- Research
- Open access
- Published:
Visfatin (NAMPT) affects global gene expression in porcine anterior pituitary cells during the mid-luteal phase of the oestrous cycle
Journal of Animal Science and Biotechnology volume 15, Article number: 96 (2024)
Abstract
Background
The pituitary belongs to the most important endocrine glands involved in regulating reproductive functions. The proper functioning of this gland ensures the undisturbed course of the oestrous cycle and affects the female’s reproductive potential. It is believed that visfatin, a hormone belonging to the adipokine family, may regulate reproductive functions in response to the female’s metabolic state. Herein we verified the hypothesis that suggests a modulatory effect of visfatin on the anterior pituitary transcriptome during the mid-luteal phase of the oestrous cycle.
Results
RNA-seq analysis of the porcine anterior pituitary cells revealed changes in the expression of 202 genes (95 up-regulated and 107 down-regulated in the presence of visfatin, when compared to the non-treated controls), assigned to 318 gene ontology terms. We revealed changes in the frequency of alternative splicing events (235 cases), as well as long noncoding RNA expression (79 cases) in the presence of the adipokine. The identified genes were associated, among others, with reproductive system development, epithelial cell proliferation, positive regulation of cell development, gland morphogenesis and cell chemotaxis.
Conclusions
The obtained results indicate a modulatory influence of visfatin on the regulation of the porcine transcriptome and, in consequence, pituitary physiology during the mid-luteal phase of the oestrous cycle.
Introduction
The pituitary is one of the most important endocrine organs in the organism. This small, inconspicuous structure, through the secretion of several tropic hormones, mediates the functioning of several target organs such as adrenals and gonads. Due to its structure, consisting of the nervous and glandular parts, the pituitary processes the central nervous system derived signals into the hormonal commands forwarded to the endocrine tissues. Due to its location, and regulatory abilities, the pituitary is a crucial point of the regulatory loops, such as the hypothalamic-pituitary-adrenal (HPA) or hypothalamic-pituitary-gonadal (HPG) axes. The secretory function of the anterior pituitary (AP) may be regulated not only by the hypothalamic-derived releasing hormones, like growth hormone releasing hormone or gonadoliberin, or the hormones sent as the feedback response from target tissues such as luteinizing hormone (LH), follicle-stimulating hormone (FSH) or prolactin (PRL), but also by several other factors and hormones, like steroid hormones, prostaglandins, cytokines, neuropeptides, or activin, inhibin and follistatin [1,2,3,4,5,6,7]. A growing body of evidence indicates that the pituitary actions may also be modulated by hormones involved in metabolism regulation such as adipokines.
Adipokines, a group of hormones produced mainly by the white adipose tissue, exert a pleiotropic action in the organism, however, their primary function is the integration of energy metabolism. Previous studies indicated the regulatory effect of adipokines, i.a., leptin, adiponectin, chemerin or apelin, on the secretory functions of the pituitary [8,9,10,11,12,13,14,15,16]. Visfatin (Nicotinamide phosphoribosyltransferase, NAMPT, VIS), also included in the adipokines group, was described for the first time in 1994 as a pre-B cell colony enhancing factor (PBEF) [17]. The hormone was qualified as an adipokine when its insulin-mimetic effects were proven for the first time [18]. The human NAMPT gene is located at 7q22.3 and encodes 52–54 kDa protein product [17, 19]. NAMPT protein may exist in the organism in two distinct isoforms: intra- (iNAMPT) and extracellular (eNAMPT) [20], from which iNAMPT is considered an intracellular enzyme and the adipokines’ specific actions are addressed only to the extracellular form. Until now no specific VIS receptor has been identified.
Besides its insulin-mimetic actions, VIS was found to exert proinflammatory influence through the up-regulation of cytokines production in monocytes [21]. It has also been proved, that adipokine acts as an anti-apoptotic and pro-angiogenic agent [22, 23]. The hormone has also been reported as a stimulatory factor in several tissues tumorigenesis (for a review see: [24]). Several metabolic disorders, such as obesity, polycystic ovary syndrome, type 2 diabetes or atherosclerosis have been correlated with VIS plasma level deviations [25,26,27,28]. NAMPT localisation in the endocrine glands, like ovaries, testes, mammary gland, pituitary, or in the hypothalamus, the structure responsible for the regulation of the above glands, indicates its potential involvement in the endocrine axes regulation [29,30,31,32,33,34].
The results of our previous studies indicating the modulatory influence of VIS on LH and FSH secretion by the porcine anterior pituitary cells (APc) [35] inspired us to set up a hypothesis assuming the regulatory role of adipokine in this important gland. Herein, we verified the hypothesis, assuming the modulatory effect of VIS on the global gene expression in the porcine APc. The studies were conducted using Illumina Next Generation Sequencing (NGS) technology. The obtained RNAseq results were used for the analysis of differentially expressed genes (DEGs), as well as for the determination of changes in the expression of long noncoding RNAs (DELs) and frequency of alternative splicing events (DASs). The revealed DEGs were assigned to specific Gene Ontology (GO) categories and analysed for the potential interaction networks. Since the secretory functions of the pituitary are subjected to the cyclic changes related to the phase of the oestrous cycle of the animals, the research was conducted with the use of animals from d 10 to 12 of the oestrous cycle, when the secretory activity of the corpus luteum (CL), and hence, whole HPG axis is high, but does not change as dynamically as in the case of the follicular phase. Moreover, on those days, the secretion of luteinizing (LH), as well as follicle stimulating (FSH) hormones does not change as dynamically as during the pre- and postovulatory phases of the cycle which could affect the obtained results [36, 37]. Another justification for choosing this particular period is a very similar activity of CL during the mid-luteal (d 10–12) phase of the cycle and during gestation, the obtained herein results may, in the future, serve for the investigation of the VIS effect on the HPG axis during pregnancy.
Materials and methods
Animals and tissue collection
All studies were conducted in following the Polish Act on the protection of animals used for scientific or educational purposes, enacted on January 15, 2015 (Polish Journal of Law of 2015 item 266) as well as directive 2010/63/EU of the European Parliament of September 22, 2010, on the protection of animals used for a scientific purpose. For the experiment, the glandular parts of the pituitaries were obtained from the crossbreed gilts (age of 7–8 months and weight of 130–150 kg) on d 10 to 12 of the oestrous cycle and descended for slaughter at the commercial abattoir. The day of the oestrous cycle was confirmed based on the morphology of the ovaries (large, centrally closed, pink corpora lutea, soft in touch, with the inner cavity filled with luteal tissue and blood vessels at the surface; presence of medium, 4.0–6.0 mm of diameter, follicles) [38]. Within a few minutes after slaughter, APs were placed in ice-cold phosphate buffer solution (PBS) enriched with glucose and a mix of antibiotics (Antibiotic Antimycotic Solution, Merck, USA), and immediately transported to the laboratory. The number of APc isolated from one pituitary gland was approximately 9 to 10 million, whereas about 18 million APc were necessary to conduct one experiment (n = 1). Therefore, 2 to 3 APs were collected and digested together. The in vitro cell cultures were conducted in five independent replicates (n = 5).
In vitro cell cultures and total RNA isolation
The APc were isolated as described by Kawamata et al. [39], with modifications. The cells were washed with Dulbecco’s medium (Sigma-Aldrich, USA) enriched with 0.1% bovine serum albumin (Sigma-Aldrich, USA) and antibiotics (Antibiotic Antimycotic Solution, Merck, USA), and homogenized with a scalpel blade. The homogenates were digested with 0.2% collagenase solution (Sigma-Aldrich, USA) at 37 °C for 30 min. Isolated APc were centrifuged three times at 1,000 × g (10 min, room temperature) in 15-mL tubes containing Dulbecco’s medium. The remained tissue was treated with 0.2% collagenase and 0.25% pancreatin (Sigma-Aldrich, USA) for 10 min. The digestion steps were repeated until the complete digestion of the material. After each digestion, the obtained cells were collected and washed in Dulbecco’s medium, as described above. The cell suspensions were pooled and filtered through a 70-μm mesh filter. The total amount and viability of APc were estimated using a haemocytometer and trypan blue dye (Sigma-Aldrich, USA), the mean percentage of viable cells after isolation was 95.65% ± 1.3%. The cells were suspended in McCoy’s 5 A medium (Sigma-Aldrich, USA), enriched with 10% horse serum (Sigma-Aldrich, USA) and 2.5% foetal calf serum (Sigma-Aldrich, USA) and seeded onto 6-well culture plates (1.5 × 106 cells/2 mL medium/well). The cells were preincubated for 72 h at 37 °C in a water-saturated atmosphere with 5% CO2 and 95% air. After 48 h of preincubation, 1 mL of fresh, serum-free McCoy’s 5 A medium was added to each well. After preincubation media were replaced with fresh, enriched McCoy’s 5 A medium and incubated for another 24 h in the presence of human recombinant VIS (final concentration of 100 ng/mL; Cat. # 8424-VF, RD System, USA). The hormone concentration was determined based on Reverchon et al. [32] and our preliminary studies. Since VIS was diluted directly in McCoy’s medium, cells descended for the control group (n = 5) were incubated in the pure medium without the presence of any treatment. The potential effect of VIS on cell viability was excluded with the use of the Alamar Blue test. Cell viability and toxicity of the treatment were determined using the alamarBlue™ assay according to the manufacturer’s instructions. The mean percentage of viable cells after incubation was 103.6% ± 6.2%.
After the incubation, total RNA from APc was isolated using Extrazol (BLIRT, Poland), following the manufacturer’s protocol. The purity (A260/A280) and quantity (wavelength 260 nm, A260) of the obtained RNA samples were determined spectrophotometrically using Infinite M200 Pro (Tecan, Männedorf, Switzerland). Bioanalyzer 2100 (Agilent Technology, USA) was used to determine the RNA integrity. Only samples which passed RNA integrity number (RIN) > 8 criteria were qualified for the further transcriptome high-throughput sequencing (RNA-Seq) analysis and the quantitative real-time polymerase chain reaction (qPCR) and reverse transcription polymerase chain reaction (RT-PCR) validations.
Library construction and sequencing
The NGS sequencing procedure, as well as preliminary bioinformatic analysis, was conducted by LCSciences (USA). Approximately 5 µg of total RNA was used to remove ribosomal RNA (rRNA) from the samples with the use of Ribo-Zero Gold rRNA Removal kit (Illumina, USA). The remaining RNA samples were fragmented into short fragments using divalent cation buffers at elevated temperatures (NEBNext® Magnesium RNA Fragmentation Module, NEB, USA). The obtained samples were reverse-transcribed to cDNA using SuperScript™II Reverse Transcriptase (Invitrogen, USA). Subsequently, cDNA was used to synthesise U-labeled second-stranded DNA using E. coli DNA polymerase I (NEB, USA), RNase H (NEB, USA) and dUTPSolution (Thermo Fisher, USA). The obtained double-stranded cDNA (dscDNA) was subjected to the end repair and A-tailing processes. In the end, the obtained fragments of dscDNA were ligated with specific adaptors. Quality control analysis and the quantification of the obtained libraries were performed with the use of Agilent Technologies 2100 Bioanalyzer High Sensitivity DNA Chip. Illumina’s NovaSeq 6000 sequencing system (Illumina, San Diego, CA, USA) was used to conduct the 2 × 150 paired-ended sequencing analysis with a minimum sequencing depth of 40 million reads per sample.
Bioinformatic analysis
To investigate the regulatory mechanisms affected by VIS in the porcine APc, the expression of genes, as well as long noncoding RNAs (lncRNAs) and alternative splicing (AS) events were analysed. The in silico analyses were performed as described in our previous study [40], with modifications. Visualization of the obtained data was conducted using SRplot online tools (http://www.bioinformatics.com.cn/).
Transcripts assembly and differentially expressed transcripts processing
To remove the reads containing adapters, and low-quality scores (QPhred score ≤ 20; reads containing more than 5% of unknown nucleotides), Cutadapt (v.1.9) [41] and custom Python scripts were applied. The quality of the obtained raw reads was determined using FastQC (v.0.11.9) [42]. HISAT2 (v.2.0.4) [43] script was applied to map the filtered reads to Sus scrofa domestica genome (v.107; [44]) deposited in Ensembl database [45]. The mapped reads were assembled using StringTie (v.1.3.4d) [46]. The transcriptomes from all samples were merged to reconstruct a comprehensive transcriptome using custom Python scripts and gffcompare (v.0.9.8) [47]. Subsequently, StringTie and ballgown [48] were used to estimate the expression levels of all transcripts.
To determine the expression levels for mRNAs and lncRNAs by calculating FPKM (fragments per kilobase of transcript per million fragments mapped), the StringTie script was applied. The differences in expression of mRNAs and lncRNAs between control and VIS-treated groups were determined with DESeq2 [49] software (and edgeR between 2 samples; [50]). Due to the peri-physiological concentration of the used treatment, the expected changes in the genes and lncRNA expression were rather discrete. Therefore, the cutoff level for DEGs or differentially expressed lncRNAs (DELs) was defined at the level of 0.56 (log2FC ≥ 0.56). All results with P < 0.05 were considered statistically significant.
Functional annotation of target genes
The gene set enrichment analysis was performed using GSEA software v.4.1.0 [51] and Molecular Signatures Database (MSigDB). Enrichment ontology and pathway analysis were performed based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) [52] and GO [53, 54] databases. The KEGG and GO terms with P < 0.05 were defined as significant.
Interaction network of DEGs
The interaction network between chosen DEGs was generated using the GeneMania Prediction Server [55]. From the list of DEGs with log2FC > 0.56, 15 were chosen. The selected gene products were found to be involved in several processes such as the regulation of pituitary functioning, apoptosis or angiogenesis. The interaction networks were generated based on the known interplay between genes: co-expression, genetic interactions, co-localization (cis-interactions), physical interactions, shared protein domains and signalling pathways.
Identification of lncRNAs, target gene prediction and functional analysis
To identify lncRNAs candidates whose expression differed between the porcine APc treated and non-treated with VIS, the transcripts overlapping with known mRNAs, known lncRNAs and transcripts shorter than 200 bp were excluded. The remained sequences were analyzed with the use of a Coding Potential Calculator (CPC) (v.0.9-r2) [56] and a Coding-Non-Coding Index (CNCI) (v.2.0) [57] tools to predict transcripts with coding potential. Transcripts with a CPC score < 0.5 and a CNCI score < 0 were qualified and considered as novel lncRNAs. The remaining transcripts with class codes (i, j, o, u, x) were considered as known lncRNAs. Subsequently, to identify potential cis interactions of lncRNAs with the neighbouring genes, a custom Python script was applied to select 100,000 upstream and downstream coding genes. To identify differences in lncRNA sequences’ expression between the probes from control and VIS-treated groups, DESeq2 software was applied. The functional analysis of the target genes for lncRNAs was conducted and the results with a false discovery rate (FDR; q) < 0.05 were considered statistically significant. The obtained DELs were descended into one of five categories, according to class code generated by StringTie software [46]: (i) a transfrag falling entirely within a reference intron (intronic); (j) potentially novel isoform or fragment at least one splice junction is shared with a reference transcript; (o) generic exonic overlap with a reference transcript; (u) unknown, intergenic transcript (intergenic); and (x) exonic overlap with reference on the opposite strand (antisense). The relatively close localization of DELs and DEGs on the same chromosome (up to 100,000 bp) were qualified into the category cis-interactions.
Alternative splicing analysis
To identify AS events, as well as to analyze differential alternative splicing events (DASs) between control and VIS-treated groups, a replicate multivariate analysis of transcript splicing (rMATS) (v.4.1.1) script was applied [51]. Trimmed reads with 120 bp length were applied to estimate the percent of splicing inclusion (PSI) in a range of splicing sites at intron/exon junctions. AS events which passed the criteria of q < 0.05 and the absolute value of inclusion level difference, |ΔPSI| > 0.1 were considered statistically significant. The revealed DASs were classified into one of five alternative splicing types, as follows: alternative 3′ splice site (A3), 5′ splice site (A5SS), mutually exclusive exons (MXE), retention intron (RI), and skipping exon (SE).
Validation of DEGs and DELs by quantitative real-time PCR (qPCR) method
To validate the results obtained from NGS, qPCR analysis was conducted. The analysis was conducted using Aria Mx Real-time PCR System (Agilent Technology, USA) as described by Smolinska et al. [58], with the use of the same RNA as in the RNAseq method (n = 5/group). Specific primer pairs used to amplify parts of 5 chosen DEGs: CD180 molecule (CD180), inhibin subunit beta E (INHBE), regucalcin (RGN), midkine (MDK) and angiogenin (ANG), as well as for 2 chosen DELs: ENSSSCT00000101418 and ENSSSCT00000069084. The following reference genes were used: ubiquitin C (UBC), and 18S ribosomal RNA (18sRNA). Primer sequences, as well as qPCR reaction conditions, were detailed in Table 1. The constitutively expressed genes UBC and 18sRNA were used as the internal controls to verify the method. The reaction mixture contained the following components: cDNA, primers, Sensitive RT HS-PCR Mix SYBR (12.5 µL), and ROX (0.24 µL; reference dye), along with RNase-free water to a final volume of 20 µL. The negative non-template controls (NTCs) contained water instead of cDNA, or the reverse transcription step was omitted. All reactions were run in duplicates. To confirm the specificity of qPCR reactions, at the end of the experiment, the analysis of the melting curve was carried out. The purity of the products was confirmed with the use of agarose gel electrophoresis. The calculation of validated DEGs/DELs relative expression was conducted with the use of the comparative cycle threshold method (ΔΔCT) and normalized using the geometrical means of the reference genes’ Ct values [59]. The normality of qPCR data distributions was confirmed by the Shapiro–Wilk test (P > 0.05). The results of the experiments were analyzed with the use of a Student t-test using Statistica software (Statsoft Inc., USA). Values for P < 0.05 were considered statistically significant.
Polymerase chain reaction (RT-PCR; DASs validation)
The characteristics of SE events in 5 chosen genes: G protein-coupled receptor 173 (GPR173), MSL complex subunit 1 (MSL1), CASP8 and FADD-like apoptosis regulator (CFLAR), mitogen-activated protein kinase 6 (MAPK6), and phosphatidylinositol 4-kinase alpha (PI4KA), were conducted with the use of Labcycler 48s (Syngen Biotech, Poland) and StartWarm HS-PCR Mix (A&A Biotechnology, Poland). The reaction mixture (in a final volume of 25 µL) contained 12.5 µL of Hot Start PCR Mix, forward and reverse primers, nuclease-free deionized water, and cDNA. NTCs were prepared as described in the previous section. PCR conditions and primers’ concentrations were determined experimentally in a temperature gradient and, together with the primer sequences, are detailed in Table 1. The obtained PCR products were analysed on 1.5% agarose gels containing Midori Green Advance dye (Nippon Genetics Europe, Germany).
Results
Quality control and statistics of reads
A total of 479,237,334 raw paired-end reads contained an average of 47,923,733 reads per sample revealed. The total alignment to the Sus scrofa gave 48%–77% of coverage. The average number of 47.92 million valid reads, and 33.24 million mapped reads passed the Illumina producers’ criteria allowing for enriching the present study with DASs analysis. On average, 68.68% of reads were mapped uniquely, whereas 1.09% were found to be mapped in multiple loci. About 61.54% of the obtained reads were mapped to coding DNA sequence regions (CDS), whereas the rest belonged to introns (34.8%) or intergenic regions (3.66%). The percentage of uniquely mapped reads results from the adopted method of constructing sequencing libraries (ribo-zero), and the high percentage of nucleotides mapped to CDS regions confirm a high proportion of coding transcripts in the obtained reads. The relatively high percentage of nucleotides mapped to introns indicated the chance of detecting large changes in intron retention type of alternative splicing. Taking into consideration the applied library constructing method, a large part of lncRNA coding sequences was automatically assigned to the intron group at the first step of the analysis. The lncRNA sequences were separated at the further steps of analyses. The correctness of the results for the obtained readings was confirmed in the validation process described below. All the statistics for the individual samples have been detailed in Supplementary File 1. Raw reads were deposited in the Functional Genomics Data Collection (ArrayExpress) database under the common project accession number E-MTAB-13,922.
DEGs and functional annotations (GO; KEGG)
Out of a total of 18,132 identified genes, 202 met the criteria of log2FC >|0.56| and P < 0.05, and have been presented in Fig. 1 and 2. Among all 202 obtained DEGs, 95 were upregulated and 107 were downregulated under the influence of VIS (Supplementary File 2). To discover possible functions of DEGs identified in the APc treated with VIS, the genes were classified into one of the following GO categories: ‘biological processes’ (BP), ‘cellular components’ (CC) and ‘molecular function’ (MF). DEGs were assigned to 318 GO terms (P < 0.05). Among them, 208 terms were assigned to BP, 44 to CC and 66 to MF category (Supplementary File 3). In the BP category, the most of DEGs were connected with positive regulation of transcription by RNA polymerase II (GO:0045944; 29 DEGs), negative regulation of transcription by RNA polymerase II (GO:0000122; 26 DEGs) and translation (GO:0006412; 22DEGs). In the CC category, the most enriched terms were nucleus (GO:0005634; 116 DEGs), ribosome (GO:0005840; 23 DEGs) and chromatin (GO:0000785; 15 DEGS). In the MF category, the biggest number of DEGs was related to DNA binding (GO:0003677; 38 DEGs), RNA polymerase II cis-regulatory region sequence-specific DNA binding (GO:0000978; 26 DEGs), structural constituent of ribosome (GO:0003735; 21 DEGs) and sequence-specific DNA binding (GO:0043565; 20 DEGs). The results of GO analysis have been presented in Fig. 3. Figure 4 presents the enrichment of GO terms with chosen, important DEGs.
The hierarchical clustering heatmap of chosen genes whose expression was altered under the influence of visfatin. Each column represents biological replicates of control (CTR1-5) or visfatin (VIS1-5). Different colours of brackets represent the normalized (Z-score; red-green scale) expression values for DEGs in each biological replicate
Visualization of the number of genes whose expression changed in the presence of visfatin. The X-axis represents logarithmic fold changes in expression (log2FC), whereas the Y-axis shows the negative decimal logarithm of the P-values. The horizontal line refers to the negative logarithmic P-value cut-off ( P = 1.3). The vertical lines mark the fold change cut-offs (log2FC > |0.56|)
Results of Gene Ontology (GO) analysis of genes whose expression differed under the influence of visfatin (P < 0.05, log2FC > |0.56|). A Bar plot of GO components enriched by DEGs evaluated in the anterior pituitary cells treated with visfatin. Orange bars represent GO terms connected with biological processes (BP). Green bars represent GO terms related to the cellular component (CC) category. Dark blue bars represent GO terms connected with the molecular function (MF) category. The vertical axis contains the names of GO terms. The horizontal axis describes the enrichment score based on the number of significant DEGs. Net plots present the GO enrichment of processes connected with BP (B), CC (C) and MF (D)categories. Different colour lines mean different GO terms. Different size of the dots reflects the number of significant DEGs assigned to GO terms. The colour of the dots reflects changes in the expression of the involved DEGs (log2FC)
The visualization of the statistically significant, differentially expressed genes (DEGs) and their enrichment in ontology terms. Circos plot presents selected Gene Ontology terms associated with porcine anterior pituitary cells’ DEGs with expression altered under the influence of visfatin ( P < 0.05, log2FC>|0.56|). Different colours of lines mean different GO terms, whereas the colour of bars assigned to the genes’ symbols represents the ratio between up- and downregulated genes. Red bars represent GO terms with the predominance of upregulated DEGs, and blue bars show terms with a predominance of downregulated DEGs
The analysis of KEGG database revealed 16 signalling pathways (P < 0.05), from which the most enriched were those connected with Coronavirus disease-COVID-19 (ko05171;19 DEGs), Ribosome (ko03010; 18 DEGs), Amyotrophic lateral sclerosis (ko05014; 15 DEGs) and Pathways of neurodegeneration - multiple diseases (ko05022; 15 DEGs). The results of the KEGG databases analysis have been presented in Fig. 5. The detailed results of the analysis have been attached in Supplementary File 3.
Scatterplot diagram presenting the enrichment of KEGG pathways with DEGs under the influence of visfatin (P < 0.05, log2FC > |0.56|). The vertical axis describes the revealed pathways. The horizontal axis describes the enrichment score calculated based on the negative decimal logarithm of the pathway’s P-value. The size of the dots reflects the number of DEGs involved in the pathway
Interaction networks between DEGs
The interaction network was created between 34 chosen DEGs, involved in, i.a., positive regulation of cell-cell adhesion, tissue remodelling, cytokine activity, interleukin-6 production, leukocyte migration, angiogenesis and embryonic hemopoiesis. GeneMania was used to predict the relations between the chosen genes (query genes) in seven different types of interaction as follows: physical interactions (Fig. 6A), co-expression and genetic interactions (Fig. 6B), as well as co-localization, pathways, predicted and shared protein domains (Fig. 6C). The analysis considered an additional 20 automatically generated genes, which were necessary to indicate the observed networks (interacting genes). Moreover, using different colours, we indicated the contribution of genes in specific biological functions: reproductive system development (red), epithelial cell proliferation (blue), positive regulation of cell development (orange), gland morphogenesis (purple), cell chemotaxis (green) and regulation of phospholipase C activity (light purple; refer to the key in Fig. 6). Co-expression of the genes was found in 99 interactions and co-localization was observed in 5 interactions. In 15 cases, the genes participated in common pathways. Genetic interaction was revealed in 28 cases. Physical interactions were present in 59 cases, and in 23 cases interactions were based on the shared protein domains. In 18 cases, the interaction between genes was assigned to the ‘predicted’ category. The complete list of gene interactions is presented in Supplementary File 6.
Identification of lncRNAs, target gene prediction and functional analysis
Out of the total number of 11,204 identified lncRNA candidates, 79 passed the criteria of FDR < 0.05 and log2FC > 0.56 and were qualified as DELs. Out of them, 44 were annotated with ENSEMBL gene IDs, and the other 34 were predicted during the procedure for identifying novel lncRNAs. In the obtained group of DELs, 47 were upregulated, whereas 32 were downregulated, in the VIS-treated group (Table 2). The analysis revealed 83 interactions. Among them, 63 results indicated the colocalization in a range of 100,000 bp, 15 were in close proximity (10,000 bp) and 3 were in very close proximity (1,000 bp). Only four of the revealed lncRNAs showed a high correlation with the target gene (Pearson Correlation Coefficient > 0.8), however, their target genes did not pass the criteria for DEGs. Because we did not identify any significant DEL–DEG pairs, data on the protein-coding genes and DELs co-localization were not included in further functional analyses and the following GO analysis was conducted for genes connected with DELs, whose did not fulfil the DEGs definition.
The GO analysis of the obtained results revealed 64 processes in which genes connected with the identified lncRNAs were involved. Among them, 45 were related to the BP cluster, 7 to CC, and 11 to MF. One of the revealed processes, transport across blood-brain barrier (GO:0150104; 2 DELs), has not been categorized. In the BP category the most enriched processes were the defense response to virus (GO:0051607; 13 DELs), defense response (GO:0006952; 11 DELs), B cell differentiation (GO:0030183; 10 DELs) and adaptive immune response (GO:0002250; 10 DELs). The most enriched processes in the CC category were cell cortex (GO:0005938; 10 DELs), stress fiber (GO:0001725; 6 DELs) and endocytic vesicle (GO:0030139; 6 DELs). The most enriched positions in the MF function were RNA polymerase II transcription regulatory region sequence-specific DNA binding (GO:0000977; 20 DELs), cytokine receptor binding (GO:0005126; 9 DELs) and type I interferon receptor binding (GO:0005132; 8 DELs). The GO analysis results of DELs were detailed in Supplementary File 4.
Alternative splicing analysis results
The analysis of AS occurrence revealed 21,736 events, from which we identified 235 as belonging to the DASs group (|ΔPSI| > 0.1; Fig. 7). Among all the revealed events, we identified 11 A3SS, 4 A5SS, 27 MXE, 8 RI, and 77 SE events. Figure 8 presents the Sashimi plots of the chosen DASs (GPR173, MSL1, CFLAR, MAPK6 and PI4KA). The detailed results of the DASs analysis are provided in Supplementary File 5.
Visualisation of results obtained during alternative splicing (AS) events analysis of AP cells transcriptome under the influence of visfatin. A The Volcano plot describes the differences in AS events frequency between the control and visfatin-treated groups. The X-axis describes ΔPSI-values for each AS event and the Y-axis shows the negative decimal logarithm of the adjusted P-value. The horizontal line refers to the negative logarithmic adjusted P-value cut-off (q = 1.3). The vertical lines refer to the ΔPSI-value cut-off (ΔPSI > |0.1|). B Bar graph presenting the occurrence of differentially expressed AS events (DAS). The vertical axis presents the number of DASs, and the horizontal axis presents different types of alternative splicing events. Different colours of dots (A) and bars (B) refer to the different types of significant AS. Events: blue — alternative 3′ splicing sites (A3SS), yellow — alternative 5′ splicing sites (A5SS), red — mutually exclusive exons (MXE), dark green — retention intron (RI), and purple — skipping exon (SE)
Quantitative visualisation (Sashimi plot) of differential alternative splicing events (DASs) statistically significant in the changes of percentage splicing inclusion (q < 0.05 and |ΔPSI| > 0.1) between visfatin-treated group (VIS1; purple) and control group (CTRL; red) samples. The numbers in each upper right corner of the tracks present the percentage of splicing inclusion (PSI) values. Splicing differentiations are covered by the number of reads mapped in the range of junction sites. The upper black tracks show the genomic localisation of splicing events. The plot was generated in ggsashimi Python script. Below of plots, we present validation results of chosen DASs using the PCR method. The images show the inclusion and skipping exon levels between VIS and the control group. Abbreviations: GPR173 - G protein-coupled receptor 173, MSL1 - MSL complex subunit 1, CFLAR - CASP8 and FADD like apoptosis regulator, MAPK6 - mitogen-activated protein kinase 6, PI4KA - phosphatidylinositol 4-kinase alpha
Quantitative real-time PCR and PCR analyses results
To validate the obtained RNA-seq results, five DEGs (CD180, INHBE, RGN, MDK and ANG), as well as 2 DELs (ENSSSCT00000101418 and ENSSSCT00000069084) were selected for the qPCR experiment. For the validation of DASs analysis, five AS genes were chosen (GPR173, MSL1, CFLAR, MAPK6 and PI4KA). QPCR analyses of DEGs and DELs (Fig. 9A and B) and PCR analysis of DASs (Fig. 8) confirmed the data obtained during the NGS experiment. Validation results confirmed the veracity and accuracy of the RNA-Seq, as well as the data analysis methods used in the present study.
Quantitative real-time PCR validation of RNA-seq results for differentially expressed genes (A) and long noncoding RNAs (B) in the visfatin-treated porcine anterior pituitary cells. Panel (C) presents the correlation between the obtained qPCR and RNAseq results. Validation was performed for CD180, INHBE, RGN, MDK and ANG genes, as well as for ENSSSCT00000069084 and ENSSSCT00000101418 lncRNAs with the use of UBC (ubiquitin C) and 18sRNA (18S ribosomal RNA) reference genes (* P-value < 0.05; ** P-value < 0.01). Abbreviations: CTRL - control group, VIS - visfatin-treated group, CD180 - CD180 molecule, INHBE - inhibin subunit beta E, RGN - regucalcin, MDK – midkine, ANG - angiogenin
Discussion
To our knowledge, this is the first study describing the influence of VIS on the global gene expression in mammalian cells. Until today, the only study describing the influence of the hormone on the transcriptome was conducted with the in vivo use of VIS in chicks [60]. Herein, we revealed the significant effect of the hormone on the expression of several genes and lncRNAs, as well as on the occurrence of DASs in the porcine APc. The created interaction analysis presents a complex network of genes interplaying with each other on different levels, from which the most common was co-expression and physical interactions. Despite the low number of direct interactions between them, their interdependence through the intermediate genes despite completely different functions indicates the complex and pleiotropic influence of VIS in the porcine pituitary.
Visfatin, as one of the adipokines responsible for the regulation of metabolism, is also perceived as a potential factor integrating energy balance and endocrine system regulation in response to the hormonal status of the organism. The expression of the VIS gene and protein has been reported in different endocrine organs such as sheep, mice, porcine and human pituitaries [34, 61, 62], human, bovine, porcine and murine ovaries [32, 62,63,64], rat, pig, mice, chicken and human testes [33, 65,66,67,68]. Besides the direct expression of VIS in the endocrine glands, the hormone expression was also confirmed in mice and porcine hypothalami, the overriding structure responsible for the regulation of the endocrine functions [29, 67]. Therefore, it seems that VIS may play a significant role in the HPG axis’ functioning. Evidence for this may be provided by the research results of Maillard et al. [62], who indicated the inhibitory influence of adipokine on the secretion of LH by the mice LβT2 cells. The hormone was also found to stimulate the secretion of adrenocorticotrophic hormone (ACTH) by rat corticotroph cells [69]. It was also proven that VIS is involved in the regulation of ovarian and testicular steroidogenesis [32, 70, 71].
During the present study, we indicated the influence of VIS on the expression of genes connected with the regulation of pituitary secretory properties. The adipokine stimulated the expression of progesterone receptor (PGR) and early growth response 2 (EGR2) genes and inhibited the expression of inhibin subunit beta E (INHBE), PROP Paired-Like Homeobox 1 (PROP1), and dopamine receptor 2 (DRD2) genes. Progesterone receptors (PRs) mediate the actions of the progestin hormones influencing reproductive functions. PRs belong to the group of intracellular factors acting as ligand-modulated transcription agents [72]. The enhanced expression of the member of the ‘steroid hormone mediated signalling pathway’ (GO: GO:0043401), PGR, in the APc under the influence of VIS indicates its potential involvement in the regulation of the gland sensitivity to P4 and, indirectly, on the relationship between the pituitary and ovaries, and ovarian processes, such as steroidogenesis, as indicated by Reverchon et al. [32]. Another member of the mentioned term, retinoid X receptor gamma (RXRG) has also a proven role in the regulation of progesterone, as well as oestradiol-mediated transcription processes [73]. Besides its regulatory influence on the APc physiology through PGR, VIS affected also several other genes connected with the regulation of females’ reproductive functions. We revealed the inhibition of INHBE gene expression under the influence of VIS. Inhibins and activins belong to the group of peptide hormones of the transforming growth factor-β superfamily and play a pivotal role in the regulation of the reproductive system functioning [74], controlling, i.a., FSH release from the pituitary [75]. The direct perfusion of inhibin into the pituitary causes a drastic decrease in follicle-stimulating hormone subunit beta (FSHβ) gene expression and, for more, decreases the half-life of FSHβ subunit mRNA [76, 77].
These indicates that VIS may be involved in the regulation of gonadotrophins production during the oestrous cycle which seems to be confirmed by the revealed change in the occurrence of skipping exon events in the G protein-coupled receptor 173 (GPR173) gene (ΔPSI = 0.103) encoding the phoenixin receptor. Phoenixin is a neuropeptide with a proven regulatory role in gonadotrophins’ secretion through the modulation of gonadoliberin (GnRH) receptor expression [78]. It was hypothesized that phoenixin may regulate the preovulatory LH surge required for ovarian cyclicity [78, 79]. Since Stein et al. [79] reported that silencing of the GPR173 gene abrogates the phoenixin-induced LH secretion, we may conclude that through the modulation of GPR173 splicing frequency and INHBE expression, VIS affects the production of gonadotrophins, and hence, affects the ovarian functions. The confirmation of the above may be supported by the results of our studies indicating the modulatory effect of VIS on basal and GnRH-stimulated secretion of LH and FSH by the porcine APc [35].
Besides the modulatory influence of VIS on the gonadotroph cells in the AP, its influence on the expression of genes mentioned above indicates its regulatory effect on the secretion of other pituitary-derived hormones such as PRL, growth hormone (GH) and thyroid stimulating hormone (TSH). We revealed the inhibitory effect of adipokine on the expression of the PROP1 gene, which product is crucial for pituitary development and physiology. The pituitaries of PROP1 null mice were found to be hypoplastic and the gonadotroph cell differentiation was delayed [80]. Patients with the defect in the PROP1 gene are characterized by the impaired production of GH, TSH and PRL, as well as LH and FSH [81]. On the other hand, the overexpression of the PROP1 gene was observed in pituitary adenomas which indicates its crucial role in pituitary development [82]. The product of another VIS-affected gene encodes EGR2, also connected with GH production which inhibition results in the somatotroph cell population ablation [83]. The next revealed gene, DRD2, encodes dopamine receptor D2. At the level of the pituitary, dopamine inhibits PRL secretion via the DRD2 receptor [84,85,86]. VIS influence on the lactotroph cells’ proliferation and secretory functions through modulation of DRD2 expression may be additionally supported by its effect on the modulation of DASs in the latent transforming growth factor beta binding protein 4 (LTBP4) gene (RI; ΔPSI = 0.163). LTBP4 is responsible for enhancing the secretion of the TGF-β latent form, regulating the availability and activation of the growth factor. TGF-β was found to cooperate with dopamine in the regulation of lactotroph functions [87, 88]. The above-presented findings indicating the modulatory influence of VIS on the expression of genes connected with the pituitary development and production of hormones reveal the existence of complex mechanisms of the pituitary trophic cell regulation, in which VIS may play a crucial role.
Besides its primary function as the metabolic regulator, VIS plays an important role in several processes such as apoptosis, cell proliferation, or angiogenesis. VIS, through the mitogen-activated protein kinase/phosphoinositide 3-kinase (MAPK/PI3K) pathway, may inhibit the apoptosis in the pancreatic MIN6 β-cell line [89]. Xiang et al. [90] reported that the suppression of apoptosis in rat pancreatic β-cell lines is mediated through AMPK and ERK1/2 signalling pathways. In contrast, Li et al. [91] identified that in macrophages the anti-apoptotic influence of VIS is achieved through the interleukin-6/STAT3 signalling mechanism. The above suggests that VIS controls cells’ apoptosis through many different transduction pathways and its activity is complex and multi-pronged. The presented data analysis revealed changes in the expression and occurrence of AS events in several genes connected with apoptosis. VIS inhibited the expression of apoptosis-associated tyrosine kinase (AATK) and KLF transcription factor 2 (KLF2) and enhanced the expression of regucalcin (RGN), caspase activity and apoptosis inhibitor 1 (CAAP1), and signal transducing adaptor family member 1 (STAP1) genes. AATK acts as the inductor of the apoptosis process [92,93,94,95]. The overexpression of AATK in melanoma cells caused their growth arrest and induced apoptosis, whereas its epigenetic silencing was observed in human and murine cancer cell lines [96,97,98]. The product of another gene, KLF2, a member of Kruppel-like transcriptional factors, is known for its growth inhibitory and apoptosis induction properties and its expression suppression has been reported in many types of cancer [99,100,101]. Contrary to the previous genes, the products RGN, CAAP1, and STAP1, have been found to exert an anti-apoptotic effect and the disruption in their expression was reported in many cancers [102,103,104,105,106,107]. The mentioned earlier suggestion of VIS involvement in the regulation of the apoptosis process may also be confirmed by its influence on lncRNA expression and AS events in apoptosis-related genes. The analyses revealed a significant influence of VIS on lncRNA addressed to GO terms connected to apoptosis-related signalling pathways, ‘positive regulation of intrinsic apoptotic signaling pathway by p53 class mediator’ (GO:1,902,255) and ‘positive regulation of NIK/NF-kappaB signaling’ (GO:1,901,224). We indicated the modulatory effect of the adipokine on the expression of ENSSSCT00000101418 lncRNA transcript interacting with the cytoskeleton-associated protein 5 (CKAP5) gene which is engaged in apoptosis. Cells of different tumour cell lines were reported to be arrested in metaphase and underwent cell death after silencing CKAP5 [108]. We identified AS modifications of mRNA encoding MSL1 (SE; ΔPSI= −0.314), CFLAR (SE; ΔPSI=−0.208), solute carrier family 25 member 23 (SLC25A23; RI; ΔPSI= −0.243), and ILK associated serine/threonine phosphatase (ILKAP; RI; ΔPSI = 0.127) genes, whose products are connected with the process of apoptosis [109,110,111,112,113]. The above confirms that VIS is an important factor in the regulation of APc survival through its role in the control of apoptosis.
Another important feature of VIS is the hormone’s influence on the process of angiogenesis. Through the stimulation of vascular endothelial growth factors (VEGF) and matrix metalloproteinases (MMP) production, the adipokine enhances endothelial angiogenesis. Additionally to the previous, VIS was also reported to inhibit the production of MMPs inhibitors, TIMP Metallopeptidase Inhibitors 1 and 2 (TIMP-1 and TIMP-2) [114]. Another study confirms the pro-angiogenic actions of VIS which takes place through the stimulation of interleukin-6 expression in endothelial cells [115]. Herein we revealed the modulatory effect of VIS on the gene connected with ‘positive regulation of vasculogenesis’ (GO:2001214), CD34, as well as on the expression of several genes connected with angiogenesis, like the endosialin gene (CD248), KLF2, MDK, and ANG. VIS caused an increase in the expression of the CD248 gene which was reported as a potentially important marker of embryonic and tumour vascularization. Since CD248 has not been reported in endothelial cells, it is considered a marker of neighbouring pericytes and stromal cells influencing the process of new vessel formation [116,117,118,119]. Contrary to CD248, KLF2 product has been found to inhibit endothelial cell activation and proliferation, as well as VEGF-A-mediated angiogenesis [120]. The VIS-mediated stimulation of the CD248 gene and suppression of the KLF2 gene suggest its pro-angiogenic features. However, VIS also suppressed the expression of other pro-angiogenic factors, ANG, MDK, and CD34. Angiogenin is a crucial factor for the induction of angiogenesis through the activation of cell migration, invasion, proliferation, and formation of tubular structures [121]. In tumorigenesis, ANG, secreted by developing tumour cells, ensures their nutrition, through the stimulation of vessels, as well as the promotion of cells’ proliferation [122,123,124]. Midkine, besides its role in the promotion of angiogenesis, takes also part in the regulation of cell migration and neurogenesis. The downregulation of MDK results in suppression of angiogenesis, independently of VEGF [125,126,127]. CD34 is considered a marker of the vascularization process [128]. The ambiguous influence of VIS on the expression of genes connected with angiogenesis suggests that adipokine may play a role in maintaining homeostasis of the angiogenesis process through the regulation of new vessel formation. That thesis may be also supported by the results concerning the alteration in lncRNAs’ expression and AS occurrence. We discovered that the presence of VIS altered the frequency of AS events in the integrin subunit alpha V (ITGAV) gene (A3ss; ΔPSI= −0.128). The product of ITGAV, integrin αV, which together with integrin β3 forms a vitronectin receptor, has been found to play a key role in the new vessel formation. The suppression of the vitronectin receptor by its antagonists resulted in the inhibition of the vessels’ formation in many experimental models [129,130,131,132,133]. What is more, we identified also VIS-induced changes in the expression of lncRNAs connected with cytoplasmic polyadenylation element binding protein 1 (CPEB1) and AKT serine/threonine kinase 3 (AKT3) genes (ENSSSCT00000069084 and MSTRG.1914, respectively). Both genes were found to be involved in angiogenesis [134,135,136,137]. The alteration in the expression of their regulatory lncRNAs taken together with the proven influence of VIS on the expression of DEGs and AS events frequency confirms the adipokine’s involvement in the regulation of this process in the glandular part of the porcine pituitary.
Conclusions
To our knowledge, this is the first study concerning the influence of VIS on the global gene expression in the mammalian APc. The revealed influence of the adipokine on the expression of genes, such as those important for the secretory functions of the gland as PGR, INHBE, and DRD2, taken together with its influence on genes involved in the apoptosis (RGN) and angiogenesis (ANG), presents VIS as an important agent in the regulation of pituitary physiology. Taking into consideration that all three mentioned processes are crucial in the process of tumorigenesis, the results presented herein may help to understand this process and the potential role of VIS in pituitary tumour formation, however, further, in-depth studies are necessary.
Availability of data and materials
The datasets supporting the conclusions of this article are available in the Functional Genomics Data Collection (ArrayExpress) repository and can be accessed with the E-MTAB-13922 accession number.
Abbreviations
- 18sRNA:
-
18S ribosomal RNA
- A3SS:
-
Alternative 3′ splice site (alternative splicing type)
- A5SS:
-
5′ splice site (alternative splicing type)
- AATK:
-
Apoptosis-associated tyrosine kinase
- ACTH:
-
Adrenocorticotropic hormone
- ANG:
-
Angiogenin
- AP:
-
Anterior pituitary
- APc:
-
Anterior pituitary cells
- AS:
-
Alternative splicing
- BP:
-
Biological processes (GO category)
- CAAP1:
-
Caspase activity and apoptosis inhibitor 1
- CC:
-
Cellular components (GO category)
- CD180:
-
CD180 molecule
- CD248:
-
Endosialin
- CFLAR:
-
CASP8 and FADD-like apoptosis regulator
- CKAP5:
-
Cytoskeleton associated protein 5
- CPEB1:
-
Cytoplasmic polyadenylation element binding protein 1
- DASs:
-
Differential alternative splicing events
- DEGs:
-
Differentially expressed genes
- DELs:
-
Differentially expressed lncRNAs
- DRD2:
-
Dopamine receptor 2
- EGR2:
-
Early growth response 2
- FC:
-
Fold change
- FDR:
-
False discovery rate
- FSH:
-
Follicle-stimulating hormone
- GH:
-
Growth hormone
- GnRH:
-
Gonadotrophin-releasing hormone
- GO:
-
Gene Ontology
- GPR173:
-
G protein-coupled receptor 173
- HPG:
-
Hypothalamic-pituitary-gonadal axis
- INHBE:
-
Inhibin subunit beta E
- KEGG:
-
Kyoto Encyclopedia of Genes and Genomes
- KLF2:
-
KLF transcription factor 2
- LH:
-
Luteinizing hormone
- lncRNAs:
-
Long noncoding RNAs
- MAPK6:
-
Mitogen-activated protein kinase 6
- MDK:
-
Midkine
- MF:
-
Molecular function (GO category)
- MMP:
-
Matrix metalloproteinases
- MSL1:
-
MSL complex subunit 1
- MXE:
-
Mutually exclusive exons (alternative splicing type)
- NAMPT:
-
Nicotinamide phosphoribosyltransferase
- NGS:
-
Next generation sequencing
- P4 :
-
Progesterone
- PGR:
-
Progesterone receptor
- PI4KA:
-
Phosphatidylinositol 4-kinase alpha
- PRL:
-
Prolactin
- PROP1:
-
PROP Paired-Like Homeobox 1
- PRs:
-
Progesterone receptors
- qPCR:
-
Quantitative real-time polymerase chain reaction
- RGN:
-
Regucalcin
- RI:
-
Retention intron (alternative splicing type)
- RT-PCR:
-
Reverse transcription polymerase chain reaction
- SE:
-
Skipping exon (alternative splicing type)
- TSH:
-
Thyroid stimulating hormone
- UBC:
-
Ubiquitin C
- VEGF:
-
Vascular endothelial growth factors
- VIS:
-
Visfatin
References
Bilezikjian LM, Blount AL, Leal AMO, Donaldson CJ, Fischer WH, Vale WW. Autocrine/paracrine regulation of pituitary function by activin, inhibin and follistatin. Mol Cell Endocrinol. 2004;225:29–36.
Aguilera G, Aguilera G. Regulation of the hypothalamic-pituitary-adrenal axis by neuropeptides. Horm Mol Biol Clin Investig. 2011;7:327–36.
Jones TH, Kennedy RL. Cytokines and hypothalamic-pituitary function. Cytokine. 1993;5:531–8.
Hedge GA. Roles for the prostaglandins in the regulation of anterior pituitary secretion. Life Sci. 1977;20:17–33.
Stefaneanu L. Pituitary sex steroid receptors: localization and function. Endocr Pathol. 1997;8:91–108.
Donda A, Reymond F, Rey F, Lemarchand-Beraud T. Sex steroids modulate the pituitary parameters involved in the regulation of TSH secretion in the rat. Acta Endocrinol (Copenh). 1990;122:577–84.
Chowen JA, Frago LM, Argente J. The regulation of GH secretion by sex steroids. Eur J Endocrinol. 2004;151 Suppl 3:U95.
Sharma R, Biedenharn KR, Fedor JM, Agarwal A. Lifestyle factors and reproductive health: taking control of your fertility. Reprod Biol Endocrinol. 2013;11:66. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1477-7827-11-66.
Goazigo ARL, Alvear-Perez R, Zizzari P, Epelbaum J, Bluet-Pajot MT, Llorens-Cortes C. Cellular localization of apelin and its receptor in the anterior pituitary: evidence for a direct stimulatory action of apelin on ACTH release. Am J Physiol Endocrinol Metab. 2007;292(1):E7–15.
Kisielewska K, Rytelewska E, Gudelska M, Kiezun M, Dobrzyn K, Bogus-Nowakowska K, et al. Expression of chemerin receptors CMKLR1, GPR1 and CCRL2 in the porcine pituitary during the oestrous cycle and early pregnancy and the effect of chemerin on MAPK/Erk1/2, Akt and AMPK signalling pathways. Theriogenology. 2020;157:181–98.
Dobrzyn K, Smolinska N, Kiezun M, Szeszko K, Rytelewska E, Kisielewska K, et al. Adiponectin: a new regulator of female reproductive system. Int J Endocrinol. 2018;29:7965071.
Rak A, Mellouk N, Froment P, Dupont J. Adiponectin and resistin: potential metabolic signals affecting hypothalamo-pituitary gonadal axis in females and males of different species. Reproduction. 2017;153:R215–26.
Kiezun M, Smolinska N, Maleszka A, Dobrzyn K, Szeszko K, Kaminski T. Adiponectin expression in the porcine pituitary during the estrous cycle and its effect on LH and FSH secretion. Am J Physiol Endocrinol Metab. 2014;307:E1038–46.
Yu WH, Kimura M, Walczewska A, Karanth S, Mccann SM. Role of leptin in hypothalamic-pituitary function. Proc Natl Acad Sci U S A. 1997;94:1023–8.
Popovic V, Damjanovic S, Dieguez C, Casanueva FF. Leptin and the pituitary. Pituitary. 2001;4:7–14.
Lloyd RV, Jin L, Tsumanuma I, Vidal S, Kovacs K, Horvath E, et al. Leptin and leptin receptor in anterior pituitary function. Pituitary. 2001;4:33–47.
Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colony-enhancing factor. Mol Cell Biol. 1994;14:1431.
Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M, Kishimoto K, et al. Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science. 2005;307:426–30.
Wu X, Cooper RS, Borecki I, Hanis C, Bray M, Lewis CE, et al. A combined analysis of genomewide linkage scans for body mass index from the National Heart, Lung, and Blood Institute Family Blood Pressure Program. Am J Hum Genet. 2002;70:1247–56.
Shackelford RE, Mayhall K, Maxwell NM, Kandil E, Coppola D. Nicotinamide phosphoribosyltransferase in malignancy: a review. Genes Cancer. 2013;4:447–56.
Moschen AR, Kaser A, Enrich B, Mosheimer B, Theurl M, Niederegger H, et al. Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol. 2007;178:1748–58.
Kim SR, Bae SK, Choi KS, Park SY, Jun HO, Lee JY, et al. Visfatin promotes angiogenesis by activation of extracellular signal-regulated kinase 1/2. Biochem Biophys Res Commun. 2007;357:150–6.
Jia SH, Li Y, Parodo J, Kapus A, Fan L, Holstein OD, et al. Pre–B cell colony–enhancing factor inhibits neutrophil apoptosis in experimental inflammation and clinical sepsis. J Clin Invest. 2004;113:1318–27.
Grolla AA, Travelli C, Genazzani AA, Sethi JK. Extracellular nicotinamide phosphoribosyltransferase, a new cancer metabokine. Br J Pharmacol. 2016;173:2182–94.
Stastny J, Bienertova-Vasku J, Vasku A. Visfatin and its role in obesity development. Diabetes Metab Syndr. 2012;6:120–4.
Chan TF, Chen YL, Chen HH, Lee CH, Jong SB, Tsai EM. Increased plasma visfatin concentrations in women with polycystic ovary syndrome. Fertil Steril. 2007;88:401–5.
Kang YS, Song HK, Lee MH, Ko GJ, Cha DR. Plasma concentration of visfatin is a new surrogate marker of systemic inflammation in type 2 diabetic patients. Diabetes Res Clin Pract. 2010;89:141–9.
Liu SW, Qiao S, Bin, Yuan JS, Liu DQ. Association of plasma visfatin levels with inflammation, atherosclerosis and acute coronary syndromes (ACS) in humans. Clin Endocrinol (Oxf). 2009;71:202–7.
Kaminski T, Kiezun M, Zaobidna E, Dobrzyn K, Wasilewska B, Mlyczynska E, et al. Plasma level and expression of visfatin in the porcine hypothalamus during the estrous cycle and early pregnancy. Sci Rep. 2023;11:8698.
Reverchon M, Cornuau M, Cloix L, Ramé C, Guerif F, Royère D, et al. Visfatin is expressed in human granulosa cells: regulation by metformin through AMPK/SIRT1 pathways and its role in steroidogenesis. Mol Hum Reprod. 2013;19:313–26.
Annie L, Gurusubramanian G, Roy VK. Changes in the localization of ovarian visfatin protein and its possible role during estrous cycle of mice. Acta Histochem. 2020;122:151630.
Reverchon M, Rame C, Bunel A, Chen W, Froment P, Dupont J. Visfatin (NAMPT) improves in vitro IGF1-induced steroidogenesis and IGF1 receptor signaling through SIRT1 in bovine granulosa cells. Biol Reprod. 2016;94:54–5.
Yonezawa T, Haga S, Kobayashi Y, Takahashi T, Obara Y. Visfatin is present in bovine mammary epithelial cells, lactating mammary gland and milk, and its expression is regulated by cAMP pathway. FEBS Lett. 2006;580:6635–43.
Szymanska K, Zaobidna E, Rytelewska E, Mlyczynska E, Kurowska P, Dobrzyn K, et al. Visfatin in the porcine pituitary gland: expression and regulation of secretion during the oestrous cycle and early pregnancy. Sci Rep. 2023;13:18253.
Szymanska K, Rytelewska E, Zaobidna E, Kiezun M, Gudelska M, Kopij G, et al. The effect of visfatin on the functioning of the porcine pituitary gland: an in vitro study. Cells. 2023;12:2835.
Van de Wiel DF, Erkens J, Koops W, Vos E, Van Landeghem AA. Periestrous and midluteal time courses of circulating LH, FSH, prolactin, estradiol-17 β and progesterone in the domestic pig. Biol Reprod. 1981;24:223–33.
Ziecik A, Krzymowska H, Tilton JE. Porcine LH levels during the estrous cycle, gestation, parturition and early lactation. J Anim Sci. 1982;54:1221–6.
Akins EL, Morrissette MC. Gross ovarian changes during estrous cycle of swine. Am J Vet Res. 1968;29:1953–7.
Kawamata Y, Fujii R, Fukusumi S, Habata Y, Hosoya M, Hinuma S, et al. Analyses for susceptibility of rat anterior pituitary cells to prolactin-releasing peptide. Endocrine. 2000;12:215–21.
Orzechowska K, Kopij G, Paukszto L, Dobrzyn K, Kiezun M, Jastrzebski J, et al. Chemerin effect on transcriptome of the porcine endometrium during implantation determined by RNA-sequencing. Biol Reprod. 2022;107:557–73.
Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 2011;17:10–2.
Andrews S. FastQC: a quality control tool for high throughput sequence data. 2010. Available online at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc , Accessed 19 Feb 2021 .
Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357–60.
Ensembl Gene Annotation (107) Sus scrofa. 2022. Available online at: https://www.ensembl.org/info/genome/genebuild/2022_05_Sus_scrofa_gene_annotation.pdf. Accessed 9 Feb 2024.
Yates AD, Achuthan P, Akanni W, Allen J, Allen J, Alvarez-Jarreta J, et al. Ensembl 2020. Nucleic Acids Res. 2020;48:D682.
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;33:290–5.
Pertea G, Pertea M. GFF utilities: GffRead and GffCompare. F1000Res. 2020;28:9.
Fu J, Frazee AC, Collado-Torres L, Jaffe AE, Leek JT. Ballgown: Flexible, isoform-level differential expression analysis. 2022. https://www.bioconductor.org/packages/release/bioc/html/ballgown.html . Accessed 24 Mar 2023 .
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.
Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–61.
Carbon S, Dietze H, Lewis SE, Mungall CJ, Munoz-Torres MC, Basu S, et al. Expansion of the gene ontology knowledgebase and resources. Nucleic Acids Res. 2017;45:D331.
Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene Ontology: tool for the unification of biology. Nat Genet. 2000;25:25–9.
Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010;38:W214–20.
Kang YJ, Yang DC, Kong L, Hou M, Meng YQ, Wei L, et al. CPC2: a fast and accurate coding potential calculator based on sequence intrinsic features. Nucleic Acids Res. 2017;45:W12–6.
Sun L, Luo H, Bu D, Zhao G, Yu K, Zhang C, et al. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 2013;41:e166-166.
Smolinska N, Kiezun M, Dobrzyn K, Rytelewska E, Kisielewska K, Gudelska M, et al. Expression of chemerin and its receptors in the porcine hypothalamus and plasma chemerin levels during the oestrous cycle and early pregnancy. Int J Mol Sci. 2019;20:3887.
Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2-∆∆CT method. Methods. 2001;25:402–8.
Li Z, Liu X, Zhang P, Han R, Sun G, Jiang R, et al. Comparative transcriptome analysis of hypothalamus-regulated feed intake induced by exogenous visfatin in chicks. BMC Genomics. 2018;19:249.
Dupré SM, Burt DW, Talbot R, Downing A, Mouzaki D, Waddington D, et al. Identification of melatonin-regulated genes in the ovine pituitary pars tuberalis, a target site for seasonal hormone control. Endocrinology. 2008;149:5527–39.
Maillard V, Elis S, Desmarchais A, Hivelin C, Lardic L, Lomet D, et al. Visfatin and resistin in gonadotroph cells: expression, regulation of LH secretion and signalling pathways. Reprod Fertil Dev. 2017;29:2479–95.
Shen CJ, Tsai EM, Lee JN, Chen YL, Lee CH, Chan TF. The concentrations of visfatin in the follicular fluids of women undergoing controlled ovarian stimulation are correlated to the number of oocytes retrieved. Fertil Steril. 2010;93:1844–50.
Palin MF, Labrecque B, Beaudry D, Mayhue M, Bordignon V, Murphy BD. Visfatin expression is not associated with adipose tissue abundance in the porcine model. Domest Anim Endocrinol. 2008;35:58–73.
Chen H, Xia T, Zhou L, Chen X, Gan L, Yao W, et al. Gene organization, alternate splicing and expression pattern of porcine visfatin gene. Domest Anim Endocrinol. 2007;32:235–45.
Thomas S, Kratzsch D, Schaab M, Scholz M, Grunewald S, Thiery J, et al. Seminal plasma adipokine levels are correlated with functional characteristics of spermatozoa. Fertil Steril. 2013;99:1256-1263.e3.
Ocón-Grove OM, Krzysik-Walker SM, Maddineni SR, Hendricks GL, Ramachandran R. NAMPT (visfatin) in the chicken testis: influence of sexual maturation on cellular localization, plasma levels and gene and protein expression. Reproduction. 2010;139:217–26.
Gurusubramanian G, Roy VK. Expression of visfatin in alloxan-induced diabetic rat testis. Acta Histochem. 2014;116:1462–8.
Celichowski P, Jopek K, Szyszka M, Milecka P, Tyczewska M, Sakhanova S, et al. Extracellular nampt (eNampt/visfatin/PBEF) directly and indirectly stimulates ACTH and CCL2 protein secretion from isolated rat corticotropes. Adv Clin Exp Med. 2021;30:967–80.
Hameed W, Yousaf I, Latif R, Aslam M. Effect of visfatin on testicular steroidogenesis in purified leydig cells. J Ayub Med Coll Abbottabad. 2012;24:3–4.
Annie L, Gurusubramanian G, Roy VK. Inhibition of visfatin/NAMPT affects ovarian proliferation, apoptosis, and steroidogenesis in pre-pubertal mice ovary. J Steroid Biochem Mol Biol. 2020;204:105763.
Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, et al. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–9.
Joseph C, Al-Izzi S, Alsaleem M, Kurozumi S, Toss MS, Arshad M, et al. Retinoid X receptor gamma (RXRG) is an independent prognostic biomarker in ER-positive invasive breast cancer. Br J Cancer. 2019;121:776–85.
Muttukrishna S, Tannetta D, Groome N, Sargent I. Activin and follistatin in female reproduction. Mol Cell Endocrinol. 2004;225:45–56.
Ling N, Ying SY, Ueno N, Shimasaki S, Esch F, Hotta M, et al. Pituitary FSH is released by a heterodimer of the β-subunits from the two forms of inhibin. Nature. 1986;321(6072):779–82.
Carroll RS, Corrigan AZ, Vale W, Chin WW. Activin stabilizes follicle-stimulating hormone-beta messenger ribonucleic acid levels. Endocrinology. 1991;129:1721–6.
Weiss J, Crowley WF Jr, Halvorson LM, Jameson JL. Perifusion of rat pituitary cells with gonadotropin-releasing hormone, activin, and inhibin reveals distinct effects on gonadotropin gene expression and secretion. Endocrinology. 1993;132:2307–11.
Yosten GLC, Lyu RM, Hsueh AJW, Avsian-Kretchmer O, Chang JK, Tullock CW, et al. A novel reproductive peptide, phoenixin. J Neuroendocrinol. 2013;25:206–15.
Stein LM, Tullock CW, Mathews SK, Garcia-Galiano D, Elias CF, Samson WK, et al. Hypothalamic action of phoenixin to control reproductive hormone secretion in females: importance of the orphan G protein-coupled receptor Gpr173. Am J Physiol Regul Integr Comp Physiol. 2016;311:R489–96.
Nasonkin IO, Ward RD, Raetzman LT, Seasholtz AF, Saunders TL, Gillespie PJ, et al. Pituitary hypoplasia and respiratory distress syndrome in Prop1 knockout mice. Hum Mol Genet. 2004;13:2727–35.
Deladoëy J, Flück C, Büyükgebiz A, Kuhlmann BV, Eblé A, Hindmarsh PC, et al. “Hot spot” in the PROP1 gene responsible for combined pituitary hormone deficiency. J Clin Endocrinol Metab. 1999;84:1645–50.
Araujo RV, Chang CV, Cescato VAS, Fragoso MCBV, Bronstein MD, Mendonca BB, et al. PROP1 overexpression in corticotrophinomas: evidence for the role of PROP1 in the maintenance of cells committed to corticotrophic differentiation. Clinics (Sao Paulo). 2013;68:887–91.
Bouchoucha YX, Charnay P, Gilardi-Hebenstreit P. Ablation of Egr2-positive cells in male mouse anterior pituitary leads to atypical isolated GH deficiency. Endocrinology. 2013;154:272–80.
Al Kahtane A, Chaiseha Y, El Halawani M. Dopaminergic regulation of avian prolactin gene transcription. J Mol Endocrinol. 2003;31:185–96.
Lincoln GA, Clarke IJ. Evidence that melatonin acts in the pituitary gland through a dopamine-independent mechanism to mediate effects of daylength on the secretion of prolactin in the ram. J Neuroendocrinol. 1995;7:637–43.
Wang C, Liu Y, Wang H, Wu H, Gong S, Chen W, et al. Molecular characterization and differential expression of multiple goose dopamine D2 receptors. Gene. 2014;535:177–83.
Recouvreux MV, Camilletti MA, Rifkin DB, Becu-Villalobos D, Díaz-Torga G. Thrombospondin-1 (TSP-1) analogs ABT-510 and ABT-898 inhibit prolactinoma growth and recover active pituitary transforming growth factor-β1 (TGF-β1). Endocrinology. 2012;153:3861–71.
Rifkin DB. Latent transforming growth factor-β (TGF-β) binding proteins: orchestrators of TGF-β availability. J Biol Chem. 2005;280:7409–12.
Cheng Q, Dong W, Qian L, Wu J, Peng Y. Visfatin inhibits apoptosis of pancreatic β-cell line, MIN6, via the mitogen-activated protein kinase/phosphoinositide 3-kinase pathway. J Mol Endocrinol. 2011;47:13–21.
Xiang RL, Mei M, Su YC, Li L, Wang JY, Wu LL. Visfatin protects rat pancreatic β-cells against IFN-γ-induced apoptosis through AMPK and ERK1/2 signaling pathways. Biomed Environ Sci. 2015;28:169–77.
Li Y, Zhang Y, Dorweiler B, Cui D, Wang T, Woo CW, et al. Extracellular nampt promotes macrophage survival via a nonenzymatic interleukin-6/STAT3 signaling mechanism. J Biol Chem. 2008;283:34833–43.
Nishino H, Saito T, Wei R, Takano T, Tsutsumi K, Taniguchi M, et al. The LMTK1—TBC1D9B—Rab11A cascade regulates dendritic spine formation via endosome trafficking. J Neurosci. 2019;39:9491–502.
Takano T, Urushibara T, Yoshioka N, Saito T, Fukuda M, Tomomura M, et al. LMTK1 regulates dendritic formation by regulating movement of Rab11A-positive endosomes. Mol Biol Cell. 2014;25:1755–68.
Raghunath M, Patti R, Bannerman P, Lee CM, Baker S, Sutton LN, et al. A novel kinase, AATYK induces and promotes neuronal differentiation in a human neuroblastoma (SH-SY5Y) cell line. Mol Brain Res. 2000;77:151–62.
Gaozza E, Baker SJ, Vora RK, Reddy EP. AATYK: a novel tyrosine kinase induced during growth arrest and apoptosis of myeloid cells. Oncogene. 1997;15(25):3127–35.
Ding LY, Hou YC, Kuo IY, Hsu TY, Tsai TC, Chang HW, et al. Epigenetic silencing of AATK in acinar to ductal metaplasia in murine model of pancreatic cancer. Clin Epigenetics. 2020;12:87.
Haag T, Herkt CE, Walesch SK, Richter AM, Dammann RH. The apoptosis associated tyrosine kinase gene is frequently hypermethylated in human cancer and is regulated by epigenetic mechanisms. Genes Cancer. 2014;5:365–74.
Ma S, Rubin BP. Apoptosis-associated tyrosine kinase 1 inhibits growth and migration and promotes apoptosis in melanoma. Lab Invest. 2014;94:430–8.
Xu TP, Liu XX, Xia R, Yin L, Kong R, Chen WM, et al. SP1-induced upregulation of the long noncoding RNA TINCR regulates cell proliferation and apoptosis by affecting KLF2 mRNA stability in gastric cancer. Oncogene. 2015;34(45):5648–61.
Nie FQ, Sun M, Yang JS, Xie M, Xu TP, Xia R, et al. Long noncoding RNA ANRIL promotes non-small cell lung cancer cell proliferation and inhibits apoptosis by silencing KLF2 and P21 expression. Mol Cancer Ther. 2015;14:268–77.
Lian Y, Yan C, Xu H, Yang J, Yu Y, Zhou J, et al. A novel lncRNA, LINC00460, affects cell proliferation and apoptosis by regulating KLF2 and CUL4A expression in colorectal cancer. Mol Ther Nucleic Acids. 2018;12:684–97.
Zhang Y, Johansson E, Miller ML, Jänicke RU, Ferguson DJ, Plas D, et al. Identification of a conserved anti-apoptotic protein that modulates the mitochondrial apoptosis pathway. PLoS One. 2011;6:e25284.
Correia S, Alves MG, Oliveira PF, Alves MR, van Pelt AMM, Cavaco JE, et al. Transgenic overexpression of regucalcin leads to suppression of thapsigargin- and actinomycin D-induced apoptosis in the testis by modulation of apoptotic pathways. Andrology. 2014;2:290–8.
Toda J, Ichii M, Oritani K, Shibayama H, Tanimura A, Saito H, et al. Signal-transducing adapter protein-1 is required for maintenance of leukemic stem cells in CML. Oncogene. 2020;39(34):5601–15.
Toda J, Ichii M, Shibayama H, Saito H, Kitai Y, Muromoto R, et al. Role of signal transducing adaptor protein-1 (STAP-1) in chronic myelogenous leukemia stem cells. Blood. 2018;132:4245.
Zhang HM, Li H, Wang GX, Wang J, Xiang Y, Huang Y, et al. MKL1/miR-5100/CAAP1 loop regulates autophagy and apoptosis in gastric cancer cells. Neoplasia. 2020;22:220–30.
Marques R, Maia CJ, Vaz C, Correia S, Socorro S. The diverse roles of calcium-binding protein regucalcin in cell biology: from tissue expression and signalling to disease. Cell Mol Life Sci. 2013;71:93–111.
Chatterjee S, Somu Naidu G, Grobe H, Ezra A, Ramishetti S, Sprinzak D, et al. Therapeutic gene silencing of CKAP5 leads to lethality in genetically unstable cancer cells. Sci Adv. 2023;9:eade4800.
Kunisky AK, Anyaeche VI, Herron RS, Park CY, Hwang HW. Shift in MSL1 alternative polyadenylation in response to DNA damage protects cancer cells from chemotherapeutic agent-induced apoptosis. Cell Rep. 2021;37:109815.
Tamura S, Toriumi S, Saito JI, Awano K, Kudo TA, Kobayashi T. PP2C family members play key roles in regulation of cell survival and apoptosis. Cancer Sci. 2006;97:563–7.
Zhou W, Cao H, Yang X, Cong K, Wang W, Chen T, et al. Of nuclear localization signal in the N terminus of integrin-linked kinase-associated phosphatase (ILKAP) and its essential role in the down-regulation of RSK2 protein signaling. J Biol Chem. 2013;288:6259–71.
Hoffman NE, Chandramoorthy HC, Shanmughapriya S, Zhang XQ, Vallem S, Doonan PJ, et al. SLC25A23 augments mitochondrial Ca2 + uptake, interacts with MCU, and induces oxidative stress-mediated cell death. Mol Biol Cell. 2014;25:936–47.
Fulda S. Targeting c-FLICE-like inhibitory protein (CFLAR) in cancer. Expert Opin Ther Targets. 2013;17:195–201.
Adya R, Tan BK, Punn A, Chen J, Randeva HS. Visfatin induces human endothelial VEGF and MMP-2/9 production via MAPK and PI3K/Akt signalling pathways: novel insights into visfatin-induced angiogenesis. Cardiovasc Res. 2008;78:356–65.
Kim JY, Bae YH, Bae MK, Kim SR, Park HJ, Wee HJ, et al. Visfatin through STAT3 activation enhances IL-6 expression that promotes endothelial angiogenesis. Biochim Biophys Acta. 2009;1793:1759–67.
Christian S, Winkler R, Helfrich I, Boos AM, Besemfelder E, Schadendorf D, et al. Endosialin (Tem1) is a marker of tumor-associated myofibroblasts and tumor vessel-associated mural cells. Am J Pathol. 2008;172:486–94.
Bagley RG, Honma N, Weber W, Boutin P, Rouleau C, Shankara S, et al. Endosialin/TEM 1/CD248 is a pericyte marker of embryonic and tumor neovascularization. Microvasc Res. 2008;76:180–8.
Hong CL, Yu IS, Pai CH, Chen JS, Hsieh MS, Wu HL, et al. CD248 regulates wnt signaling in pericytes to promote angiogenesis and tumor growth in lung cancer. Cancer Res. 2022;82:3734–50.
MacFadyen J, Savage K, Wienke D, Isacke CM. Endosialin is expressed on stromal fibroblasts and CNS pericytes in mouse embryos and is downregulated during development. Gene Expr Patterns. 2007;7:363–9.
Bhattacharya R, Senbanerjee S, Lin Z, Mir S, Hamik A, Wang P, et al. Inhibition of vascular permeability factor/vascular endothelial growth factor-mediated angiogenesis by the Kruppel-like factor KLF2. J Biol Chem. 2005;280:28848–51.
Tello-Montoliu A, Patel JV, Lip GYH. Angiogenin: a review of the pathophysiology and potential clinical applications. J Thromb Haemost. 2006;4:1864–74.
Li S, Hu MG, Sun Y, Yoshioka N, Ibaragi S, Sheng J, et al. Angiogenin mediates androgen-stimulated prostate cancer growth and enables castration resistance. Mol Cancer Res. 2013;11:1203–14.
Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Curry WT, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.
Tsuji T, Sun Y, Kishimoto K, Olson KA, Liu S, Hirukawa S, et al. Angiogenin is translocated to the nucleus of HeLa cells and is involved in ribosomal RNA transcription and cell proliferation. Cancer Res. 2005;65:1352–60.
Shin DH, Yeon Jo J, Kim SH, Choi M, Han C, Choi BK, et al. Midkine is a potential therapeutic target of tumorigenesis, angiogenesis, and metastasis in non-small cell lung cancer. Cancers (Basel). 2020;12:2402.
Zhang N, Deuel TF. Pleiotrophin and midkine, a family of mitogenic and angiogenic heparin-binding growth and differentiation factors. Curr Opin Hematol. 1999;6:44–50.
Hao H, Maeda Y, Fukazawa T, Yamatsuji T, Takaoka M, Bao XH, et al. Inhibition of the growth factor MDK/Midkine by a novel small molecule compound to treat non-small cell lung cancer. PLoS One. 2013;8:e71093.
Vieira SC, Silva BB, Pinto GA, Vassallo J, Moraes NG, Santana JOI, et al. CD34 as a marker for evaluating angiogenesis in cervical cancer. Pathol Res Pract. 2005;201:313–8.
Brooks PC, Clark RAF, Cheresh DA. Requirement of vascular integrin αvβ3 for angiogenesis. Science. 1994;264:569–71.
Dechantsreiter MA, Planker E, Mathä B, Lohof E, Hölzemann G, Jonczyk A, et al. N-Methylated cyclic RGD peptides as highly active and selective αvβ3 integrin antagonists. J Med Chem. 1999;42:3033–40.
Hammes HP, Brownlee M, Jonzcyk A, Sutter A, Preissner KT. Subcutaneous injection of a cyclic peptide antagonist of vitronectin receptor-type integrins inhibits retinal neovascularization. Nat Med. 1996;2:529–33.
Brooks PC, Montgomery AMP, Rosenfeld M, Reisfeld RA, Hu T, Klier G, et al. Integrin αvβ3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell. 1994;79:1157–64.
Brooks PC, Strömblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA. Antiintegrin αvβ3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest. 1995;96:1815–22.
Li Y, Cai B, Shen L, Dong Y, Lu Q, Sun S, et al. MiRNA-29b suppresses tumor growth through simultaneously inhibiting angiogenesis and tumorigenesis by targeting Akt3. Cancer Lett. 2017;397:111–9.
Calderone V, Gallego J, Fernandez-Miranda G, Garcia-Pras E, Maillo C, Berzigotti A, et al. Sequential functions of CPEB1 and CPEB4 regulate pathologic expression of vascular endothelial growth factor and angiogenesis in chronic liver disease. Gastroenterology. 2016;150:982–97.
Ortiz-Zapater E, Pineda D, Martínez-Bosch N, Fernández-Miranda G, Iglesias M, Alameda F, et al. Key contribution of CPEB4-mediated translational control to cancer progression. Nat Med. 2011;18(1):83–90.
Liby TA, Spyropoulos P, Buff Lindner H, Eldridge J, Beeson C, Hsu T, et al. Akt3 controls vascular endothelial growth factor secretion and angiogenesis in ovarian cancer cells. Int J Cancer. 2012;130:532–43.
Martyniak M, Zglejc K, Franczak A, Kotwica G. Expression of 3β-hydroxysteroid dehydrogenase and P450 aromatase in porcine oviduct during the oestrous cycle. J Anim Feed Sci. 2016;25:235–43.
Martyniak M, Franczak A, Kotwica G. Interleukin-1β system in the oviducts of pigs during the oestrous cycle and early pregnancy. Theriogenology. 2017;96:31–41.
Funding
This research was supported by the Polish National Science Center research grants: OPUS 16 [grant number: 2018/31/B/NZ9/00781], OPUS 20 [grant number: 2020/39/B/NZ9/01061] and SONATA 16 [grant number: 2020/39/D/NZ9/01009].
Author information
Authors and Affiliations
Contributions
KD: Conceptualization, methodology, validation, formal analysis, investigation, data curation, visualization, writing— original draft preparation, writing—review and editing. MK and EZ: Investigation. MG, BZ: Validation. GK: Validation and data curation. LP: Formal analysis and data curation. AR: Writing—review and editing. NS: Investigation, resources, writing—review and editing, funding acquisition. TK: Conceptualization, methodology, writing—review and editing, supervision, project administration, funding acquisition.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was conducted according to the Polish Act of the protection of animals used for educational or scientific purposes of 15 January 2015 and directive 2010/63/EU (Journal of Laws Dz.U. 2015 No. item 266) of the European Parliament of 22 September 2010 (2010/63/EU) on the protection of animals used for scientific objectives, so this study did not require the consent of the competent ethics committee for animal experiments.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no conflict of interest.
Supplementary Information
Supplementary
File 1. General statistics of high-throughput RNA sequencing and mapping to the porcine genome.
Supplementary
File 2. List of differentially expressed genes.
Supplementary
File 3. Gene Ontology (GO) analysis generated from up-regulated and down-regulated DE-genes.
Supplementary
File 4. Gene Ontology (GO) analysis generated from up-regulated and down-regulated DELs.
Supplementary File 5
. Results obtained during the analysis of differentially expressed AS events
Supplementary
File 6. Complete list of gene interactions generated by GeneMania.
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.
About this article
Cite this article
Dobrzyn, K., Kopij, G., Kiezun, M. et al. Visfatin (NAMPT) affects global gene expression in porcine anterior pituitary cells during the mid-luteal phase of the oestrous cycle. J Animal Sci Biotechnol 15, 96 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-024-01054-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-024-01054-z