sequencing and GC-MS. The authors also thank Dr. Liu for her meticulous revision of language. More importance, special thanks to Yue Liu for his contribution to data processing. Additionally, the authors would like to particularly thank her parents and Dr. Dong , for their continuing love and support.

Author details

1Laboratory of Animal Physiology and Biochemistry, Animal embryo Center, College of Animal Science, Qingdao Agricultural University, Qingdao, Shandong, People’s Republic of China200109. 2Laboratory of Animal molecular , Shandong black cattle breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao, Shandong, People’s Republic of China200109.

References

[1] Li Y , Chen X , Sun H , et al. Long non-coding RNAs in the regulation of skeletal myogenesis and muscle diseases. Cancer Letters. 417(2017)58-64.

[2] Sun, X, Li,et al. The developmental transcriptome sequencing of bovine skeletal muscle reveals a long noncoding RNA, lncMD, promotes muscle differentiation by sponging miR-125b. Biochimica Et Biophysica Acta. (2016).

[3] Yingwei, Yue, Congfei, et al. A LncRNA promotes myoblast proliferation by up-regulating GH1.. In vitro cellular & developmental biology. Animal. (2017).

[4] Billerey C , Boussaha M , Diane Esquerré, et al. Identification of large intergenic non-coding RNAs in bovine muscle using next-generation transcriptomic sequencing. BMC Genomics. 15(2014)499.

[5] Lu W Q. Feeding and management techniques of Luxi cattle in fattening period. Beijing Agriculture. 25(2015)130-131 .

[6] Liu R L , et al. Screening of Skeletal Muscle Differential Genes Based on Transcriptome. North China Agricultural Journal. 33 (2018)64-72.

[7] Cao, T, Shi, L.G, Zhang, L.L,et al. Comparative Study on Fetal Muscle Fiber of Wuzhishan Pig and Changbai Pig during 65d Gestation. Journal of Animal Ecology. 35(2014)37-40.

[8] Zhao J, Ohsumi TK, Kung JT, Ogawa Y, Grau DJ, Sarma K, Song JJ, Kingston RE,et al. Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell. 40(2010)939-53.

[9] Wang L , Feng Z , Wang X , et al. DEGseq: an R package for identifying differentially expressed genes from RNA SEQ data . Bioinformatics. 26 (2010)136-138.

[10] Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol. 11(2010)R106.

[11] Cabili M N , Trapnell C , Goff L , et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes & Development. 25( 2011)1915-1927.

[12] Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol. 11(2010)R14.

[13] Li D, Bao P, Yin Z, Sun L, Feng J, He Z, Jin M, et al. Exploration of the involvement of LncRNA in HIV-associated encephalitis using bioinformatics. PeerJ. 6(2018)e5721. 

[14] Steinbacher P, Haslett J R, Sanger A M, et al. Evolution of myogenesis in fish: a sturgeon view of the mechanisms of muscle development . Anat Embryol (Berl). 211(2006)311-322.

[15] Zhou Zhijia. Relationship between muscle histological characteristics and meat quality . Breeding technology consultant. 8( 2010) 57-58.

[16] Hendricks H B. Relation of porcine muscle fiber type and size to postmortem shortening. Anim Sci. 32(1971)57.

[17] Agliano F, Rathinam V.A, Medvedev A.E, Vanaja S.K, Vella A.T. Long Noncoding RNAs in Host-Pathogen Interactions. Trends Immunol. (2019).

[18] Briggs J.A, Wolvetang E.J, Mattick J.S, Rinn J.L, Barry G. Mechanisms of Long Non-coding RNAs in Mammalian Nervous System Development, Plasticity, Disease, and Evolution. Neuron. 88(2015)861-877.

[19] Peters E.L, Van der Linde S.M, Vogel I.S.P, Haroon M, Offringa C, De Wit G.M.J, Koolwijk P, Van der Laarse W.J, Jaspers R.T. IGF−1 Attenuates Hypoxia-Induced Atrophy but Inhibits Myoglobin Expression in C2C12 Skeletal Muscle Myotubes. Int. J. Mol. Sci. 18(2017)1889.

[20] Xue Z, Huang K, Cai C, Cai L, Jiang C.Y, Feng Y, Liu Z, Zeng Q, Cheng L, Sun Y.E, et al. Genetic programs in human and mouse early embryos revealed by single-cell RNA sequencing. Nature. 500(2013)593-597.

[21] Sun M, Gadad S.S, Kim D.S, Kraus W.L. Discovery, Annotation, and Functional Analysis of Long Noncoding RNAs Controlling Cell-Cycle Gene Expression and Proliferation in Breast Cancer Cells. Mol. Cell. 59(2015)698-711.

[22] Sun L, Luo H, Bu D, Zhao G, Yu K, Zhang C, Liu Y, Chen R, Zhao Y. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 41(2013)e166.

[23] Langfelder P, Zhang B, Horvath S. Defining clusters from a hierarchical cluster tree: The Dynamic Tree Cut package for R. Bioinform. 24(2008)719-720.

[24] Cohen B A , Mitra R D , Hughes J D , et al. A computational analysis of whole-genome expression data reveals chromosomal domains of gene expression. Nature Genetics.26(2000), 183-186.

[25] Salmena L, Poliseno L, Tay Y, et al. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language?. Cell. 146(2011) 353-358.

[26] Kartha R V, Subramanian S. Competing endogenous RNAs (ceRNAs): new entrants to the intricacies of gene regulation. Frontiers in genetics. 5 (2014) 8.

[27] Cesana M, Cacchiarelli D, Legnini I, et al. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147(2011) 358-369.

[28] Mathew S.J., Hansen J.M., Merrell A.J., Murphy M.M., Lawson J.A., Hutcheson D.A., Hansen M.S., Angus-Hill M., Kardon G. Connective tissue fibroblasts and Tcf4 regulate myogenesis. Dev. (Camb. Engl.) .138(2011)371-384.

[29] Bennett R.D., Caride A.J., Mauer A.S. Interaction with the IQ3 motif of myosin-10 is required for calmodulin-like protein-dependent filopodial extension. Febs Lett. 582( 2008)2377-2381.

[30] Choi J Y , Shin D , Lee H J , et al. Comparison of long noncoding RNA between muscles and adipose tissues in Hanwoo beef cattle. Animal cells and systems the official publication of the Zoological Society of Korea. 23(2018)1-9.

[31] Lewis B P, Burge C B, Bartel D P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are micro RNA targets. Cell. 120(2005)15-20.

[32] Grimson A, Farh K K, Johnston W K, et al. Micro RNA targeting specificity in mammals: determinants beyond seed pairing. Molecular Cell. 27(2007) 91-105.